1
|
Houlder EL, Gago S, Vere G, Furlong-Silva J, Conn D, Hickey E, Khan S, Thomson D, Shepherd MW, Lebedinec R, Brown GD, Horsnell W, Bromley M, MacDonald AS, Cook PC. Aspergillus-mediated allergic airway inflammation is triggered by dendritic cell recognition of a defined spore morphotype. J Allergy Clin Immunol 2024:S0091-6749(24)01240-5. [PMID: 39581297 DOI: 10.1016/j.jaci.2024.10.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 10/26/2024] [Accepted: 10/31/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Exposure to fungi, especially Aspergillus fumigatus, can elicit potent allergic inflammation that triggers and worsens asthmatic disease. Dendritic cells (DCs) initiate allergic inflammatory responses to allergic stimuli. However, it is unclear if Af spores during isotropic growth (early spore swelling) can activate DCs to initiate allergic responses or if germination is required. This lack of basic understanding of how Af causes disease is a barrier to developing new treatments. OBJECTIVE We sought to show that a precise Af morphotype stage during spore swelling can trigger DCs to mediate allergic inflammatory responses and ascertain if antifungal therapeutics can be effective at suppressing this process. METHODS We used an Af strain deficient in pyrimidine biosynthesis (ΔpyrG) to generate populations of Af spores arrested at different stages of isotropic growth (swelling) via temporal removal of uracil and uridine from growth media. These arrested spore stages were cultured with bone marrow-derived DCs (BMDCs), and their activation was measured via flow cytometry and ELISA to examine which growth stage was able to activate BMDCs. These BMDCs were then adoptively transferred into the airways to assess if they were able to mediate allergic inflammation in naïve recipient mice. Allergic airway inflammation in vivo was determined via flow cytometry, ELISA, and real-time quantitative PCR. This system was also used to determine if antifungal drug (itraconazole) treatment could alter early stages of spore swelling and therefore BMDC activation and in vivo allergic inflammation upon adoptive transfer. RESULTS We found that Af isotropic growth is essential to trigger BMDC activation and mediate allergic airway inflammation. Furthermore, using time-arrested Af stages, we found that at least 3 hours in growth media enabled spores to swell sufficiently to activate BMDCs to elicit allergic airway inflammation in vivo. Incubation of germinating Af with itraconazole reduced spore swelling and partially reduced their ability to activate BMDCs to elicit in vivo allergic airway inflammation. CONCLUSION Our results have pinpointed the precise stage of Af development when germinating spores are able to activate DCs to mediate downstream allergic airway inflammation. Furthermore, we have identified that antifungal therapeutics partially reduced the potential of Af spores to stimulate allergic responses, highlighting a potential mechanism by which antifungal treatment might help prevent the development of fungal allergy.
Collapse
Affiliation(s)
- Emma L Houlder
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom; Leiden University Center for Infectious Disease, Leiden University Medical Centre, Leiden, The Netherlands
| | - Sara Gago
- Manchester Fungal Infection Group, Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| | - George Vere
- Department of Biosciences, Medical Research Council Centre for Medical Mycology at the University of Exeter, Faculty of Health and Life Sciences, Exeter, United Kingdom
| | - Julio Furlong-Silva
- Department of Biosciences, Medical Research Council Centre for Medical Mycology at the University of Exeter, Faculty of Health and Life Sciences, Exeter, United Kingdom
| | - Daniel Conn
- Department of Biosciences, Medical Research Council Centre for Medical Mycology at the University of Exeter, Faculty of Health and Life Sciences, Exeter, United Kingdom
| | - Emer Hickey
- Department of Biosciences, Medical Research Council Centre for Medical Mycology at the University of Exeter, Faculty of Health and Life Sciences, Exeter, United Kingdom
| | - Saba Khan
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Darren Thomson
- Manchester Fungal Infection Group, Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom; Department of Biosciences, Medical Research Council Centre for Medical Mycology at the University of Exeter, Faculty of Health and Life Sciences, Exeter, United Kingdom
| | - Mark W Shepherd
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Ressa Lebedinec
- Manchester Fungal Infection Group, Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| | - Gordon D Brown
- Department of Biosciences, Medical Research Council Centre for Medical Mycology at the University of Exeter, Faculty of Health and Life Sciences, Exeter, United Kingdom
| | - William Horsnell
- Department of Biosciences, Medical Research Council Centre for Medical Mycology at the University of Exeter, Faculty of Health and Life Sciences, Exeter, United Kingdom
| | - Mike Bromley
- Manchester Fungal Infection Group, Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| | - Andrew S MacDonald
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Peter C Cook
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom; Department of Biosciences, Medical Research Council Centre for Medical Mycology at the University of Exeter, Faculty of Health and Life Sciences, Exeter, United Kingdom.
| |
Collapse
|
2
|
Li Y, Fu W, Xiang J, Ren Y, Li Y, Zhou M, Yu J, Luo Z, Liu E, Fu Z, Liu B, Ding F. Long-chain acyl-CoA synthetase 4-mediated mitochondrial fatty acid metabolism and dendritic cell antigen presentation. Inflamm Res 2024; 73:819-839. [PMID: 38472395 DOI: 10.1007/s00011-024-01868-7] [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: 12/21/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
OBJECTIVE This study aims to investigate the role of Acyl-CoA synthetase 4 (ACSL4) in mediating mitochondrial fatty acid metabolism and dendritic cell (DC) antigen presentation in the immune response associated with asthma. METHODS RNA sequencing was employed to identify key genes associated with mitochondrial function and fatty acid metabolism in DCs. ELISA was employed to assess the levels of fatty acid metabolism in DCs. Mitochondrial morphology was evaluated using laser confocal microscopy, structured illumination microscopy, and transmission electron microscopy. Flow cytometry and immunofluorescence were utilized to detect changes in mitochondrial superoxide generation in DCs, followed by immunofluorescence co-localization analysis of ACSL4 and the mitochondrial marker protein COXIV. Subsequently, pathological changes and immune responses in mouse lung tissue were observed. ELISA was conducted to measure the levels of fatty acid metabolism in lung tissue DCs. qRT-PCR and western blotting were employed to respectively assess the expression levels of mitochondrial-associated genes (ATP5F1A, VDAC1, COXIV, TFAM, iNOS) and proteins (ATP5F1A, VDAC1, COXIV, TOMM20, iNOS) in lung tissue DCs. Flow cytometry was utilized to analyze changes in the expression of surface antigens presented by DCs in lung tissue, specifically the MHCII molecule and the co-stimulatory molecules CD80/86. RESULTS The sequencing results reveal that ACSL4 is a crucial gene regulating mitochondrial function and fatty acid metabolism in DCs. Inhibiting ACSL4 reduces the levels of fatty acid oxidases in DCs, increases arachidonic acid levels, and decreases A-CoA synthesis. Simultaneously, ACSL4 inhibition leads to an increase in mitochondrial superoxide production (MitoSOX) in DCs, causing mitochondrial rupture, vacuolization, and sparse mitochondrial cristae. In mice, ACSL4 inhibition exacerbates pulmonary pathological changes and immune responses, reducing the fatty acid metabolism levels within lung tissue DCs and the expression of mitochondria-associated genes and proteins. This inhibition induces an increase in the expression of MHCII antigen presentation molecules and co-stimulatory molecules CD80/86 in DCs. CONCLUSIONS The research findings indicate that ACSL4-mediated mitochondrial fatty acid metabolism and dendritic cell antigen presentation play a crucial regulatory role in the immune response of asthma. This discovery holds promise for enhancing our understanding of the mechanisms underlying asthma pathogenesis and potentially identifying novel targets for its prevention and treatment.
Collapse
Affiliation(s)
- Yan Li
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2 Road, Yuzhong Dis, Chongqing, 400014, People's Republic of China
- Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing Medical University, Chongqing, People's Republic of China
| | - Wenlong Fu
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2 Road, Yuzhong Dis, Chongqing, 400014, People's Republic of China
- Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing Medical University, Chongqing, People's Republic of China
| | - JinYing Xiang
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2 Road, Yuzhong Dis, Chongqing, 400014, People's Republic of China
- Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yinying Ren
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2 Road, Yuzhong Dis, Chongqing, 400014, People's Republic of China
- Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yuehan Li
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2 Road, Yuzhong Dis, Chongqing, 400014, People's Republic of China
- Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing Medical University, Chongqing, People's Republic of China
| | - Mi Zhou
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2 Road, Yuzhong Dis, Chongqing, 400014, People's Republic of China
- Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing Medical University, Chongqing, People's Republic of China
| | - Jinyue Yu
- Bristol Medical School, University of Bristol, Bristol, UK
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Zhengxiu Luo
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2 Road, Yuzhong Dis, Chongqing, 400014, People's Republic of China
- Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing Medical University, Chongqing, People's Republic of China
| | - Enmei Liu
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2 Road, Yuzhong Dis, Chongqing, 400014, People's Republic of China
- Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhou Fu
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2 Road, Yuzhong Dis, Chongqing, 400014, People's Republic of China
- Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing Medical University, Chongqing, People's Republic of China
| | - Bo Liu
- Department of Cardiothoracic Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2 Road, Yuzhong Dis, Chongqing, 400014, People's Republic of China.
- Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing Medical University, Chongqing, People's Republic of China.
| | - Fengxia Ding
- Department of Respiratory Medicine, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2 Road, Yuzhong Dis, Chongqing, 400014, People's Republic of China.
- Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
3
|
Lin CY, Cheng CW, Ko JL, Lue KH, Liu YF. Multiplexed transcriptional profiling of Dermatophagoides house dust mites allergens in human epithelium cells. ENVIRONMENTAL TOXICOLOGY 2024; 39:2229-2239. [PMID: 38124673 DOI: 10.1002/tox.24109] [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: 11/01/2023] [Revised: 12/03/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023]
Abstract
Allergic asthma, a chronic disease characterized by airway inflammation, poses a significant public health concern. It is well-established that house dust mites (HDMs) are common inducers of allergic responses in individuals, particularly children. In central Taiwan, our research team observed that over 80% of allergic children exhibited sensitization to various HDMs species. This investigation aims to bridge the gap between these observations and a better understanding of the early fundamental mechanisms for preventing allergic diseases. Specifically, our study delves into the impact of crude extracts of HDMs on human epithelial BEAS-2B cells. Our findings, based on RNA sequencing (RNA-seq) analysis, shed light on how three major Dermatophagoides HDMs allergens activate a common Toll-like receptor signaling pathway in human epithelial cells within a 4-h treatment. During this process, the nuclear transcription factor NF-κB translocated into the cell nucleus within 30 min of allergen stimulation, triggering the expression of pro-inflammatory genes such as IL-6 and IL-8 over 4 h. Additionally, when the cells were treated with specific Dermatophagoides microceras (Der m) allergens, it resulted in the upregulation of genes that regulate type 1 diabetes mellitus (T1DM) signaling pathways. This led to the mediation of IL-12A inflammation. Furthermore, there was an increase in gene sets associated with cilia function and the microtubule cytoskeleton in human epithelial cells after treatment with a combination of Der m allergens and Dexamethasone. Additionally, OMICs analysis was conducted to examine the effects of HDMs allergenic stimulation on human epidermal cells. We aimed to improve our understanding of the molecular mechanisms within cells and identify potential targets and natural products in the treatment of asthma caused by HDMs allergens.
Collapse
Affiliation(s)
- Chia-Yang Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Family Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chun-Wen Cheng
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Jiunn-Liang Ko
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ko-Huang Lue
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of Allergy, Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yu-Fan Liu
- Division of Allergy, Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
4
|
Camp B, Jorde I, Sittel F, Pausder A, Jeron A, Bruder D, Schreiber J, Stegemann-Koniszewski S. Comprehensive analysis of lung macrophages and dendritic cells in two murine models of allergic airway inflammation reveals model- and subset-specific accumulation and phenotypic alterations. Front Immunol 2024; 15:1374670. [PMID: 38529288 PMCID: PMC10961404 DOI: 10.3389/fimmu.2024.1374670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/27/2024] [Indexed: 03/27/2024] Open
Abstract
Introduction Allergic asthma has been mainly attributed to T helper type 2 (Th2) and proinflammatory responses but many cellular processes remain elusive. There is increasing evidence for distinct roles for macrophage and dendritic cell (DC) subsets in allergic airway inflammation (AAI). At the same time, there are various mouse models for allergic asthma that have been of utmost importance in identifying key inflammatory pathways in AAI but that differ in the allergen and/or route of sensitization. It is unclear whether and how the accumulation and activation of specialized macrophage and DC subsets depend on the experimental model chosen for analyses. Methods In our study, we employed high-parameter spectral flow cytometry to comprehensively assess the accumulation and phenotypic alterations of different macrophage- and DC-subsets in the lung in an OVA- and an HDM-mediated mouse model of AAI. Results We observed subset-specific as well as model-specific characteristics with respect to cell numbers and functional marker expression. Generally, alveolar as opposed to interstitial macrophages showed increased MHCII surface expression in AAI. Between the models, we observed significantly increased numbers of alveolar macrophages, CD103+ DC and CD11b+ DC in HDM-mediated AAI, concurrent with significantly increased airway interleukin-4 but decreased total serum IgE levels. Further, increased expression of CD80 and CD86 on DC was exclusively detected in HDM-mediated AAI. Discussion Our study demonstrates a model-specific involvement of macrophage and DC subsets in AAI. It further highlights spectral flow cytometry as a valuable tool for their comprehensive analysis under inflammatory conditions in the lung.
