151
|
Kariyawasam HH, James LK, Gane SB. Dupilumab: Clinical Efficacy of Blocking IL-4/IL-13 Signalling in Chronic Rhinosinusitis with Nasal Polyps. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1757-1769. [PMID: 32440101 PMCID: PMC7217316 DOI: 10.2147/dddt.s243053] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 04/29/2020] [Indexed: 12/17/2022]
Abstract
In September 2019, The Lancet published details of two large Phase III double-blind placebo-controlled studies (LIBERTY NP SINUS-24 and LIBERTY NP SINUS-52) confirming the clinical efficacy of the biologic dupilumab in simultaneously blocking both IL-4/IL-13 signalling in chronic rhinosinusitis with nasal polyps (CRSwNP). The studies demonstrated that dupilumab (Dupixent®, Sanofi and Regeneron) 300mg subcutaneously administered was clinically effective when added for patients with moderate to severe CRSwNP already maintained on the standard intranasal steroid mometasone furoate. Duration of treatment ranged from injections either 2 weekly for 24 weeks (SINUS-24) or every 2 weeks for 52 weeks or finally every 2 weeks for 24 weeks stepping down thereafter to every 4 weeks for a further 28 weeks (SINUS-52). Rapid improvements in all important parameters of disease burden were seen with such improvement maintained even where the frequency of injections was decreased. In patients with co-existent asthma, lung function and asthma control scores improved. This is consistent with the one airway hypothesis of shared T2 inflammatory programmes driving both disease syndromes. The studies formed the basis for FDA registration and clinical launch in the US, and EMA approval in Europe. Dupilumab presents a significant new treatment option in an area of urgent unmet therapeutic need in CRSwNP. Should dupilumab prove to be as effective in the real-life clinical environment as it has been in the studies, then a paradigm shift from sinonasal surgery to medical treatment of CRSwNP may need to occur in the ENT community. Questions in relation to best patient selection, combined upper and lower airway therapeutic pathways, long-term safety along with health economics and cost constraints ought now to be addressed.
Collapse
Affiliation(s)
- Harsha H Kariyawasam
- Department of Specialist Allergy and Clinical Immunology, Royal National ENT Hospital, University College London Hospitals NHS Foundation Trust, London, UK.,Department of Rhinology, Royal National ENT Hospital, University College London Hospitals NHS Foundation Trust, London, UK.,UCL Ear Institute , University College London, London, UK
| | - Louisa K James
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Simon B Gane
- Department of Rhinology, Royal National ENT Hospital, University College London Hospitals NHS Foundation Trust, London, UK.,UCL Ear Institute , University College London, London, UK
| |
Collapse
|
152
|
Norlander AE, Peebles RS. Innate Type 2 Responses to Respiratory Syncytial Virus Infection. Viruses 2020; 12:E521. [PMID: 32397226 PMCID: PMC7290766 DOI: 10.3390/v12050521] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a common and contagious virus that results in acute respiratory tract infections in infants. In many cases, the symptoms of RSV remain mild, however, a subset of individuals develop severe RSV-associated bronchiolitis. As such, RSV is the chief cause of infant hospitalization within the United States. Typically, the immune response to RSV is a type 1 response that involves both the innate and adaptive immune systems. However, type 2 cytokines may also be produced as a result of infection of RSV and there is increasing evidence that children who develop severe RSV-associated bronchiolitis are at a greater risk of developing asthma later in life. This review summarizes the contribution of a newly described cell type, group 2 innate lymphoid cells (ILC2), and epithelial-derived alarmin proteins that activate ILC2, including IL-33, IL-25, thymic stromal lymphopoietin (TSLP), and high mobility group box 1 (HMGB1). ILC2 activation leads to the production of type 2 cytokines and the induction of a type 2 response during RSV infection. Intervening in this innate type 2 inflammatory pathway may have therapeutic implications for severe RSV-induced disease.
Collapse
Affiliation(s)
| | - R. Stokes Peebles
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-2650, USA;
| |
Collapse
|
153
|
Regulatory T cells are a double-edged sword in pulmonary fibrosis. Int Immunopharmacol 2020; 84:106443. [PMID: 32334385 DOI: 10.1016/j.intimp.2020.106443] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/23/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023]
Abstract
Pulmonary fibrosis (PF) is a chronic progressive interstitial lung disease. The pathogenesis of PF has not been clearly elucidated, and there is no obvious effective treatment to arrest the progression of PF to date. A long-term chronic inflammatory response and inappropriate repair process after lung injury are important causes and pathological processes of PF. As an influential type of the body's immune cells, regulatory T cells (Tregs) play an irreplaceable role in inhibiting the inflammatory response and promoting the repair of lung tissue. However, the exact roles of Tregs in the process of PF have not been clearly established, and the available literature concerning the roles of Tregs in PF are contradictory. First, Tregs can advance the progression of pulmonary fibrosis by secreting platelet-derived growth factor (PDGF), transforming growth factor-β (TGF-β) and other related factors, promoting epithelial-mesenchymal transition (EMT) and affecting the Th1 and Th2 balance, etc. Second, Tregs can inhibit PF by promoting the repair of epithelial cell damage, inhibiting the accumulation of fibroblasts, and strongly inhibiting the production and function of other related pro-inflammatory factors and pro-inflammatory cells. Accordingly, in this review, we focus on the multiple roles of Tregs in different models and different pulmonary fibrosis phases, thereby providing theoretical support for a better understanding of the multiple roles of these cells in PF and a theoretical basis for identifying targets for PF therapy.
Collapse
|
154
|
Prediction of response to biological treatment with monoclonal antibodies in severe asthma. Biochem Pharmacol 2020; 179:113978. [PMID: 32305434 DOI: 10.1016/j.bcp.2020.113978] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/17/2020] [Indexed: 12/11/2022]
Abstract
In recent years, major developments have occurred in severe asthma management. Different asthma phenotypes and subgroups have been identified and new treatment options have become available. A total of five monoclonal antibodies are currently approved in severe asthma treatment: omalizumab, mepolizumab, reslizumab, benralizumab and dupilumab. These drugs have been shown to reduce exacerbations and to have an oral corticosteroid-sparing effect in many severe asthma patients. However, biological treatment is not successful in all patients and should be discontinued in non-responsive patients. Treating the right patient with the right biologic, and therefore biologic response prediction, has become a major point of interest in severe asthma management. A variety of response outcomes is utilized in the different clinical trials, as well as a huge range of potential predicting factors. Also, regarding the timing of the response evaluation, there are considerable differences between studies. This review summarizes the results from studies on predicting responses and responders to biological treatment in severe asthma, taking into account clinical, functional and inflammatory parameters assessed prior to the start of treatment as well as following a few months of therapy. In addition, future perspectives are discussed, highlighting the need for more research to improve patient identification and treatment responses in the field of biological treatment in severe asthma.
Collapse
|
155
|
Lewis BW, Vo T, Choudhary I, Kidder A, Bathula C, Ehre C, Wakamatsu N, Patial S, Saini Y. Ablation of IL-33 Suppresses Th2 Responses but Is Accompanied by Sustained Mucus Obstruction in the Scnn1b Transgenic Mouse Model. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:1650-1660. [PMID: 32060135 PMCID: PMC7714586 DOI: 10.4049/jimmunol.1900234] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 01/14/2020] [Indexed: 12/14/2022]
Abstract
Cystic fibrosis is characterized by dehydration of the airway surface liquid layer with persistent mucus obstruction. Th2 immune responses are often manifested as increased mucous cell density (mucous cell metaplasia) associated with mucus obstruction. IL-33 is a known inducer of Th2 immune responses, but its roles in mucus obstruction and related phenotypes in a cystic fibrosis-like lung disease model (i.e., Scnn1b-Tg-positive [Tg+]) mouse, remain unclear. Accordingly, IL-33 knockout (IL-33KO) Tg+ mice were examined and compared with IL-33 heterozygous (IL-33HET) Tg+ mice. As compared with IL-33HET/Tg+ mice, IL-33KO/Tg+ mice had complete absence of bronchoalveolar lavage fluid eosinophilia, accompanied with significant reduction in bronchoalveolar lavage fluid concentration of IL-5, a cytokine associated with eosinophil differentiation and recruitment, and IL-4, a major Th2 cytokine. As compared with IL-33HET/Tg+ mice, IL-33KO/Tg+ mice had significantly reduced levels of Th2-associated gene signatures (Slc26a4, Clca1, Retnla, and Chi3l4), along with complete loss of intracellular mucopolysaccharide staining in the airway epithelium. As compared with IL-33HET/Tg+ mice, although the IL-33KO/Tg+ mice had significantly reduced levels of MUC5AC protein expression, they showed no reduction in the degree of mucus obstruction, MUC5B protein expression, bacterial burden, and neonatal mortality. Interestingly, the histological features, including subepithelial airway inflammation and alveolar space enlargement, were somewhat exaggerated in IL-33KO/Tg+ mice compared with IL-33HET/Tg+ mice. Taken together, our data indicate that although IL-33 modulates Th2 inflammatory responses and MUC5AC protein production, mucus obstruction is not dependent on IL-33.