Collapse
Affiliation(s)
- Belinda Camp
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Ilka Jorde
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Franka Sittel
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Alexander Pausder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Andreas Jeron
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
- Department of Pediatrics, Ludwig-Maximilians University of Munich, Munich, Germany
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jens Schreiber
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Sabine Stegemann-Koniszewski
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
5
|
Shan Y, Wu J, Dai X, Yuan C, Jiang J, Yan H, Tang L, Dong Y, Ren L, Pan Q, Ji J, Zhao X. Jiangqi Pingxiao formula regulates dendritic cell apoptosis in an autophagy-dependent manner through the AMPK/mTOR pathway in a murine model of OVA-induced asthma. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117405. [PMID: 37952734 DOI: 10.1016/j.jep.2023.117405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/16/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Allergic asthma is a recurring respiratory condition that typically manifests during childhood or adolescence. It is characterized by a dominant type II immune response triggered by the identification and capturing of inhaled allergens by dendritic cells (DCs). Jiangqi Pingxiao Formula (JQPXF), a prescription medicine used for the treatment of pediatric asthma, has been clinically proven to be both safe and effective. However, its mechanism of action in the treatment of asthma has not been fully been fully elucidated. Recent research suggests that several natural compounds have the potential to target dendritic cells (DCs) and alleviate ovalbumin (OVA)-induced asthma, which may also be found within JQPXF. AIM OF THE STUDY This study aimed to elucidate the effect of JQPXF on OVA-induced asthma model and its molecular mechanism targeting DCs. MATERIALS AND METHODS The main constituents of JQPXF were analyzed by ultra performance liquid chromatography (UPLC). An asthma model was established by OVA. Hematoxylin-eosin staining and measurement of respiratory function was used to evaluate the treatment effect of JQPXF on asthmatic mice. Cytokine (IL-5, IL-13 and IgE) concentrations were determined by enzyme-linked immunosorbent assay (ELISA). Flow cytometry was employed to evaluate inflammatory cell infiltration (T helper 2 cells and DCs) in vivo and DC survival in vivo and vitro. Western blot and immunofluorescence were used to verify the molecular mechanisms. RESULTS The results suggest that JQPXF can ameliorate pathological conditions and improve lung function in asthmatic mice, as well as the Th2 cells. Treatment with JQPXF significantly reduced the number of DCs and increased the number of Propidium iodide+ (PI) DCs. Furthermore, JQPXF upregulated protein levels of the pro-apoptotic factors Cleaved-caspase-3 and Bax, while downregulating the anti-apoptotic factor Bcl-2. Simultaneously, JQPXF increased autophagy levels by facilitating p62 degradation and promoting translation from LC3B I to LC3B II of DCs in vitro, as well as reducing the integrated optical density (IOD) of p62 within the CD11c-positive area in the lung. 3-Methyladenine (3-MA) was used to block autophagic flux and the apoptotic effect of JQPXF on DCs was abolished in vitro, with the number of DCs decreased by JQPXF being reversed in vivo. We further investigated the upstream key regulator of autophagy, the AMPK/mTOR pathway, and found that JQPXF increased AMPK phosphorylation while decreasing mTOR phosphorylation levels. Additionally, we employed Compound C (CC) as an AMPK inhibitor to inhibit this signaling pathway, and our findings revealed that both autophagic flux and apoptotic levels in DCs were abolished in vitro. CONCLUSIONS In summary, we have demonstrated that JQPXF could alleviate type II inflammation in an asthmatic model by promoting the apoptosis of DCs through an autophagy-dependent mechanism, achieved by regulating the AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yiwen Shan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiabao Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaohan Dai
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chen Yuan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjin Jiang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hua Yan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lingling Tang
- School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yingmei Dong
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lishun Ren
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qingyun Pan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianjian Ji
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Xia Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
6
|
Yang G, Zeng XH, Geng XR, Liu JQ, Mo LH, Luo XQ, Liu HZ, Zhang YY, Yang LT, Huang QM, Xiao XJ, Liu J, Xu LZ, Liu DB, Liu XY, Liu ZQ, Yang PC. The transcription factor XBP1 in dendritic cells promotes the T H2 cell response in airway allergy. Sci Signal 2023; 16:eabm9454. [PMID: 37368951 DOI: 10.1126/scisignal.abm9454] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 06/06/2023] [Indexed: 06/29/2023]
Abstract
Dendritic cells (DCs) that express T cell immunoglobulin domain molecule-4 (TIM4), a cell surface receptor for phosphatidylserine, induce T helper 2 (TH2) cell responses and allergic reactions. We elucidated the role of the transcription factor X-box-binding protein-1 (XBP1) in the induction of the TH2 cell response through its role in generating TIM4+ DCs. We found that XBP1 was required for TIM4 mRNA and protein expression in airway DCs in response to the cytokine interleukin-2 (IL-2) and that this pathway was required for TIM4 expression on DCs in response to the allergens PM2.5 and Derf1. The IL-2-XBP1-TIM4 axis in DCs contributed to Derf1/PM2.5-induced, aberrant TH2 cell responses in vivo. An interaction between the guanine nucleotide exchange factor Son of sevenless-1 (SOS1) and the GTPase RAS promoted XBP1 and TIM4 production in DCs. Targeting the XBP1-TIM4 pathway in DCs prevented or alleviated experimental airway allergy. Together, these data suggest that XBP1 is required for TH2 cell responses by inducing the development of TIM4+ DCs, which depends on the IL-2-XBP1-SOS1 axis. This signaling pathway provides potential therapeutic targets for the treatment of TH2 cell-dependent inflammation or allergic diseases.
Collapse
Affiliation(s)
- Gui Yang
- Department of Otolaryngology, Head and Neck Surgery, Longgang Central Hospital, Shenzhen, China
| | - Xian-Hai Zeng
- Longgang ENT Hospital, Shenzhen, China
- Shenzhen ENT Institute, Shenzhen, China
| | - Xiao-Rui Geng
- Longgang ENT Hospital, Shenzhen, China
- Shenzhen ENT Institute, Shenzhen, China
| | - Jiang-Qi Liu
- Longgang ENT Hospital, Shenzhen, China
- Shenzhen ENT Institute, Shenzhen, China
| | - Li-Hua Mo
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China
- Institute of Allergy and Immunology, Shenzhen University School of Medicine and State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Shenzhen, China
- Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Xiang-Qian Luo
- Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Hua-Zhen Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China
- Institute of Allergy and Immunology, Shenzhen University School of Medicine and State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Shenzhen, China
| | - Yuan-Yi Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China
- Institute of Allergy and Immunology, Shenzhen University School of Medicine and State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Shenzhen, China
| | - Li-Teng Yang
- Department of General Practice Medicine and Respirology, Third Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, China
| | - Qin-Miao Huang
- Department of General Practice Medicine and Respirology, Third Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, China
| | - Xiao-Jun Xiao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China
- Institute of Allergy and Immunology, Shenzhen University School of Medicine and State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Shenzhen, China
| | - Jie Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China
- Institute of Allergy and Immunology, Shenzhen University School of Medicine and State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Shenzhen, China
| | - Ling-Zhi Xu
- Department of Immunology, Weifang Medical University, Weifang, China
| | - Da-Bo Liu
- Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Xiao-Yu Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China
- Institute of Allergy and Immunology, Shenzhen University School of Medicine and State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Shenzhen, China
| | - Zhi-Qiang Liu
- Longgang ENT Hospital, Shenzhen, China
- Shenzhen ENT Institute, Shenzhen, China
| | - Ping-Chang Yang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China
- Institute of Allergy and Immunology, Shenzhen University School of Medicine and State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Shenzhen, China
| |
Collapse
|
7
|
Striz I, Golebski K, Strizova Z, Loukides S, Bakakos P, Hanania N, Jesenak M, Diamant Z. New insights into the pathophysiology and therapeutic targets of asthma and comorbid chronic rhinosinusitis with or without nasal polyposis. Clin Sci (Lond) 2023; 137:727-753. [PMID: 37199256 PMCID: PMC10195992 DOI: 10.1042/cs20190281] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/22/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023]
Abstract
Asthma and chronic rhinosinusitis with nasal polyps (CRSwNP) or without (CRSsNP) are chronic respiratory diseases. These two disorders often co-exist based on common anatomical, immunological, histopathological, and pathophysiological basis. Usually, asthma with comorbid CRSwNP is driven by type 2 (T2) inflammation which predisposes to more severe, often intractable, disease. In the past two decades, innovative technologies and detection techniques in combination with newly introduced targeted therapies helped shape our understanding of the immunological pathways underlying inflammatory airway diseases and to further identify several distinct clinical and inflammatory subsets to enhance the development of more effective personalized treatments. Presently, a number of targeted biologics has shown clinical efficacy in patients with refractory T2 airway inflammation, including anti-IgE (omalizumab), anti-IL-5 (mepolizumab, reslizumab)/anti-IL5R (benralizumab), anti-IL-4R-α (anti-IL-4/IL-13, dupilumab), and anti-TSLP (tezepelumab). In non-type-2 endotypes, no targeted biologics have consistently shown clinical efficacy so far. Presently, multiple therapeutical targets are being explored including cytokines, membrane molecules and intracellular signalling pathways to further expand current treatment options for severe asthma with and without comorbid CRSwNP. In this review, we discuss existing biologics, those under development and share some views on new horizons.
Collapse
Affiliation(s)
- Ilja Striz
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Subdivision of Allergology and Clinical Immunology, Institute for Postgraduate Education in Medicine, Prague, Czech Republic
| | - Kornel Golebski
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands
| | - Zuzana Strizova
- Institute of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Stelios Loukides
- Department of Respiratory Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Petros Bakakos
- First Respiratory Medicine Department, National and Kapodistrian University of Athens, Athens, Greece
| | - Nicola A. Hanania
- Section of Pulmonary and Critical Care Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Milos Jesenak
- Department of Pulmonology and Phthisiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital in Martin, Slovakia
- Department of Pediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital in Martin, Slovakia
- Department of Clinical Immunology and Allergology, University Hospital in Martin, Slovakia
| | - Zuzana Diamant
- Department of Microbiology Immunology and Transplantation, KU Leuven, Catholic University of Leuven, Belgium
- Department of Respiratory Medicine and Allergology, Institute for Clinical Science, Skane University Hospital, Lund University, Lund, Sweden
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
8
|
Targeting the Semaphorin3E-plexinD1 complex in allergic asthma. Pharmacol Ther 2023; 242:108351. [PMID: 36706796 DOI: 10.1016/j.pharmthera.2023.108351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 01/09/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023]
Abstract
Asthma is a heterogenous airway disease characterized by airway inflammation and remodeling. It affects more than 300 million people worldwide and poses a significant burden on society. Semaphorins, discovered initially as neural guidance molecules, are ubiquitously expressed in various organs and regulate multiple signaling pathways. Interestingly, Semaphorin3E is a critical molecule in lung pathophysiology through its role in both lung development and homeostasis. Semaphorin3E binds to plexinD1, mediating regulatory effects on cell migration, proliferation, and angiogenesis. Recent in vitro and in vivo studies have demonstrated that the Semaphorin3E-plexinD1 axis is implicated in asthma, impacting inflammatory and structural cells associated with airway inflammation, tissue remodeling, and airway hyperresponsiveness. This review details the Semaphorin3E-plexinD1 axis in various aspects of asthma and highlights future directions in research including its potential role as a therapeutic target in airway allergic diseases.