Collapse
Affiliation(s)
- Brandon W Lewis
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA 70803
| | - Thao Vo
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA 70803
| | - Ishita Choudhary
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA 70803
| | - Allison Kidder
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA 70803
| | - Chandra Bathula
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA 70803
| | - Camille Ehre
- Marsico Lung Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and
| | - Nobuko Wakamatsu
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA 70803
| | - Sonika Patial
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA 70803
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA 70803;
| |
Collapse
|
156
|
Davies ER, Perotin JM, Kelly JFC, Djukanovic R, Davies DE, Haitchi HM. Involvement of the epidermal growth factor receptor in IL-13-mediated corticosteroid-resistant airway inflammation. Clin Exp Allergy 2020; 50:672-686. [PMID: 32096290 PMCID: PMC7317751 DOI: 10.1111/cea.13591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/12/2020] [Accepted: 02/19/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Effective treatment for severe asthma is a significant unmet need. While eosinophilic inflammation caused by type 2 cytokines is responsive to corticosteroid and biologic therapies, many severe asthmatics exhibit corticosteroid-unresponsive mixed granulocytic inflammation. OBJECTIVE Here, we tested the hypothesis that the pro-allergic cytokine, IL-13, can drive both corticosteroid-sensitive and corticosteroid-resistant responses. RESULTS By integration of in vivo and in vitro models of IL-13-driven inflammation, we identify a role for the epidermal growth factor receptor (EGFR/ERBB1) as a mediator of corticosteroid-unresponsive inflammation and bronchial hyperresponsiveness driven by IL-13. Topological data analysis using human epithelial transcriptomic data from the U-BIOPRED cohort identified severe asthma groups with features consistent with the presence of IL-13 and EGFR/ERBB activation, with involvement of distinct EGFR ligands. Our data suggest that IL-13 may play a dual role in severe asthma: on the one hand driving pathologic corticosteroid-refractory mixed granulocytic inflammation, but on the other hand underpinning beneficial epithelial repair responses, which may confound responses in clinical trials. CONCLUSION AND CLINICAL RELEVANCE Detailed dissection of those molecular pathways that are downstream of IL-13 and utilize the ERBB receptor and ligand family to drive corticosteroid-refractory inflammation should enhance the development of new treatments that target this sub-phenotype(s) of severe asthma, where there is an unmet need.
Collapse
Affiliation(s)
- Elizabeth R Davies
- Brooke Laboratories, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Jeanne-Marie Perotin
- Brooke Laboratories, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,National Institute for Health Research (NIHR) Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Joanne F C Kelly
- Brooke Laboratories, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Ratko Djukanovic
- Brooke Laboratories, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,National Institute for Health Research (NIHR) Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Donna E Davies
- Brooke Laboratories, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,National Institute for Health Research (NIHR) Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Hans Michael Haitchi
- Brooke Laboratories, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,National Institute for Health Research (NIHR) Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Institute for Life Sciences, University of Southampton, Southampton, UK
| | | |
Collapse
|
157
|
Wang X, Xu C, Ji J, Cai Y, Shu Y, Chao Y, Wu X, Zou C, Wu X, Tang L. IL-4/IL-13 upregulates Sonic hedgehog expression to induce allergic airway epithelial remodeling. Am J Physiol Lung Cell Mol Physiol 2020; 318:L888-L899. [PMID: 32130032 DOI: 10.1152/ajplung.00186.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We have previously demonstrated that upregulation of Sonic hedgehog (SHH) expression in allergic airway epithelia essentially contributes to the goblet cell metaplasia and mucous hypersecretion. However, the mechanism underlying the upregulation of SHH expression remains completely unknown. In cultured human airway epithelial cells, IL-4/IL-13 but not IL-5 robustly induces the mRNA and protein expression of SHH and in turn activates SHH signaling by promoting the JAK/STAT6-controlling transcription of SHH gene. Moreover, intratracheal instillation of IL-4 and/or IL-13 robustly activates STAT6 and concomitantly upregulates SHH expression in mouse airway epithelia, whereas, in Club cell 10-kDa protein (CC10)-positive airway epithelial cells of children with asthma, activated STAT6 closely correlates with the increased expression of SHH and high activity of SHH signaling. Finally, intratracheal inhibition of STAT6 by AS-1517499 significantly diminished the allergen-induced upregulation of SHH expression, goblet cell phenotypes, and airway hyperresponsiveness, in an ovalbumin- or house dust mite-induced mouse model with allergic airway inflammation,. Together, upregulation of SHH expression by IL-4/IL-13-induced JAK/STAT6 signaling contributes to allergic airway epithelial remodeling, and this study thus provides insight into how morphogen signaling is coordinated with Th2 cytokine pathways to regulate tissue remodeling in chronic airway diseases.
Collapse
Affiliation(s)
- Xiangzhi Wang
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Chengyun Xu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of CFDA for Respiratory Drug Research, Zhejiang University School of Medicine, Hangzhou, China
| | - Junyan Ji
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Yuqing Cai
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Yingying Shu
- National Clinical Research Center for Child Health, Hangzhou, China.,Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunqi Chao
- National Clinical Research Center for Child Health, Hangzhou, China.,Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiling Wu
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| | - Chaochun Zou
- National Clinical Research Center for Child Health, Hangzhou, China.,Department of Endocrinology, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ximei Wu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of CFDA for Respiratory Drug Research, Zhejiang University School of Medicine, Hangzhou, China
| | - Lanfang Tang
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
158
|
Sánchez-Ortiz M, Cruz MJ, Sánchez-Díez S, Villar A, Ojanguren I, Muñoz X. Immunomodulatory effect of pigeon serum in an acute and chronic murine model of bird fanciers lung. ENVIRONMENTAL RESEARCH 2020; 182:108981. [PMID: 31830693 DOI: 10.1016/j.envres.2019.108981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/29/2019] [Accepted: 11/29/2019] [Indexed: 06/10/2023]
Abstract
INTRODUCTION Since the immunopathological mechanisms of bird fancier's lung (BFL) are not well known, we created two models of the disease (acute and chronic BFL) to study and compare the pathways involved in its immunopathogenesis. MATERIALS AND METHODS C57BL/6 mice were used. Two intraperitoneal injections of 100 μL of commercial pigeon serum (PS) or saline (SAL) were administered with an interval of 48 h in between. Subsequently, intranasal instillations of 40 μL of PS or SAL were performed three days a week, for three weeks in the acute model (AC/PS) and for twelve weeks in the chronic model (CR/PS). Total lung capacity (TLC) was assessed. Pulmonary inflammation was evaluated in bronchoalveolar lavage (BAL), and total serum immunoglobulin (Ig) G was measured in serum samples 24 h, 7 days and 14 days after the last exposure. Histological studies of lungs were assessed. RESULTS A drop in TLC was observed in treated mice. This decrease was more marked in the CR/PS group (p < 0.001). Neutrophil and lymphocyte counts increased in both AC/PS and CR/PS groups (p < 0.01). The extent of airway inflammation was also examined in the histological analysis of the lungs, which showed predominant perivascular and peribronchiolar inflammation, with centrilobular oedema and subpleural inflammation in the AC/PS group. In the CR/PS group, the changes were greater, with increased levels of IL-5, IL-17F, IL-13 and IL-10 and decreased levels of IL-2. CONCLUSIONS Bronchial inflammation is present in acute and chronic models of HP following exposure to PS. Our results support the role of neutrophils and IL-17 in the development of the disease and an evolution towards a Th-2 immune response in chronic HP. These models may serve as a tool for future studies of the pathogenesis of HP.