Collapse
|
9
|
Han M, Ma J, Ouyang S, Wang Y, Zheng T, Lu P, Zheng Z, Zhao W, Li H, Wu Y, Zhang B, Hu R, Otsu K, Liu X, Wan Y, Li H, Huang G. The kinase p38α functions in dendritic cells to regulate Th2-cell differentiation and allergic inflammation. Cell Mol Immunol 2022; 19:805-819. [PMID: 35551270 PMCID: PMC9243149 DOI: 10.1038/s41423-022-00873-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 04/02/2022] [Accepted: 04/15/2022] [Indexed: 12/24/2022] Open
Abstract
Dendritic cells (DCs) play a critical role in controlling T helper 2 (Th2) cell-dependent diseases, but the signaling mechanism that triggers this function is not fully understood. We showed that p38α activity in DCs was decreased upon HDM stimulation and dynamically regulated by both extrinsic signals and Th2-instructive cytokines. p38α-specific deletion in cDC1s but not in cDC2s or macrophages promoted Th2 responses under HDM stimulation. Further study showed that p38α in cDC1s regulated Th2-cell differentiation by modulating the MK2−c-FOS−IL-12 axis. Importantly, crosstalk between p38α-dependent DCs and Th2 cells occurred during the sensitization phase, not the effector phase, and was conserved between mice and humans. Our results identify p38α signaling as a central pathway in DCs that integrates allergic and parasitic instructive signals with Th2-instructive cytokines from the microenvironment to regulate Th2-cell differentiation and function, and this finding may offer a novel strategy for the treatment of allergic diseases and parasitic infection.
Collapse
Affiliation(s)
- Miaomiao Han
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 200031, Shanghai, China
| | - Jingyu Ma
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Suidong Ouyang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, 523808, Dongguan, China
| | - Yanyan Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, 523808, Dongguan, China
| | - Tingting Zheng
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, 523808, Dongguan, China
| | - Peishan Lu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, 523808, Dongguan, China
| | - Zihan Zheng
- Biomedical Analysis Center, Army Medical University, 400038, Chongqing, China
| | - Weiheng Zhao
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.,Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Hongjin Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.,Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 200437, Shanghai, China
| | - Yun Wu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, 523808, Dongguan, China
| | - Baohua Zhang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.,Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, 200072, Shanghai, China
| | - Ran Hu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.,Basic Department of Cancer Center, Shanghai Tenth People's Hospital of Tongji University, 200072, Shanghai, China
| | - Kinya Otsu
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan.,School of Cardiovascular Medicine and Sciences, King's College London, London, SE59NU, UK
| | - Xinguang Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, 523808, Dongguan, China
| | - Ying Wan
- Biomedical Analysis Center, Army Medical University, 400038, Chongqing, China.
| | - Huabin Li
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 200031, Shanghai, China.
| | - Gonghua Huang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China. .,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, 523808, Dongguan, China.
| |
Collapse
|
10
|
Semaphorin3E/plexinD1 Axis in Asthma: What We Know So Far! ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:205-213. [PMID: 34019271 DOI: 10.1007/978-3-030-68748-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Semaphorin3E belongs to the large family of semaphorin proteins. Semaphorin3E was initially identified as axon guidance cues in the neural system. It is universally expressed beyond the nervous system and contributes to regulating essential cell functions such as cell migration, proliferation, and adhesion. Binding of semaphorin3E to its receptor, plexinD1, triggers diverse signaling pathways involved in the pathogenesis of various diseases from cancer to autoimmune and allergic disorders. Here, we highlight the novel findings on the role of semaphorin3E in airway biology. In particular, we highlight our recent findings on the function and potential mechanisms by which semaphorin3E and its receptor, plexinD1, impact airway inflammation, airway hyperresponsiveness, and remodeling in the context of asthma.
Collapse
|
11
|
Percier P, De Prins S, Tima G, Beyaert R, Grooten J, Romano M, Denis O. Aspergillus fumigatus Recognition by Dendritic Cells Negatively Regulates Allergic Lung Inflammation through a TLR2/MyD88 Pathway. Am J Respir Cell Mol Biol 2021; 64:39-49. [PMID: 32970964 DOI: 10.1165/rcmb.2020-0083oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Aspergillus fumigatus is an opportunistic fungal pathogen responsible for a spectrum of clinical manifestations. Dendritic cells recognize pathogen-associated molecular patterns of Aspergillus via two main receptor families, Toll-like receptors (TLRs) and C-type lectin receptors (CLR). Here, the importance of TLR and CLR signaling in the regulation of T-helper cell type 2 (Th2) responses was analyzed using a mouse model based on the transfer of bone marrow-derived dendritic cells (BMDCs) pulsed with A. fumigatus conidia. BMDCs were generated from mice deficient in either MyD88 or MALT1 (mucosa-associated lymphoid tissue lymphoma translocation protein 1). Both the MyD88 and MALT1 signaling pathway in BMDCs contributed to the production of inflammatory cytokines induced by A. fumigatus conidia. Mice sensitized with MyD88-/- BMDCs pulsed in vitro with A. fumigatus conidia showed an exacerbated allergic inflammation, with stronger eosinophil recruitment in the BAL and higher Th2 cytokine production compared with mice sensitized with wild-type or MALT1-/- BMDCs. This exacerbation was not observed when MyD88-/- BMDCs were pulsed with Cladosporium sphaerospermum, a nonpathogenic mold. A lack of TLR2 signaling recapitulated the exacerbation of the A. fumigatus Th2 response observed in the absence of MyD88 signaling, whereas TLR2 agonist dampened the response induced with A. fumigatus and C. sphaerospermum conidia. IL-10 production by BMDCs in response to A. fumigatus was dependent on the expression of TLR2 and MyD88. IL-10-/- BMDCs exacerbated, whereas MyD88-/- BMDCs supplemented with exogenous IL-10 decreased the allergic pulmonary inflammation. These results indicate that TLR2/MyD88-specific recognition of PAMPs from A. fumigatus conidia can upregulate IL-10 production and downregulate lung eosinophilia and the development of a Th2 response.
Collapse
Affiliation(s)
| | | | - Giresse Tima
- Service Viral Diseases, Sciensano, Brussels, Belgium
| | - Rudi Beyaert
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, Vlaams Instituut voor Biotechnologie, Ghent, Belgium.,Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium; and
| | - Johan Grooten
- Laboratory of Molecular Immunology, Department of Biomedical Molecular Biology, Ghent University, Zwijnaarde, Belgium
| | | | | |
Collapse
|
12
|
Pollaris L, Decaesteker T, Van den Broucke S, Jonckheere AC, Cremer J, Verbeken E, Maes T, Devos FC, Vande Velde G, Nemery B, Hoet PHM, Vanoirbeek JAJ. Involvement of Innate Lymphoid Cells and Dendritic Cells in a Mouse Model of Chemical-induced Asthma. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2021; 13:295-311. [PMID: 33474863 PMCID: PMC7840869 DOI: 10.4168/aair.2021.13.2.295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/02/2020] [Accepted: 06/09/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE Exposure to low concentrations of toluene diisocyanate (TDI) leads to immune-mediated chemical-induced asthma. The role of the adaptive immune system has already been thoroughly investigated; nevertheless, the involvement of innate immune cells in the pathophysiology of chemical-induced asthma is still unresolved. The aim of the study is to investigate the role of innate lymphoid cells (ILCs) and dendritic cells (DCs) in a mouse model for chemical-induced asthma. METHODS On days 1 and 8, BALB/c mice were dermally treated (20 μL/ear) with 0.5% TDI or the vehicle acetone olive oil (AOO; 2:3). On days 15, 17, 19, 22 and 24, the mice received an oropharyngeal challenge with 0.01% TDI or AOO (1:4). One day after the last challenge, airway hyperreactivity (AHR) to methacholine was assessed, followed by an evaluation of pulmonary inflammation and immune-related parameters, including the cytokine pattern in bronchoalveolar lavage fluid, lymphocyte subpopulations of the lymph nodes and their ex vivo cytokine production profile, blood immunoglobulins and DC and ILC subpopulations in the lungs. RESULTS Both DC and ILC2 were recruited to the lungs after multiple airway exposures to TDI, regardless of the prior dermal sensitization. However, prior dermal sensitization with TDI alone results in AHR and predominant eosinophilic airway inflammation, accompanied by a typical type 2 helper T (Th2) cytokine profile. CONCLUSIONS TDI-induced asthma is mediated by a predominant type 2 immune response, with the involvement of adaptive Th2 cells. However, from our study we suggest that the innate ILC2 cells are important additional players in the development of TDI-induced asthma.
Collapse
Affiliation(s)
- Lore Pollaris
- Centre for Environment and Health, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Tatjana Decaesteker
- Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, University of Leuven, Leuven, Belgium
| | - Sofie Van den Broucke
- Centre for Environment and Health, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Anne Charlotte Jonckheere
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, University of Leuven, Leuven, Belgium
| | - Jonathan Cremer
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, University of Leuven, Leuven, Belgium
| | - Erik Verbeken
- Department of Imaging and Pathology, University of Leuven, Leuven, Belgium
| | - Tania Maes
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Fien C Devos
- Centre for Environment and Health, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Greetje Vande Velde
- Department of Imaging and Pathology, Biomedical MRI, University of Leuven, Leuven, Belgium
| | - Benoit Nemery
- Centre for Environment and Health, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Peter H M Hoet
- Centre for Environment and Health, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Jeroen A J Vanoirbeek
- Centre for Environment and Health, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium.
| |
Collapse
|
13
|
Roberts G, Almqvist C, Boyle R, Crane J, Hogan SP, Marsland B, Saglani S, Woodfolk JA. Developments in the mechanisms of allergy in 2018 through the eyes of Clinical and Experimental Allergy, Part I. Clin Exp Allergy 2020; 49:1541-1549. [PMID: 31833127 DOI: 10.1111/cea.13532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the first of two linked articles, we describe the development in the mechanisms underlying allergy as described by Clinical & Experimental Allergy and other journals in 2018. Experimental models of allergic disease, basic mechanisms and clinical mechanisms are all covered.
Collapse
Affiliation(s)
- Graham Roberts
- Clinical and Experimental Sciences and Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,The David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Isle of Wight, UK
| | - Catarina Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Pediatric Allergy and Pulmonology Unit at Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Robert Boyle
- Department of Paediatrics, Imperial College London, London, UK
| | - Julian Crane
- Department of Medicine, University of Otago Wellington, Wellington, New Zealand
| | - Simon P Hogan
- Department of Pathology, Mary H Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Ben Marsland
- Department of Immunology and Pathology, Monash University, Melbourne, Vic., Australia
| | - Segal Saglani
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Judith A Woodfolk
- Division of Asthma, Allergy and Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
14
|
Jirmo AC, Busse M, Happle C, Skuljec J, Dalüge K, Habener A, Grychtol R, DeLuca DS, Breiholz OD, Prinz I, Hansen G. IL-17 regulates DC migration to the peribronchial LNs and allergen presentation in experimental allergic asthma. Eur J Immunol 2020; 50:1019-1033. [PMID: 32142593 DOI: 10.1002/eji.201948409] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/30/2020] [Accepted: 03/05/2020] [Indexed: 01/04/2023]
Abstract
IL-17 is associated with different phenotypes of asthma, however, it is not fully elucidated how it influences induction and maintenance of asthma and allergy. In order to determine the role of IL-17 in development of allergic asthma, we used IL-17A/F double KO (IL-17A/F KO) and WT mice with or without neutralization of IL-17 in an experimental allergic asthma model and analyzed airway hyperresponsiveness, lung inflammation, T helper cell polarization, and DCs influx and activation. We report that the absence of IL-17 reduced influx of DCs into lungs and lung draining LNs. Compared to WT mice, IL-17A/F KO mice or WT mice after neutralization of IL-17A showed reduced airway hyperresponsiveness, eosinophilia, mucus hypersecretion, and IgE levels. DCs from draining LNs of allergen-challenged IL-17A/F KO mice showed a reduction in expression of migratory and costimulatory molecules CCR7, CCR2, MHC-II, and CD40 compared to WT DCs. Moreover, in vivo stimulation of adoptively transferred antigen-specific cells was attenuated in lung-draining LNs in the absence of IL-17. Thus, we report that IL-17 enhances airway DC activation, migration, and function. Consequently, lack of IL-17 leads to reduced antigen-specific T cell priming and impaired development of experimental allergic asthma.
Collapse
Affiliation(s)
- Adan Chari Jirmo
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Mandy Busse
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Christine Happle
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Jelena Skuljec
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Kathleen Dalüge
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Anika Habener
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Ruth Grychtol
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - David S DeLuca
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Oliver D Breiholz
- Research Core Unit Genomics (RCUG), Hannover Medical School, Hannover, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.,Excellence Cluster RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
15
|
Blockade of CD40L inhibits immunogenic maturation of lung dendritic cells: Implications for the role of lung iNKT cells in mouse models of asthma. Mol Immunol 2020; 121:167-185. [PMID: 32229377 DOI: 10.1016/j.molimm.2020.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 03/15/2020] [Accepted: 03/15/2020] [Indexed: 12/15/2022]
Abstract
Some studies have shown that maturation of dendritic cells (DCs) is modulated directly by pathogen components via pattern recognition receptors such as Toll-like receptors, but also by signal like CD40 ligand (CD40 L or CD154) mediated by activated T cells. Several reports indicate that invariant natural killer T (iNKT) cells up-regulate CD40 L upon stimulation and thereby induce activation and maturation of DCs through crosslink with CD40. Our previous findings indicated that iNKT cells promote Th2 cell responses through the induction of immunogenic maturation of lung DCs (LDCs) in the asthmatic murine, but its mechanism remains unclear. Therefore, we investigated the immunomodulatory effects of blockade of CD40 L using anti-CD40 L treatment on Th2 cell responses and immunogenic maturation of LDCs, and further analyzed whether these influences of blockade of CD40 L were related to lung iNKT cells using iNKT cell-deficient mice and the combination treatment of specific iNKT cell activation with anti-CD40 L treatment in murine models of asthma. Our findings showed that blockade of CD40 L using anti-CD40 L treatment attenuated Th2 cell responses in wild-type (WT) mice, but not in CD1d-deficient mice sensitized and challenged with ovalbumin (OVA) or house dust mite (HDM). Meanwhile, blockade of CD40 L down-regulated immunogenic maturation of LDCs in WT mice, but not in CD1d-deficient mice sensitized and challenged with OVA. Additionally, agonistic anti-CD40 treatment reversed the inhibitory effects of anti-CD40 L treatment on Th2 cell responses and LDC activation in an OVA-induced mouse model of asthma. Furthermore, LDCs from asthmatic mice treated with anti-CD40 L could significantly reduce the influence on Th2 cell responses in vivo and in vitro. Finally, α-Galactosylceramide plus anti-CD40 L treatment stimulated lung iNKT cells, but suppressed Th2 cell responses in the asthmatic mice. Taken together, our data raise an evidence that blockade of CD40 L attenuates Th2 cell responses through the inhibition of immunogenic maturation of LDCs, which may be at least partially related to lung iNKT cells in murine models of asthma.
Collapse
|
16
|
Hudon Thibeault AA, Laprise C. Cell-Specific DNA Methylation Signatures in Asthma. Genes (Basel) 2019; 10:E932. [PMID: 31731604 PMCID: PMC6896152 DOI: 10.3390/genes10110932] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/08/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022] Open
Abstract
Asthma is a complex trait, often associated with atopy. The genetic contribution has been evidenced by familial occurrence. Genome-wide association studies allowed for associating numerous genes with asthma, as well as identifying new loci that have a minor contribution to its phenotype. Considering the role of environmental exposure on asthma development, an increasing amount of literature has been published on epigenetic modifications associated with this pathology and especially on DNA methylation, in an attempt to better understand its missing heritability. These studies have been conducted in different tissues, but mainly in blood or its peripheral mononuclear cells. However, there is growing evidence that epigenetic changes that occur in one cell type cannot be directly translated into another one. In this review, we compare alterations in DNA methylation from different cells of the immune system and of the respiratory tract. The cell types in which data are obtained influences the global status of alteration of DNA methylation in asthmatic individuals compared to control (an increased or a decreased DNA methylation). Given that several genes were cell-type-specific, there is a great need for comparative studies on DNA methylation from different cells, but from the same individuals in order to better understand the role of epigenetics in asthma pathophysiology.
Collapse
Affiliation(s)
- Andrée-Anne Hudon Thibeault
- Département des sciences fondamentales, Université du Québec à Chicoutimi (UQAC), Saguenay, G7H 2B1 QC, Canada;
- Centre intersectoriel en santé durable (CISD), Université du Québec à Chicoutimi (UQAC), Saguenay, G7H 2B1 QC, Canada
- Quebec Respiratory Health Network, Quebec, G1V 4G5 QC, Canada
| | - Catherine Laprise
- Département des sciences fondamentales, Université du Québec à Chicoutimi (UQAC), Saguenay, G7H 2B1 QC, Canada;
- Centre intersectoriel en santé durable (CISD), Université du Québec à Chicoutimi (UQAC), Saguenay, G7H 2B1 QC, Canada
- Quebec Respiratory Health Network, Quebec, G1V 4G5 QC, Canada
| |
Collapse
|
17
|
Chen X, Qiu C. Respiratory tract mucous membrane microecology and asthma. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:495. [PMID: 31700931 PMCID: PMC6803190 DOI: 10.21037/atm.2019.09.06] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
According to the world health organization, the increasing incidence of asthma is placing a heavy burden on the social economy. Its high rate of disability and mortality has become a serious social and public health problem. Asthma is a heterogeneous disease in which genetic polymorphism interacts with environmental factors. Because the pathogenesis of asthma is not completely clear, there is no specific treatment. In 2010, 16S rRNA gene sequencing showed that lungs have many different microbial communities in both healthy and sick states. These microbial communities and respiratory mucosa constitute the respiratory mucosal microecology. When the respiratory mucosal microecology changes, it can play a key role in the occurrence and development of asthma and other respiratory diseases by regulating the immune mechanism. This paper reviews the latest research results in this field, and tries to explore the effects of changes in respiratory mucosal microecology on the pathogenesis of asthma, so as to provide new methods for early diagnosis, treatment and prevention of asthma.
Collapse
Affiliation(s)
- Xingyuan Chen
- Department of Respiratory and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen 518020, China
| | - Chen Qiu
- Department of Respiratory and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen 518020, China
| |
Collapse
|
18
|
Pan H, Zhang G, Nie H, Li S, He S, Yang J. Sulfatide-activated type II NKT cells suppress immunogenic maturation of lung dendritic cells in murine models of asthma. Am J Physiol Lung Cell Mol Physiol 2019; 317:L578-L590. [PMID: 31432714 DOI: 10.1152/ajplung.00256.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Our previous study showed that sulfatide-activated type II natural killer T (NKT) cells can prevent allergic airway inflammation in an ovalbumin (OVA)-induced murine model of asthma, but the underlying mechanism is unclear. Recently, sulfatide-activated type II NKT cells were shown to modulate the function of dendritic cells in experimental autoimmune encephalomyelitis and nonobese diabetic mice. Thus, it was hypothesized that sulfatide-activated type II NKT cells may modulate the function of lung dendritic cells (LDCs) in asthmatic mice. Our data showed that, in our mouse models, activation of type II NKT cells by sulfatide administration and adoptive transfer of sulfatide-activated type II NKT cells resulted in reduced expression of surface maturation markers and proinflammatory cytokine production of LDCs. LDCs from sulfatide-treated asthmatic mice, in contrast to LDCs from PBS-treated asthmatic mice, significantly reduced allergic airway inflammation in vivo. However, we found no influence of sulfatide-activated type II NKT cells on the phenotypic and functional maturation of bone marrow-derived dendritic cells in vitro. In addition, adoptive transfer of sulfatide-activated type II NKT cells did not influence the phenotypic and functional maturation of LDCs in CD1d-/- mice, which lack both type I and II NKT cells, immunized and challenged with OVA. Our data reveal that sulfatide-activated type II NKT cells can suppress immunogenic maturation of LDCs to reduce allergic airway inflammation in mouse models of asthma, and it is possible that the immunomodulatory effect needs type I NKT cells.
Collapse
Affiliation(s)
- Huaqin Pan
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Guqin Zhang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hanxiang Nie
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shuhua Li
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shaojun He
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiong Yang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
19
|
Del Prete A, Scutera S, Sozzani S, Musso T. Role of osteopontin in dendritic cell shaping of immune responses. Cytokine Growth Factor Rev 2019; 50:19-28. [PMID: 31126876 DOI: 10.1016/j.cytogfr.2019.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023]
Abstract
Osteopontin (OPN) is a pleiotropic cytokine produced both by immune and non-immune cells and active on different cellular targets. OPN production has been associated with several pathological conditions, including autoimmune diseases (e.g. lupus, multiple sclerosis and rheumatoid arthritis) and cancer. Emerging evidence suggests that the role of OPN has been underestimated, as it seems to be working at multiple levels of immune regulation, such as the shaping of T cell effector responses, the regulation of the tumor microenvironment, and the functional interaction with mesenchymal stromal cells. In this context, dendritic cells (DCs) play a crucial role being both an important source and a cellular target for OPN action. DC family is composed by several cell subsets endowed with specific immune functions. OPN exerts its biological functions through multiple receptors and is produced in different intracellular and secreted forms. OPN production by DC subsets is emerging as a crucial mechanism of regulation in normal and pathological conditions and starts to be exploited as a therapeutic target. This review will focus on the role of DC-derived OPN in shaping immune response and on the complex role of this cytokines in the regulation in immune response.
Collapse
Affiliation(s)
- Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Humanitas Clinical and Research Center-IRCCS Rozzano-Milano, Italy
| | - Sara Scutera
- Microbiology section, Department of Public Health and Pediatric Sciences, University of Torino, Turin, Italy
| | - Silvano Sozzani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Tiziana Musso
- Microbiology section, Department of Public Health and Pediatric Sciences, University of Torino, Turin, Italy
| |
Collapse
|
20
|
Pollaris L, Van Den Broucke S, Decaesteker T, Cremer J, Seys S, Devos FC, Provoost S, Maes T, Verbeken E, Vande Velde G, Nemery B, Hoet PHM, Vanoirbeek JAJ. Dermal exposure determines the outcome of repeated airway exposure in a long-term chemical-induced asthma-like mouse model. Toxicology 2019; 421:84-92. [PMID: 31071364 DOI: 10.1016/j.tox.2019.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/24/2019] [Accepted: 05/05/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Exposure to diisocyanates is an important cause of occupational asthma (OA) in the industrialized world. Since OA occurs after long-term exposure to diisocyanates, we developed a chronic mouse model of chemical-induced asthma where toluene diisocyanate (TDI) was administered at two different exposure sites. OBJECTIVES Evaluating the effect of long-term respiratory isocyanate exposure - with or without prior dermal exposure- on sensitization, inflammatory responses and airway hyperreactivity (AHR). METHODS On days 1 and 8, BALB/c mice were dermally treated (20 μl/ear) with 0.5% 2,4-toluene diisocyanate TDI or the vehicle acetone olive oil (AOO) (3:2). Starting from day 15, mice received intranasal instillations with 0.1% TDI of vehicle five times in a week, for five successive weeks. One day after the last instillation airway hyperreactivity (AHR) to methacholine was assessed, followed by an evaluation of pulmonary inflammation and structural lung changes. Immune-related parameters were assessed in the lungs (BAL and tissue), blood, cervical- and auricular lymph nodes. RESULTS Mice repeatedly intranasally exposed to TDI showed systemic sensitization and a mixed Th1/Th2 type immune response, without the presence of AHR. However, when mice are first dermally sensitized with TDI, followed by repeated intranasal TDI challenges, this results in a pronounced Th2 response and AHR. CONCLUSION Dermal exposure to TDI determines airway hyperreactivity after repeated airway exposure to TDI.