Collapse
Affiliation(s)
- M Sánchez-Ortiz
- Pulmonology Department. Hospital Universitari Vall d'Hebron, Barcelona, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Spain
| | - M J Cruz
- Pulmonology Department. Hospital Universitari Vall d'Hebron, Barcelona, Spain; CIBER Respiratory Diseases (Ciberes), Spain.
| | - S Sánchez-Díez
- Pulmonology Department. Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - A Villar
- Pulmonology Department. Hospital Universitari Vall d'Hebron, Barcelona, Spain; CIBER Respiratory Diseases (Ciberes), Spain
| | - I Ojanguren
- Pulmonology Department. Hospital Universitari Vall d'Hebron, Barcelona, Spain; CIBER Respiratory Diseases (Ciberes), Spain
| | - X Muñoz
- Pulmonology Department. Hospital Universitari Vall d'Hebron, Barcelona, Spain; CIBER Respiratory Diseases (Ciberes), Spain; Department of Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Spain
| |
Collapse
|
159
|
Unique IL-13Rα2/STAT3 mediated IL-13 regulation detected in lung conventional dendritic cells, 24 h post viral vector vaccination. Sci Rep 2020; 10:1017. [PMID: 31974500 PMCID: PMC6978450 DOI: 10.1038/s41598-020-57815-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/30/2019] [Indexed: 12/19/2022] Open
Abstract
This study demonstrates that 24 h following viral vector-based vaccination IL-13Rα2 functions as a master sensor on conventional dendritic cells (cDCs), abetted by high protein stability coupled with minimal mRNA expression, to rapidly regulate DC mediated IL-13 responses at the lung mucosae, unlike IL-13Rα1. Under low IL-13, IL-13Rα2 performs as a primary signalling receptor, whilst under high IL-13, acts to sequester IL-13 to maintain homeostasis, both in a STAT3-dependent manner. Likewise, we show that viral vector-derived IL-13 levels at the vaccination site can induce differential STAT3/STAT6 paradigms in lung cDC, that can get regulated collaboratively or independently by TGF-β1 and IFN-γ. Specifically, low IL-13 responses associated with recombinant Fowlpox virus (rFPV) is regulated by early IL-13Rα2, correlated with STAT3/TGF-β1 expression. Whilst, high IL-13 responses, associated with recombinant Modified Vaccinia Ankara (rMVA) is regulated in an IL-13Rα1/STAT6 dependent manner associated with IFN-γR expression bias. Different viral vaccine vectors have previously been shown to induce unique adaptive immune outcomes. Taken together current observations suggest that IL-13Rα2-driven STAT3/STAT6 equilibrium at the cDC level may play an important role in governing the efficacy of vector-based vaccines. These new insights have high potential to be exploited to improve recombinant viral vector-based vaccine design, according to the pathogen of interest and/or therapies against IL-13 associated disease conditions.
Collapse
|
160
|
Glisinski KM, Schlobohm AJ, Paramore SV, Birukova A, Moseley MA, Foster MW, Barkauskas CE. Interleukin-13 disrupts type 2 pneumocyte stem cell activity. JCI Insight 2020; 5:131232. [PMID: 31941839 DOI: 10.1172/jci.insight.131232] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/04/2019] [Indexed: 12/28/2022] Open
Abstract
The T helper 2 (Th2) inflammatory cytokine interleukin-13 (IL-13) has been associated with both obstructive and fibrotic lung diseases; however, its specific effect on the epithelial stem cells in the gas exchange compartment of the lung (alveolar space) has not been explored. Here, we used in vivo lung models of homeostasis and repair, ex vivo organoid platforms, and potentially novel quantitative proteomic techniques to show that IL-13 disrupts the self-renewal and differentiation of both murine and human type 2 alveolar epithelial cells (AEC2s). Significantly, we find that IL-13 promotes ectopic expression of markers typically associated with bronchiolar airway cells and commonly seen in the alveolar region of lung tissue from patients with idiopathic pulmonary fibrosis. Furthermore, we identify a number of proteins that are differentially secreted by AEC2s in response to IL-13 and may provide biomarkers to identify subsets of patients with pulmonary disease driven by "Th2-high" biology.
Collapse
Affiliation(s)
- Kristen M Glisinski
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
| | - Adam J Schlobohm
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
| | - Sarah V Paramore
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
| | - Anastasiya Birukova
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and
| | - M Arthur Moseley
- Duke Proteomics and Metabolomics Shared Resource, Duke University Medical Center, Durham, North Carolina, USA
| | - Matthew W Foster
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, and.,Duke Proteomics and Metabolomics Shared Resource, Duke University Medical Center, Durham, North Carolina, USA
| | | |
Collapse
|
161
|
Licari A, Castagnoli R, Tondina E, Testa G, Parisi GF, Marseglia A, Brambilla I, Marseglia GL. Novel Biologics for the Treatment of Pediatric Severe Asthma. CURRENT RESPIRATORY MEDICINE REVIEWS 2020; 15:195-204. [DOI: 10.2174/1573398x15666190521111816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/25/2019] [Accepted: 04/10/2019] [Indexed: 11/22/2022]
Abstract
Estimated to represent less than 5% of all asthmatic patients, children with severe asthma experience troublesome persistent symptoms, life-threatening attacks and side effects by oral corticosteroid treatment, that significantly impact on the quality of life and on economic costs. An accurate understanding of the mechanisms of the disease has been crucial for the discovery and development of biological therapies, for which children with severe asthma are candidates. The aim of this review is to discuss the use of approved biologics for severe asthma, providing updated evidence of novel targeted therapies in the pediatric age range.