Collapse
Affiliation(s)
- Lore Pollaris
- Centre for Environment and Health, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Sofie Van Den Broucke
- Centre for Environment and Health, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Tatjana Decaesteker
- Laboratory of Respiratory Diseases, Department of Chronic Diseases, Metabolism and Ageing, University of Leuven, Leuven, Belgium
| | - Jonathan Cremer
- Laboratory of Clinical Immunology, Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Sven Seys
- Laboratory of Clinical Immunology, Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Fien C Devos
- Centre for Environment and Health, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Sharen Provoost
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Tania Maes
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Erik Verbeken
- Translational Cell and Tissue Research, Department of Imaging and Pathology, University of Leuven, Leuven, Belgium
| | | | - Benoit Nemery
- Centre for Environment and Health, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Peter H M Hoet
- Centre for Environment and Health, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Jeroen A J Vanoirbeek
- Centre for Environment and Health, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium.
| |
Collapse
|
21
|
Chen Z, Wang L. Ovalbumin induces natural killer cells to secrete Th2 cytokines IL‑5 and IL‑13 in a mouse model of asthma. Mol Med Rep 2019; 19:3210-3216. [PMID: 30816463 PMCID: PMC6423558 DOI: 10.3892/mmr.2019.9966] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 02/14/2019] [Indexed: 01/08/2023] Open
Abstract
Asthma is a chronic lung disease characterized by an imbalance of T-helper (Th)1/Th2 cells and their cytokine profiles. Natural killer (NK) cells constitute a considerable subset of the lymphocyte population in the lungs, and provide protection against respiratory infection by fungi, bacteria and viruses. However, the mechanism by which NK cells are involved in asthma remains to be fully elucidated. The present study analyzed the dynamic changes of NK cells and their subsets during the development of the ovalbumin (OVA)-induced allergic airway response. Lung tissues were histologically examined for cell infiltration and mucus hypersecretion. The number, activity and cytokine-secreting ability of NK cells was determined by flow cytometry. The results showed that the percentage of NK cells in the lung was decreased following OVA sensitization and challenge. However, NK cells exhibited enhanced activity and secreted more Th2 cytokines (IL-5 and IL-13) following OVA challenge. Furthermore, the proportion of CD11b− NK subsets increased with the development of asthma, and CD11b− CD27− NK cells were the primary NK subset producing Th2 cytokines. These findings suggest that, although NK cells are not the crucial type of lymphocytes involved in asthma, OVA induces NK cells to secrete Th2 cytokines that may be involved in the pathogenesis of asthma.
Collapse
Affiliation(s)
- Zhangbo Chen
- Department of Pathogen Biology and Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Lu Wang
- Department of Physiology and Chinese‑German Stem Cell Center, Tongji Medical College, Huazhong University of Science and Technology, The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
22
|
Mesenchymal stem cells for inflammatory airway disorders: promises and challenges. Biosci Rep 2019; 39:BSR20182160. [PMID: 30610158 PMCID: PMC6356012 DOI: 10.1042/bsr20182160] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 12/19/2018] [Accepted: 01/03/2019] [Indexed: 02/06/2023] Open
Abstract
The regenerative and immunomodulatory characteristics of mesenchymal stem cells (MSCs) make them attractive in the treatment of many diseases. Although they have shown promising preclinical studies of immunomodulation and paracrine effects in inflammatory airway disorders and other lung diseases, there are still challenges that have to be overcome before MSCs can be safely, effectively, and routinely applied in the clinical setting. A good understanding of the roles and mechanisms of the MSC immunomodulatory effects will benefit the application of MSC-based clinical therapy. In this review, we summarize the promises and challenges of the preclinical and clinical trials of MSC therapies, aiming to better understand the role that MSCs play in attempt to treat inflammatory airway disorders.
Collapse
|
23
|
The Role of the Microbiome in Asthma: The Gut⁻Lung Axis. Int J Mol Sci 2018; 20:ijms20010123. [PMID: 30598019 PMCID: PMC6337651 DOI: 10.3390/ijms20010123] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/22/2018] [Accepted: 12/27/2018] [Indexed: 12/14/2022] Open
Abstract
Asthma is one of the most common chronic respiratory diseases worldwide. It affects all ages but frequently begins in childhood. Initiation and exacerbations may depend on individual susceptibility, viral infections, allergen exposure, tobacco smoke exposure, and outdoor air pollution. The aim of this review was to analyze the role of the gut⁻lung axis in asthma development, considering all asthma phenotypes, and to evaluate whether microbe-based therapies may be used for asthma prevention. Several studies have confirmed the role of microbiota in the regulation of immune function and the development of atopy and asthma. These clinical conditions have apparent roots in an insufficiency of early life exposure to the diverse environmental microbiota necessary to ensure colonization of the gastrointestinal and/or respiratory tracts. Commensal microbes are necessary for the induction of a balanced, tolerogenic immune system. The identification of commensal bacteria in both the gastroenteric and respiratory tracts could be an innovative and important issue. In conclusion, the function of microbiota in healthy immune response is generally acknowledged, and gut dysbacteriosis might result in chronic inflammatory respiratory disorders, particularly asthma. Further investigations are needed to improve our understanding of the role of the microbiome in inflammation and its influence on important risk factors for asthma, including tobacco smoke and host genetic features.
Collapse
|
24
|
Han M, Hu R, Ma J, Zhang B, Chen C, Li H, Yang J, Huang G. Fas Signaling in Dendritic Cells Mediates Th2 Polarization in HDM-Induced Allergic Pulmonary Inflammation. Front Immunol 2018; 9:3045. [PMID: 30619373 PMCID: PMC6308134 DOI: 10.3389/fimmu.2018.03045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 12/10/2018] [Indexed: 01/17/2023] Open
Abstract
Fas-Fas ligand (FasL) signaling plays an important role in the development of allergic inflammation, but the cellular and molecular mechanisms are still not well known. By using the bone marrow-derived dendritic cell (BMDC) transfer-induced pulmonary inflammation model, we found that house dust mite (HDM)-stimulated FAS-deficient BMDCs induced higher Th2-mediated allergic inflammation, associated with increased mucus production and eosinophilic inflammation. Moreover, FAS-deficient BMDCs promoted Th2 cell differentiation upon HDM stimulation in vitro. Compared to wild-type BMDCs, the Fas-deficient BMDCs had increased ERK activity and decreased IL-12 production upon HDM stimulation. Inhibition of ERK activity could largely increase IL-12 production, consequently restored the increased Th2 cytokine expression of OT-II CD4+ T cells activated by Fas-deficient BMDCs. Thus, our results uncover an important role of DC-specific Fas signaling in Th2 differentiation and allergic inflammation, and modulation of Fas signaling in DCs may offer a useful strategy for the treatment of allergic inflammatory diseases.
Collapse
Affiliation(s)
- Miaomiao Han
- Department of Otolaryngology-Head and Neck Surgery, Center for Allergic and Inflammatory Diseases, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ran Hu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyu Ma
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baohua Zhang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ce Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Huabin Li
- Department of Otolaryngology-Head and Neck Surgery, Center for Allergic and Inflammatory Diseases, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Jun Yang
- Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gonghua Huang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| |
Collapse
|
25
|
Singh V, Phukan UJ. Interaction of host and Staphylococcus aureus protease-system regulates virulence and pathogenicity. Med Microbiol Immunol 2018; 208:585-607. [PMID: 30483863 DOI: 10.1007/s00430-018-0573-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 11/22/2018] [Indexed: 02/06/2023]
Abstract
Staphylococcus aureus causes various health care- and community-associated infections as well as certain chronic TH2 driven inflammatory diseases. It is a potent pathogen with serious virulence and associated high morbidity. Severe pathogenicity is accredited to the S. aureus secreted virulence factors such as proteases and host protease modulators. These virulence factors promote adhesion and invasion of bacteria through damage of tight junction barrier and keratinocytes. They inhibit activation and transmigration of various immune cells such as neutrophils (and neutrophil proteases) to evade opsono-phagocytosis and intracellular bacterial killing. Additionally, they protect the bacteria from extracellular killing by disrupting integrity of extracellular matrix. Platelet activation and agglutination is also impaired by these factors. They also block the classical as well as alternative pathways of complement activation and assist in spread of infection through blood and tissue. As these factors are exquisite factors of S. aureus mediated disease development, we have focused on review of diversification of various protease-system associated virulence factors, their structural building, diverse role in disease development and available therapeutic counter measures. This review summarises the role of protease-associated virulence factors during invasion and progression of disease.
Collapse
Affiliation(s)
- Vigyasa Singh
- Molecular Bioprospection Department, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, India
| | - Ujjal Jyoti Phukan
- School of Life Science, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
26
|
Movassagh H, Koussih L, Shan L, Gounni AS. The regulatory role of semaphorin 3E in allergic asthma. Int J Biochem Cell Biol 2018; 106:68-73. [PMID: 30447428 DOI: 10.1016/j.biocel.2018.11.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 11/06/2018] [Accepted: 11/12/2018] [Indexed: 10/27/2022]
Abstract
Semaphorins were originally discovered as essential mediators involved in regulation of axonal growth during development of the nervous system. Ubiquitously expressed on various organs, they control several cellular functions by regulating essential signaling pathways. Among them, semaphorin3E binds plexinD1 as the primary receptor and mediates regulatory effects on cell migration, proliferation, and angiogenesis considered major physiological and pathological features in health and disease. Recent in vitro and in vivo experimental evidence demonstrate a key regulator role of semaphorin3E on airway inflammation, hyperresponsivenss and remodeling in allergic asthma. Herein, we aim to provide a broad overview of the biology of semaphorin family and review the recently discovered regulatory role of semaphorin3E in modulating immune cells and structural cells function in the airways. These findings support the concept of semaphorin3E/plexinD1 axis as a therapeutic target in allergic asthma.
Collapse
Affiliation(s)
- Hesam Movassagh
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Latifa Koussih
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lianyu Shan
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Abdelilah S Gounni
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
27
|
Walwyn-Brown K, Guldevall K, Saeed M, Pende D, Önfelt B, MacDonald AS, Davis DM. Human NK Cells Lyse Th2-Polarizing Dendritic Cells via NKp30 and DNAM-1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2028-2041. [PMID: 30120122 PMCID: PMC6139540 DOI: 10.4049/jimmunol.1800475] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/23/2018] [Indexed: 01/08/2023]
Abstract
Cross-talk between NK cells and dendritic cells (DCs) is important in Th1 immune responses, including antitumor immunity and responses to infections. DCs also play a crucial role in polarizing Th2 immunity, but the impact of NK cell-DC interactions in this context remains unknown. In this study, we stimulated human monocyte-derived DCs in vitro with different pathogen-associated molecules: LPS or polyinosinic-polycytidylic acid, which polarize a Th1 response, or soluble egg Ag from the helminth worm Schistosoma mansoni, a potent Th2-inducing Ag. Th2-polarizing DCs were functionally distinguishable from Th1-polarizing DCs, and both showed distinct morphology and dynamics from immature DCs. We then assessed the outcome of autologous NK cells interacting with these differently stimulated DCs. Confocal microscopy showed polarization of the NK cell microtubule organizing center and accumulation of LFA-1 at contacts between NK cells and immature or Th2-polarizing DCs but not Th1-polarizing DCs, indicative of the assembly of an activating immune synapse. Autologous NK cells lysed immature DCs but not DCs treated with LPS or polyinosinic-polycytidylic acid as reported previously. In this study, we demonstrated that NK cells also degranulated in the presence of Th2-polarizing DCs. Moreover, time-lapse live-cell microscopy showed that DCs that had internalized fluorescently labeled soluble egg Ag were efficiently lysed. Ab blockade of NK cell-activating receptors NKp30 or DNAM-1 abrogated NK cell lysis of Th2-polarizing DCs. Thus, these data indicate a previously unrecognized role of NK cell cytotoxicity and NK cell-activating receptors NKp30 and DNAM-1 in restricting the pool of DCs involved in Th2 immune responses.
Collapse
Affiliation(s)
- Katherine Walwyn-Brown
- Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Karolin Guldevall
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, SE-106 91 Stockholm, Sweden
| | - Mezida Saeed
- Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Daniela Pende
- Laboratorio Immunologia, Istituto di Ricovero e Cura a Carattere Scientifico, Ospedale Policlinico San Martino, 16132 Genova, Italy; and
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, SE-106 91 Stockholm, Sweden
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Andrew S MacDonald
- Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Daniel M Davis
- Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom;
| |
Collapse
|
28
|
Beckert H, Meyer-Martin H, Buhl R, Taube C, Reuter S. The Canonical but Not the Noncanonical Wnt Pathway Inhibits the Development of Allergic Airway Disease. THE JOURNAL OF IMMUNOLOGY 2018; 201:1855-1864. [PMID: 30135183 DOI: 10.4049/jimmunol.1800554] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/26/2018] [Indexed: 12/27/2022]
Abstract
Asthma is a syndrome with multifactorial causes, resulting in a variety of different phenotypes. Current treatment options are not curative and are sometimes ineffective in certain disease phenotypes. Therefore, novel therapeutic approaches are required. Recent findings have shown that activation of the canonical Wnt signaling pathway suppresses the development of allergic airway disease. In contrast, the effect of the noncanonical Wnt signaling pathway activation on allergic airway disease is not well described. The aim of this study was to validate the therapeutic effectiveness of Wnt-1-driven canonical Wnt signaling compared with Wnt-5a-driven noncanonical signaling in murine models. In vitro, both ligands were capable of attenuating allergen-specific T cell activation in a dendritic cell-dependent manner. In addition, the therapeutic effects of Wnt ligands were assessed in two different models of allergic airway disease. Application of Wnt-1 resulted in suppression of airway inflammation as well as airway hyperresponsiveness and mucus production. In contrast, administration of Wnt-5a was less effective in reducing airway inflammation or goblet cell metaplasia. These results suggest an immune modulating function for canonical as well as noncanonical Wnt signaling, but canonical Wnt pathway activation appears to be more effective in suppressing allergic airway disease than noncanonical Wnt activation.
Collapse
Affiliation(s)
- Hendrik Beckert
- Department of Pulmonary Medicine, University Medical Center Essen-Ruhrlandklinik, Essen, North Rhine-Westphalia 45239, Germany; and
| | - Helen Meyer-Martin
- Department of Pulmonary Medicine, III. Medical Clinic, University Medical Center of the Johannes Gutenberg University, D-55131 Mainz, Germany
| | - Roland Buhl
- Department of Pulmonary Medicine, III. Medical Clinic, University Medical Center of the Johannes Gutenberg University, D-55131 Mainz, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Medical Center Essen-Ruhrlandklinik, Essen, North Rhine-Westphalia 45239, Germany; and
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Medical Center Essen-Ruhrlandklinik, Essen, North Rhine-Westphalia 45239, Germany; and
| |
Collapse
|
29
|
Teufelberger AR, Nordengrün M, Braun H, Maes T, De Grove K, Holtappels G, O'Brien C, Provoost S, Hammad H, Gonçalves A, Beyaert R, Declercq W, Vandenabeele P, Krysko DV, Bröker BM, Bachert C, Krysko O. The IL-33/ST2 axis is crucial in type 2 airway responses induced by Staphylococcus aureus –derived serine protease–like protein D. J Allergy Clin Immunol 2018; 141:549-559.e7. [DOI: 10.1016/j.jaci.2017.05.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 04/26/2017] [Accepted: 05/08/2017] [Indexed: 01/09/2023]
|
30
|
Liu MC, Xiao HQ, Breslin LM, Bochner BS, Schroeder JT. Enhanced antigen presenting and T cell functions during late-phase allergic responses in the lung. Clin Exp Allergy 2017; 48:334-342. [PMID: 29105205 DOI: 10.1111/cea.13054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUND Allergic inflammation is a common feature of asthma and may contribute to both development and perpetuation of disease. The interaction of antigen-presenting cells (APC) with sensitized helper T lymphocytes (TC) producing Th2 cytokines may determine the inflammatory response. Recruitment of APC and TC to the lung during allergic responses has been demonstrated, but functional studies in humans have been limited. OBJECTIVE This study examined the function of APC and TC accumulating at sites of inflammation after segmental allergen challenge (SAC). METHODS Fifteen allergic patients underwent SAC, and cells from bronchoalveolar lavage (BAL) were collected after 24 hours. APC and TC from the blood and BAL were purified based on expression of the monocyte marker, CD14; the plasmacytoid dendritic cell (pDC) marker, BDCA4, identifying neuropilin-1 (NRP1); and the helper T cell marker, CD4. Functional activity was assessed using allergen-induced T cell proliferation. Flow cytometry identified cells expressing CD14 and NRP1. RESULTS SAC resulted in a 12-fold increase in mononuclear cells having the morphologic appearance of blood monocytes. Most of these cells co-expressed CD14 and NRP1. After saline challenge, BAL mononuclear cells demonstrated little APC function. Following SAC, BAL mononuclear cells showed function equal to pDC from blood and greater than blood monocytes. Purified NRP1+ cells from BAL had even greater function than pDC cells from blood (P = .008). Using consistent sources of APC, enhanced proliferation of TC from lung compared to blood was also demonstrated (P = .002). CONCLUSIONS The marked increase in APC function for allergen-specific TC proliferation during allergic inflammation is largely due to the recruitment of monocytes and dendritic cells. There is also an enhanced response in the lung TC population, consistent with recruitment of allergen-specific T cells. Interactions between recruited APC and TC may occur as an early event promoting allergic airway inflammation.
Collapse
Affiliation(s)
- M C Liu
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - H Q Xiao
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - L M Breslin
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - B S Bochner
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - J T Schroeder
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
31
|
Muehling LM, Lawrence MG, Woodfolk JA. Pathogenic CD4 + T cells in patients with asthma. J Allergy Clin Immunol 2017; 140:1523-1540. [PMID: 28442213 PMCID: PMC5651193 DOI: 10.1016/j.jaci.2017.02.025] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 02/07/2017] [Accepted: 02/20/2017] [Indexed: 02/08/2023]
Abstract
Asthma encompasses a variety of clinical phenotypes that involve distinct T cell-driven inflammatory processes. Improved understanding of human T-cell biology and the influence of innate cytokines on T-cell responses at the epithelial barrier has led to new asthma paradigms. This review captures recent knowledge on pathogenic CD4+ T cells in asthmatic patients by drawing on observations in mouse models and human disease. In patients with allergic asthma, TH2 cells promote IgE-mediated sensitization, airway hyperreactivity, and eosinophilia. Here we discuss recent discoveries in the myriad molecular pathways that govern the induction of TH2 differentiation and the critical role of GATA-3 in this process. We elaborate on how cross-talk between epithelial cells, dendritic cells, and innate lymphoid cells translates to T-cell outcomes, with an emphasis on the actions of thymic stromal lymphopoietin, IL-25, and IL-33 at the epithelial barrier. New concepts on how T-cell skewing and epitope specificity are shaped by multiple environmental cues integrated by dendritic cell "hubs" are discussed. We also describe advances in understanding the origins of atypical TH2 cells in asthmatic patients, the role of TH1 cells and other non-TH2 types in asthmatic patients, and the features of T-cell pathogenicity at the single-cell level. Progress in technologies that enable highly multiplexed profiling of markers within a single cell promise to overcome barriers to T-cell discovery in human asthmatic patients that could transform our understanding of disease. These developments, along with novel T cell-based therapies, position us to expand the assortment of molecular targets that could facilitate personalized treatments.
Collapse
Affiliation(s)
- Lyndsey M Muehling
- Allergy Division, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Va
| | - Monica G Lawrence
- Allergy Division, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Va
| | - Judith A Woodfolk
- Allergy Division, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Va.
| |
Collapse
|
32
|
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]
|
33
|
Machiels B, Dourcy M, Xiao X, Javaux J, Mesnil C, Sabatel C, Desmecht D, Lallemand F, Martinive P, Hammad H, Guilliams M, Dewals B, Vanderplasschen A, Lambrecht BN, Bureau F, Gillet L. A gammaherpesvirus provides protection against allergic asthma by inducing the replacement of resident alveolar macrophages with regulatory monocytes. Nat Immunol 2017; 18:1310-1320. [PMID: 29035391 DOI: 10.1038/ni.3857] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 09/20/2017] [Indexed: 12/12/2022]
Abstract
The hygiene hypothesis postulates that the recent increase in allergic diseases such as asthma and hay fever observed in Western countries is linked to reduced exposure to childhood infections. Here we investigated how infection with a gammaherpesvirus affected the subsequent development of allergic asthma. We found that murid herpesvirus 4 (MuHV-4) inhibited the development of house dust mite (HDM)-induced experimental asthma by modulating lung innate immune cells. Specifically, infection with MuHV-4 caused the replacement of resident alveolar macrophages (AMs) by monocytes with regulatory functions. Monocyte-derived AMs blocked the ability of dendritic cells to trigger a HDM-specific response by the TH2 subset of helper T cells. Our results indicate that replacement of embryonic AMs by regulatory monocytes is a major mechanism underlying the long-term training of lung immunity after infection.
Collapse
Affiliation(s)
- Bénédicte Machiels
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine - FARAH, University of Liège, Liège, Belgium
| | - Mickael Dourcy
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine - FARAH, University of Liège, Liège, Belgium
| | - Xue Xiao
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine - FARAH, University of Liège, Liège, Belgium
| | - Justine Javaux
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine - FARAH, University of Liège, Liège, Belgium
| | - Claire Mesnil
- Cellular and Molecular Immunology, Department of Functional Sciences, Faculty of Veterinary Medicine - GIGA, University of Liège, Liège, Belgium
| | - Catherine Sabatel
- Cellular and Molecular Immunology, Department of Functional Sciences, Faculty of Veterinary Medicine - GIGA, University of Liège, Liège, Belgium
| | - Daniel Desmecht
- Department of Pathology, Faculty of Veterinary Medicine - FARAH, University of Liège, Liège, Belgium
| | | | | | - Hamida Hammad
- VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Martin Guilliams
- VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Benjamin Dewals
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine - FARAH, University of Liège, Liège, Belgium
| | - Alain Vanderplasschen
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine - FARAH, University of Liège, Liège, Belgium
| | - Bart N Lambrecht
- VIB Center for Inflammation Research, Ghent University, Ghent, Belgium.,Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Fabrice Bureau
- Cellular and Molecular Immunology, Department of Functional Sciences, Faculty of Veterinary Medicine - GIGA, University of Liège, Liège, Belgium
| | - Laurent Gillet
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine - FARAH, University of Liège, Liège, Belgium
| |
Collapse
|
34
|
Liu H, Jakubzick C, Osterburg AR, Nelson RL, Gupta N, McCormack FX, Borchers MT. Dendritic Cell Trafficking and Function in Rare Lung Diseases. Am J Respir Cell Mol Biol 2017; 57:393-402. [PMID: 28586276 PMCID: PMC5650088 DOI: 10.1165/rcmb.2017-0051ps] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 06/06/2017] [Indexed: 12/14/2022] Open
Abstract
Dendritic cells (DCs) are highly specialized immune cells that capture antigens and then migrate to lymphoid tissue and present antigen to T cells. This critical function of DCs is well defined, and recent studies further demonstrate that DCs are also key regulators of several innate immune responses. Studies focused on the roles of DCs in the pathogenesis of common lung diseases, such as asthma, infection, and cancer, have traditionally driven our mechanistic understanding of pulmonary DC biology. The emerging development of novel DC reagents, techniques, and genetically modified animal models has provided abundant data revealing distinct populations of DCs in the lung, and allow us to examine mechanisms of DC development, migration, and function in pulmonary disease with unprecedented detail. This enhanced understanding of DCs permits the examination of the potential role of DCs in diseases with known or suspected immunological underpinnings. Recent advances in the study of rare lung diseases, including pulmonary Langerhans cell histiocytosis, sarcoidosis, hypersensitivity pneumonitis, and pulmonary fibrosis, reveal expanding potential pathogenic roles for DCs. Here, we provide a review of DC development, trafficking, and effector functions in the lung, and discuss how alterations in these DC pathways contribute to the pathogenesis of rare lung diseases.
Collapse
Affiliation(s)
- Huan Liu
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Claudia Jakubzick
- Department of Immunology and Microbiology, National Jewish Health and University of Colorado, Denver, Colorado; and
| | - Andrew R. Osterburg
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Rebecca L. Nelson
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Nishant Gupta
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati, Cincinnati, Ohio
- Cincinnati Veteran’s Affairs Medical Center, Cincinnati, Ohio
| | - Francis X. McCormack
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati, Cincinnati, Ohio
- Cincinnati Veteran’s Affairs Medical Center, Cincinnati, Ohio
| | - Michael T. Borchers
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati, Cincinnati, Ohio
- Cincinnati Veteran’s Affairs Medical Center, Cincinnati, Ohio
| |
Collapse
|
35
|
Li Y, Yu Q, Zhao W, Zhang J, Liu W, Huang M, Zeng X. Oligomeric proanthocyanidins attenuate airway inflammation in asthma by inhibiting dendritic cells maturation. Mol Immunol 2017; 91:209-217. [PMID: 28963930 DOI: 10.1016/j.molimm.2017.09.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/18/2017] [Accepted: 09/22/2017] [Indexed: 12/23/2022]
Abstract
To date, although a promising anti-inflammatory activity of oligomeric proanthocyanidins (OPCs) has been observed in asthma, the mechanism responsible for these immunomodulatory properties remains obscure. Dendritic cells (DCs) that reside in the airway have been widely perceived as an important contributor to asthma. Our study was to demonstrate OPCs' effects on maturation and immunoregulation of pulmonary CD11c+ dendritic cells (DCs). BALB/c mice were exposed to ovalbumin (OVA) to induce murine model of asthma. In addition, pulmonary DCs and bone marrow-derived DCs (BMDCs) cultures were used to evaluate impacts of OPCs on DCs function. The results obtained here indicated that OPCs treatment dramatically reduced airway inflammation, such as the infiltration of inflammatory cells and the levels of allergen-specific serum IgE and Th2 cytokines. The expression of co-stimulatory molecules especially CD86 distributed on pulmonary DCs and bone marrow-derived DCs (BMDCs) also markedly declined. The phosphorylation of cAMP responsive element-binding protein (CREB) was significantly inhibited while no changes were observed in the expression of cAMP responsive element modulator (CREM). By transferring BMDCs into the airways of naïve mice, we found that OPCs-treated DCs (DC+OVA+OPC) were much less potent in promoting CD4+ T cells proliferation than OVA-pulsed DCs (DC+OVA), followed by the ameliorated eosinophilic inflammation in airway. Our findings tailor a novel profile of OPCs in the regulation of DCs function, shedding new light on the therapeutic potential of OPCs in asthma management.