Collapse
Affiliation(s)
- Amelia Licari
- S.C. Pediatria, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Enrico Tondina
- Department of Pediatrics, University of Pavia, Pavia, Italy
| | - Giorgia Testa
- S.C. Pediatria, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giuseppe Fabio Parisi
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | | - Ilaria Brambilla
- S.C. Pediatria, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | |
Collapse
|
162
|
Wang Z, Ji N, Chen Z, Sun Z, Wu C, Yu W, Hu F, Huang M, Zhang M. MiR-1165-3p Suppresses Th2 Differentiation via Targeting IL-13 and PPM1A in a Mouse Model of Allergic Airway Inflammation. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2020; 12:859-876. [PMID: 32638565 PMCID: PMC7346992 DOI: 10.4168/aair.2020.12.5.859] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 03/06/2020] [Accepted: 03/13/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE CD4⁺T cells are essential in the pathogenesis of allergic asthma. We have previously demonstrated that microRNA-1165-3p (miR-1165-3p) was significantly reduced in T-helper type (Th) 2 cells and that miR-1165-3p was a surrogate marker for atopic asthma. Little is known about the mechanisms of miR-1165-3p in the regulation of Th2-dominated allergic inflammation. We aimed to investigate the associations between Th2 differentiation and miR-1165b-3p in asthma as well as the possible mechanisms. METHODS CD4⁺ naïve T cells were differentiated into Th1 or Th2 cells in vitro. MiR-1165-3p was up-regulated or down-regulated using lentiviral systems during Th1/Th2 differentiation. In vivo, the lentiviral particles with the miR-1165-3p enhancer were administered by tail vein injection on the first day of a house dust mite -induced allergic airway inflammation model. Allergic inflammation and Th1/Th2 differentiation were routinely monitored. To investigate the potential targets of miR-1165-3p, biotin-microRNA pull-down products were sequenced, and the candidates were further verified with a dual-luciferase reporter assay. The roles of a target protein phosphatase, Mg2+/Mn2+-dependent 1A (PPM1A), in Th2 cell differentiation and allergic asthma were further explored. Plasma PPM1A was determined by ELISA in 18 subjects with asthma and 20 controls. RESULTS The lentivirus encoding miR-1165-3p suppressed Th2-cell differentiation in vitro. In contrast, miR-1165-3p silencing promoted Th2-cell development. In the HDM-induced model of allergic airway inflammation, miR-1165-3p up-regulation was accompanied by reduced airway hyper-responsiveness, serum immunoglobulin E, airway inflammation and Th2-cell polarization. IL-13 and PPM1A were the direct targets of miR-1165-3p. The expression of IL-13 or PPM1A was inversely correlated with that of miR-1165-3p. PPM1A regulated the signal transducer and activator of transcription and AKT signaling pathways during Th2 differentiation. Moreover, plasma PPM1A was significantly increased in asthmatic patients. CONCLUSIONS MiR-1165-3p negatively may regulate Th2-cell differentiation by targeting IL-13 and PPM1A in allergic airway inflammation.