Collapse
Affiliation(s)
- Yeshan Li
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Qijun Yu
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Wenxue Zhao
- Lung Biology Center, Department of Medicine, University of California San Francisco, San Francisco, California 94143, USA
| | - Jiaxiang Zhang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Wentao Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 140 Guangzhou Road, Nanjing, Jiangsu 210017, China
| | - Mao Huang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China.
| | - Xiaoning Zeng
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
36
|
He Q, Liu L, Yang Q, Wang A, Chen S, Li R, Huang Y, Zhang G, Ding X, Yu H, Hu S, Nie H. Invariant natural killer T cells promote immunogenic maturation of lung dendritic cells in mouse models of asthma. Am J Physiol Lung Cell Mol Physiol 2017; 313:L973-L990. [PMID: 28912381 DOI: 10.1152/ajplung.00340.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 01/17/2023] Open
Abstract
Our previous study showed that invariant natural killer T (iNKT) cells might act as an adjuvant to promote Th2 inflammatory responses in an OVA-induced mouse model of allergic asthma, but the mechanism remains unknown. To clarify the underlying mechanism through which iNKT cells promote Th2 inflammatory responses, we investigated the modulatory influence of iNKT cells on phenotypic and functional maturation of lung dendritic cells (LDCs) using iNKT cell-knockout mice, specific iNKT cell activation, coculture experiments, and adoptive transfer of iNKT cells in mouse models of asthma. Our data showed that iNKT cell deficiency could downregulate surface maturation markers and proinflammatory cytokine secretion of LDCs from a mouse model of asthma. However, elevated activation of iNKT cells by α-galactosylceramide and adoptive transfer of iNKT cells could upregulate surface maturation markers and proinflammatory cytokine secretion of LDCs from mouse models of asthma. Meanwhile, iNKT cells significantly influenced the function of LDCs, markedly enhancing Th2 responses in vivo and in vitro. In addition, iNKT cell can induce LDCs expression of CD206 and RELM-α, reflecting alternative activation of LDCs in a mouse model of asthma. α-Galactosylceramide treatment significantly enhanced expression of CD40L of lung iNKT cells from a mouse model of asthma, and the coculture experiment of LDCs with iNKT cells showed that the blockade of CD40L strongly suppressed surface maturation markers and proinflammatory cytokine production by LDCs. Our data suggest that iNKT cells can promote immunogenic maturation of LDCs to enhance Th2 responses in mouse models of asthma.
Collapse
Affiliation(s)
- Qing He
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Linlin Liu
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qiaoyu Yang
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ailing Wang
- Nursing Department, Wuhan University School of Health Sciences, Wuhan, China; and
| | - Shuo Chen
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruiyun Li
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Huang
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guqin Zhang
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuhong Ding
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongying Yu
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Suping Hu
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hanxiang Nie
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China;
| |
Collapse
|
37
|
Contribution of innate immune cells to pathogenesis of severe influenza virus infection. Clin Sci (Lond) 2017; 131:269-283. [PMID: 28108632 DOI: 10.1042/cs20160484] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/19/2016] [Accepted: 11/25/2016] [Indexed: 12/12/2022]
Abstract
Influenza A viruses (IAVs) cause respiratory illness of varying severity based on the virus strains, host predisposition and pre-existing immunity. Ultimately, outcome and recovery from infection rely on an effective immune response comprising both innate and adaptive components. The innate immune response provides the first line of defence and is crucial to the outcome of infection. Airway epithelial cells are the first cell type to encounter the virus in the lungs, providing antiviral and chemotactic molecules that shape the ensuing immune response by rapidly recruiting innate effector cells such as NK cells, monocytes and neutrophils. Each cell type has unique mechanisms to combat virus-infected cells and limit viral replication, however their actions may also lead to pathology. This review focuses how innate cells contribute to protection and pathology, and provides evidence for their involvement in immune pathology in IAV infections.
Collapse
|
38
|
Gao WX, Sun YQ, Shi J, Li CL, Fang SB, Wang D, Deng XQ, Wen W, Fu QL. Effects of mesenchymal stem cells from human induced pluripotent stem cells on differentiation, maturation, and function of dendritic cells. Stem Cell Res Ther 2017; 8:48. [PMID: 28253916 PMCID: PMC5333407 DOI: 10.1186/s13287-017-0499-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/11/2017] [Accepted: 02/09/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have potent immunomodulatory effects on multiple immune cells and have great potential in treating immune disorders. Induced pluripotent stem cells (iPSCs) serve as an unlimited and noninvasive source of MSCs, and iPSC-MSCs have been reported to have more advantages and exhibit immunomodulation on T lymphocytes and natural killer cells. However, the effects of iPSC-MSCs on dendritic cells (DCs) are unclear. The aim of this study is to investigate the effects of iPSC-MSCs on the differentiation, maturation, and function of DCs. METHODS Human monocyte-derived DCs were induced and cultured in the presence or absence of iPSC-MSCs. Flow cytometry was used to analyze the phenotype and functions of DCs, and enzyme-linked immunosorbent assay (ELISA) was used to study cytokine production. RESULTS In this study, we successfully induced MSCs from different clones of human iPSCs. iPSC-MSCs exhibited a higher proliferation rate with less cell senescence than BM-MSCs. iPSC-MSCs inhibited the differentiation of human monocyte-derived DCs by both producing interleukin (IL)-10 and direct cell contact. Furthermore, iPSC-MSCs did not affect immature DCs to become mature DCs, but modulated their functional properties by increasing their phagocytic ability and inhibiting their ability to stimulate proliferation of lymphocytes. More importantly, iPSC-MSCs induced the generation of IL-10-producing regulatory DCs in the process of maturation, which was mostly mediated by a cell-cell contact mechanism. CONCLUSIONS Our results indicate an important role for iPSC-MSCs in the modulation of DC differentiation and function, supporting the clinical application of iPSC-MSCs in DC-mediated immune diseases.
Collapse
Affiliation(s)
- Wen-Xiang Gao
- 0000 0001 2360 039Xgrid.12981.33Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong People’s Republic of China
| | - Yue-Qi Sun
- 0000 0001 2360 039Xgrid.12981.33Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong People’s Republic of China
| | - Jianbo Shi
- 0000 0001 2360 039Xgrid.12981.33Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong People’s Republic of China
| | - Cheng-Lin Li
- 0000 0001 2360 039Xgrid.12981.33Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong People’s Republic of China
- grid.412615.5Centre for Stem Cell Clinical Research and Application, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong People’s Republic of China
| | - Shu-Bin Fang
- 0000 0001 2360 039Xgrid.12981.33Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong People’s Republic of China
| | - Dan Wang
- 0000 0001 2360 039Xgrid.12981.33Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong People’s Republic of China
| | - Xue-Quan Deng
- 0000 0001 2360 039Xgrid.12981.33Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong People’s Republic of China
| | - Weiping Wen
- 0000 0001 2360 039Xgrid.12981.33Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong People’s Republic of China
| | - Qing-Ling Fu
- 0000 0001 2360 039Xgrid.12981.33Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong People’s Republic of China
- grid.412615.5Centre for Stem Cell Clinical Research and Application, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong People’s Republic of China
| |
Collapse
|
39
|
Zhang Y, Xu Y, Liu S, Guo X, Cen D, Xu J, Li H, Li K, Zeng C, Lu L, Zhou Y, Shen H, Cheng H, Zhang X, Ke Y. Scaffolding protein Gab1 regulates myeloid dendritic cell migration in allergic asthma. Cell Res 2016; 26:1226-1241. [PMID: 27811945 DOI: 10.1038/cr.2016.124] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 08/13/2016] [Accepted: 08/18/2016] [Indexed: 12/14/2022] Open
Abstract
Asthma is a common allergic disorder involving a complex interplay among multiple genetic and environmental factors. Recent studies identified genetic variants of human GAB1 as a novel asthma susceptibility factor. However, the functions of Gab1 in lung remain largely unexplored. In this study, we first observed an elevation of Gab1 level in peripheral blood mononuclear cells from asthmatic patients during acute exacerbation compared with convalescence. Mice with a selectively disrupted Gab1 in myeloid dendritic cells (mDCs) considerably attenuated allergic inflammation in experimental models of asthma. Further investigations revealed a prominent reduction in CCL19-mediated migration of Gab1-deficient mDCs to draining lymph nodes and subsequent impairment of Th2-driven adaptive activation. Mechanistically, Gab1 is an essential component of the CCL19/CCR7 chemokine axis that regulates mDC migration during asthmatic responses. Together, these findings provide the first evidence for the roles of Gab1 in lung, giving us deeper understanding of asthmatic pathogenesis.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Yun Xu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Shuwan Liu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xiaohong Guo
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Dong Cen
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Jiaqi Xu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Heyuan Li
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Kaijun Li
- Lishui Central Hospital, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, China
| | - Chunlai Zeng
- Lishui Central Hospital, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, China
| | - Linrong Lu
- The Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Yiting Zhou
- Department of Biochemistry and Molecular Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Huahao Shen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Hongqiang Cheng
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, China
| | - Xue Zhang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, China
| | - Yuehai Ke
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
40
|
Olivar R, Luque A, Cárdenas-Brito S, Naranjo-Gómez M, Blom AM, Borràs FE, Rodriguez de Córdoba S, Zipfel PF, Aran JM. The Complement Inhibitor Factor H Generates an Anti-Inflammatory and Tolerogenic State in Monocyte-Derived Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2016; 196:4274-90. [PMID: 27076676 DOI: 10.4049/jimmunol.1500455] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 02/27/2016] [Indexed: 12/14/2022]
Abstract
The activation of the complement system is a key initiating step in the protective innate immune-inflammatory response against injury, although it may also cause harm if left unchecked. The structurally related soluble complement inhibitors C4b-binding protein (C4BP) and factor H (FH) exert a tight regulation of the classical/lectin and alternative pathways of complement activation, respectively, attenuating the activity of the C3/C5 convertases and, consequently, avoiding serious damage to host tissues. We recently reported that the acute-phase C4BP isoform C4BP lacking the β-chain plays a pivotal role in the modulation of the adaptive immune responses. In this study, we demonstrate that FH acts in the early stages of monocyte to dendritic cell (DC) differentiation and is able to promote a distinctive tolerogenic and anti-inflammatory profile on monocyte-derived DCs (MoDCs) challenged by a proinflammatory stimulus. Accordingly, FH-treated and LPS-matured MoDCs are characterized by altered cytoarchitecture, resembling immature MoDCs, lower expression of the maturation marker CD83 and the costimulatory molecules CD40, CD80, and CD86, decreased production of key proinflammatory Th1-cytokines (IL-12, TNF-α, IFN-γ, IL-6, and IL-8), and preferential production of immunomodulatory mediators (IL-10 and TGF-β). Moreover, FH-treated MoDCs show low Ag uptake and, when challenged with LPS, display reduced CCR7 expression and chemotactic migration, impaired CD4(+) T cell alloproliferation, inhibition of IFN-γ secretion by the allostimulated T cells, and, conversely, induction of CD4(+)CD127(low/negative)CD25(high)Foxp3(+) regulatory T cells. Thus, this novel noncanonical role of FH as an immunological brake able to directly affect the function of MoDCs in an inflammatory environment may exhibit therapeutic potential in hypersensitivity, transplantation, and autoimmunity.
Collapse
Affiliation(s)
- Rut Olivar
- Human Molecular Genetics Group, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Ana Luque
- Human Molecular Genetics Group, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Sonia Cárdenas-Brito
- Human Molecular Genetics Group, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Mar Naranjo-Gómez
- Innovation in Vesicles and Cells for Application Therapy Group, Germans Trias i Pujol Health Sciences Research Institute, 08916 Badalona, Barcelona, Spain; Nephrology Service, Germans Trias i Pujol University Hospital, 08916 Badalona, Barcelona, Spain
| | - Anna M Blom
- Section of Medical Protein Chemistry, Department of Laboratory Medicine, Lund University, 20502 Malmö, Sweden
| | - Francesc E Borràs
- Innovation in Vesicles and Cells for Application Therapy Group, Germans Trias i Pujol Health Sciences Research Institute, 08916 Badalona, Barcelona, Spain; Nephrology Service, Germans Trias i Pujol University Hospital, 08916 Badalona, Barcelona, Spain
| | | | - Peter F Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Products Research and Infection Biology, 07745 Jena, Germany
| | - Josep M Aran
- Human Molecular Genetics Group, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, 08908 Barcelona, Spain;
| |
Collapse
|
41
|
Hoffmann F, Ender F, Schmudde I, Lewkowich IP, Köhl J, König P, Laumonnier Y. Origin, Localization, and Immunoregulatory Properties of Pulmonary Phagocytes in Allergic Asthma. Front Immunol 2016; 7:107. [PMID: 27047494 PMCID: PMC4803735 DOI: 10.3389/fimmu.2016.00107] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/08/2016] [Indexed: 01/21/2023] Open
Abstract
Allergic asthma is a chronic inflammatory disease of the airways that is driven by maladaptive T helper 2 (Th2) and Th17 immune responses against harmless, airborne substances. Pulmonary phagocytes represent the first line of defense in the lung where they constantly sense the local environment for potential threats. They comprise two distinct cell types, i.e., macrophages and dendritic cells (DC) that differ in their origins and functions. Alveolar macrophages quickly take up most of the inhaled allergens, yet do not deliver their cargo to naive T cells sampling in draining lymph nodes. In contrast, pulmonary DCs instruct CD4(+) T cells develop into Th2 and Th17 effectors, initiating the maladaptive immune responses toward harmless environmental substances observed in allergic individuals. Unraveling the mechanisms underlying this mistaken identity of harmless, airborne substances by innate immune cells is one of the great challenges in asthma research. The identification of different pulmonary DC subsets, their role in antigen uptake, migration to the draining lymph nodes, and their potential to instruct distinct T cell responses has set the stage to unravel this mystery. However, at this point, a detailed understanding of the spatiotemporal resolution of DC subset localization, allergen uptake, processing, autocrine and paracrine cellular crosstalk, and the humoral factors that define the activation status of DCs is still lacking. In addition to DCs, at least two distinct macrophage populations have been identified in the lung that are either located in the airway/alveolar lumen or in the interstitium. Recent data suggest that such populations can exert either pro- or anti-inflammatory functions. Similar to the DC subsets, detailed insights into the individual roles of alveolar and interstitial macrophages during the different phases of asthma development are still missing. Here, we will provide an update on the current understanding of the origin, localization, and function of the diverse pulmonary antigen-presenting cell subsets, in particular with regard to the development and regulation of allergic asthma. While most data are from mouse models of experimental asthma, we have also included available human data to judge the translational value of the findings obtained in experimental asthma models.
Collapse
Affiliation(s)
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Inken Schmudde
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Ian P. Lewkowich
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Giessen, Germany
| | - Peter König
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Giessen, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| |
Collapse
|
42
|
T Follicular Helper Cell Plasticity Shapes Pathogenic T Helper 2 Cell-Mediated Immunity to Inhaled House Dust Mite. Immunity 2016; 44:259-73. [PMID: 26825674 DOI: 10.1016/j.immuni.2015.11.017] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 10/26/2015] [Accepted: 11/19/2015] [Indexed: 11/22/2022]
Abstract
Exposure to environmental antigens, such as house dust mite (HDM), often leads to T helper 2 (Th2) cell-driven allergic responses. However, the mechanisms underlying the development of these responses are incompletely understood. We found that the initial exposure to HDM did not lead to Th2 cell development but instead promoted the formation of interleukin-4 (IL-4)-committed T follicular helper (Tfh) cells. Following challenge exposure to HDM, Tfh cells differentiated into IL-4 and IL-13 double-producing Th2 cells that accumulated in the lung and recruited eosinophils. B cells were required to expand IL-4-committed Tfh cells during the sensitization phase, but did not directly contribute to disease. Impairment of Tfh cell responses during the sensitization phase or Tfh cell depletion prevented Th2 cell-mediated responses following challenge. Thus, our data demonstrate that Tfh cells are precursors of HDM-specific Th2 cells and reveal an unexpected role of B cells and Tfh cells in the pathogenesis of allergic asthma.
Collapse
|
43
|
Allergic Inflammation in Aspergillus fumigatus-Induced Fungal Asthma. Curr Allergy Asthma Rep 2015; 15:59. [PMID: 26288940 DOI: 10.1007/s11882-015-0561-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although fungi are pervasive in many environments, few cause disease in humans. Of these, Aspergillus fumigatus is particularly well suited to be a pathogen of the human lung. Its physical and biological characteristics combine to provide an organism that can cause tremendous morbidity and high mortality if left unchecked. Luckily, that is rarely the case. However, repeated exposure to inhaled A. fumigatus spores often results in an immune response that carries significant immunopathology, exacerbating asthma and changing the structure of the lung with chronic impacts to pulmonary function. This review focuses on the current understanding of the mechanisms that are associated with fungal exposure, sensitization, and infection in asthmatics, as well as the function of various inflammatory cells associated with severe asthma with fungal sensitization.
Collapse
|
44
|
Scanlon ST, McKenzie ANJ. The messenger between worlds: the regulation of innate and adaptive type-2 immunity by innate lymphoid cells. Clin Exp Allergy 2015; 45:9-20. [PMID: 25423949 DOI: 10.1111/cea.12464] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Although type-2 immune responses evolved primarily to defend against extracellular helminths, in part through the co-opting of tissue repair and remodeling mechanisms, they are often inappropriately directed towards relatively innocuous allergens resulting in conditions including asthma, allergic rhinitis, food allergy, and atopic dermatitis. The recent discovery of group 2 innate lymphoid cells (ILC2) has increased our understanding of the initiation of these responses and the roles played by CD4(+) T helper (Th) 2 cells in their modulation. This review focuses on the important messenger role of ILC2 in translating epithelial-derived alarmins into downstream adaptive type-2 responses via dendritic cells and T cells, with special emphasis on their roles in allergic disease.
Collapse
Affiliation(s)
- S T Scanlon
- Protein & Nucleic Acid Chemistry (PNAC) Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | | |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW The purpose of this study is to summarize recent studies of the lower respiratory microbiome in asthma, the role of innate immunity in asthma and strategies to understand complex microbiome-immune interactions in asthma. RECENT FINDINGS Recent evidence indicates that the composition of lower respiratory microbiota in asthmatic individuals, across a spectrum of disease severity, is altered compared with healthy individuals. Attributes of this altered airway microbiome have been linked to clinical and inflammatory features of asthma. The importance of innate immune cells and mucosal defense systems in asthma is increasingly appreciated and may be dysregulated in the disease. SUMMARY Interactions between the respiratory microbiome and innate mucosal immunity in asthma are complex and a challenge to dissect. Multiple avenues of investigation, leveraging a variety of methodologies, will need to be pursued to understand functional relationships to clinical and inflammatory phenotypes seen in asthma.
Collapse
|
46
|
Kopf M, Schneider C, Nobs SP. The development and function of lung-resident macrophages and dendritic cells. Nat Immunol 2015; 16:36-44. [PMID: 25521683 DOI: 10.1038/ni.3052] [Citation(s) in RCA: 394] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/10/2014] [Indexed: 12/12/2022]
Abstract
Gas exchange is the vital function of the lungs. It occurs in the alveoli, where oxygen and carbon dioxide diffuse across the alveolar epithelium and the capillary endothelium surrounding the alveoli, separated only by a fused basement membrane 0.2-0.5 μm in thickness. This tenuous barrier is exposed to dangerous or innocuous particles, toxins, allergens and infectious agents inhaled with the air or carried in the blood. The lung immune system has evolved to ward off pathogens and restrain inflammation-mediated damage to maintain gas exchange. Lung-resident macrophages and dendritic cells are located in close proximity to the epithelial surface of the respiratory system and the capillaries to sample and examine the air-borne and blood-borne material. In communication with alveolar epithelial cells, they set the threshold and the quality of the immune response.
Collapse
Affiliation(s)
- Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Christoph Schneider
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Samuel P Nobs
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| |
Collapse
|
47
|
First genomic analysis of dendritic cells from lung and draining lymph nodes in murine asthma. Int J Genomics 2015; 2015:638032. [PMID: 25811019 PMCID: PMC4355561 DOI: 10.1155/2015/638032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 02/09/2015] [Accepted: 02/09/2015] [Indexed: 01/05/2023] Open
Abstract
Asthma is the consequence of allergic inflammation in the lung compartments and lung-draining lymph nodes. Dendritic cells initiate and promote T cell response and drive it to immunity or allergy. However, their modes of action during asthma are poorly understood. In this study, an allergic inflammation with ovalbumin was induced in 38 mice versus 42 control animals. After ovalbumin aerosol challenge, conventional dendritic cells (CD11c/MHCII/CD8) were isolated from the lungs and the draining lymph nodes by means of magnetic cell sorting followed by fluorescence-activated cell sorting. A comparative transcriptional analysis was performed using gene arrays. In general, many transcripts are up- and downregulated in the CD8− dendritic cells of the allergic inflamed lung tissue, whereas few genes are regulated in CD8+ dendritic cells. The dendritic cells of the lymph nodes also showed minor transcriptional changes. The data support the relevance of the CD8− conventional dendritic cells but do not exclude distinct functions of the small population of CD8+ dendritic cells, such as cross presentation of external antigen. So far, this is the first approach performing gene arrays in dendritic cells obtained from lung tissue and lung-draining lymph nodes of asthmatic-like mice.
Collapse
|
48
|
den Haan JM, Arens R, van Zelm MC. The activation of the adaptive immune system: Cross-talk between antigen-presenting cells, T cells and B cells. Immunol Lett 2014; 162:103-12. [DOI: 10.1016/j.imlet.2014.10.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
49
|
Epithelial barrier function: at the front line of asthma immunology and allergic airway inflammation. J Allergy Clin Immunol 2014; 134:509-20. [PMID: 25085341 DOI: 10.1016/j.jaci.2014.05.049] [Citation(s) in RCA: 333] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/30/2014] [Accepted: 05/30/2014] [Indexed: 02/08/2023]
Abstract
Airway epithelial cells form a barrier to the outside world and are at the front line of mucosal immunity. Epithelial apical junctional complexes are multiprotein subunits that promote cell-cell adhesion and barrier integrity. Recent studies in the skin and gastrointestinal tract suggest that disruption of cell-cell junctions is required to initiate epithelial immune responses, but how this applies to mucosal immunity in the lung is not clear. Increasing evidence indicates that defective epithelial barrier function is a feature of airway inflammation in asthmatic patients. One challenge in this area is that barrier function and junctional integrity are difficult to study in the intact lung, but innovative approaches should provide new knowledge in this area in the near future. In this article we review the structure and function of epithelial apical junctional complexes, emphasizing how regulation of the epithelial barrier affects innate and adaptive immunity. We discuss why defective epithelial barrier function might be linked to TH2 polarization in asthmatic patients and propose a rheostat model of barrier dysfunction that implicates the size of inhaled allergen particles as an important factor influencing adaptive immunity.
Collapse
|
50
|
Key mediators in the immunopathogenesis of allergic asthma. Int Immunopharmacol 2014; 23:316-29. [PMID: 24933589 DOI: 10.1016/j.intimp.2014.05.034] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 12/20/2022]
Abstract
Asthma is described as a chronic inflammatory disorder of the conducting airways. It is characterized by reversible airway obstruction, eosinophil and Th2 infiltration, airway hyper-responsiveness and airway remodeling. Our findings to date have largely been dependent on work done using animal models, which have been instrumental in broadening our understanding of the mechanism of the disease. However, using animals to model a uniquely human disease is not without its drawbacks. This review aims to examine some of the key mediators and cells of allergic asthma learned from animal models and shed some light on emerging mediators in the pathogenesis allergic airway inflammation in acute and chronic asthma.
Collapse
|