Collapse
Affiliation(s)
- Zhengxia Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ningfei Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhongqi Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhixiao Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chaojie Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenqing Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Infectious Disease, Taizhou People's Hospital, Taizhou, China
| | - Fan Hu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mao Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Mingshun Zhang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Immunology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
163
|
Beckert H, Meyer-Martin H, Buhl R, Taube C, Reuter S. Single and Synergistic Effects of Type 2 Cytokines on Eosinophils and Asthma Hallmarks. THE JOURNAL OF IMMUNOLOGY 2019; 204:550-558. [PMID: 31862712 DOI: 10.4049/jimmunol.1901116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/19/2019] [Indexed: 12/15/2022]
Abstract
The type 2 cytokines IL-5, IL-13, and IL-4 play an important role in the induction and progression of asthma. According to the Global Initiative for Asthma guidelines, blood eosinophil numbers are one marker that helps to guide treatment decisions in patients suffering from severe forms of asthma. Effects of type 2 cytokines were analyzed, alone or in combination, on eosinophils in blood and other compartments and on the development of asthma symptoms. C57BL/6 mice received a single intranasal application of equimolar amounts of IL-5, IL-13, and IL-4, alone or in combination. Numbers, activation state, and migratory behavior of eosinophils in bone marrow (BM), blood, lung, and bronchoalveolar lavage as well as airway hyperresponsiveness and goblet cell metaplasia were evaluated. Only IL-13 was associated with airway eosinophilia, development of airway hyperresponsiveness, and goblet cell metaplasia, without any synergistic effects. IL-5 increased the number of eosinophils in BM and lung tissue but failed to affect structural changes. IL-4 had similar, but weaker, effects to IL-13. Cytokine combinations synergistically affected eosinophils but failed to enhance IL-13-driven effects on lung function or goblet cell metaplasia. IL-5 and IL-13 markedly increased eosinophil numbers locally in lung and airways and distally in blood and BM, whereas IL-5 and IL-4 only increased eosinophils in lung and BM. IL-13 together with IL-4 failed to demonstrate any synergistic effect. These insights into single and combined effects of type 2 cytokines on disease-driving mechanisms could improve understanding of the impact and effectiveness of new therapies in asthma.
Collapse
Affiliation(s)
- Hendrik Beckert
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, 45239 Essen, Germany; and
| | - Helen Meyer-Martin
- Department of Pulmonary Medicine, III, Medical Clinic, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Roland Buhl
- Department of Pulmonary Medicine, III, Medical Clinic, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, 45239 Essen, Germany; and
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, 45239 Essen, Germany; and
| |
Collapse
|
164
|
Persson EK, Verstraete K, Heyndrickx I, Gevaert E, Aegerter H, Percier JM, Deswarte K, Verschueren KHG, Dansercoer A, Gras D, Chanez P, Bachert C, Gonçalves A, Van Gorp H, De Haard H, Blanchetot C, Saunders M, Hammad H, Savvides SN, Lambrecht BN. Protein crystallization promotes type 2 immunity and is reversible by antibody treatment. Science 2019; 364:364/6442/eaaw4295. [PMID: 31123109 DOI: 10.1126/science.aaw4295] [Citation(s) in RCA: 202] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/05/2019] [Indexed: 01/05/2023]
Abstract
Although spontaneous protein crystallization is a rare event in vivo, Charcot-Leyden crystals (CLCs) consisting of galectin-10 (Gal10) protein are frequently observed in eosinophilic diseases, such as asthma. We found that CLCs derived from patients showed crystal packing and Gal10 structure identical to those of Gal10 crystals grown in vitro. When administered to the airways, crystalline Gal10 stimulated innate and adaptive immunity and acted as a type 2 adjuvant. By contrast, a soluble Gal10 mutein was inert. Antibodies directed against key epitopes of the CLC crystallization interface dissolved preexisting CLCs in patient-derived mucus within hours and reversed crystal-driven inflammation, goblet-cell metaplasia, immunoglobulin E (IgE) synthesis, and bronchial hyperreactivity (BHR) in a humanized mouse model of asthma. Thus, protein crystals may promote hallmark features of asthma and are targetable by crystal-dissolving antibodies.
Collapse
Affiliation(s)
- Emma K Persson
- Immunoregulation Unit, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Kenneth Verstraete
- Unit for Structural Biology, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Ines Heyndrickx
- Immunoregulation Unit, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Elien Gevaert
- Upper Airways Research Laboratory, ENT Department, Ghent University Hospital, Ghent, Belgium
| | - Helena Aegerter
- Immunoregulation Unit, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | | | - Kim Deswarte
- Immunoregulation Unit, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Koen H G Verschueren
- Unit for Structural Biology, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Ann Dansercoer
- Unit for Structural Biology, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Delphine Gras
- Aix Marseille University, INSERM, INRA, C2VN, Marseille, France
| | - Pascal Chanez
- Aix Marseille University, INSERM, INRA, C2VN, Marseille, France.,Clinique des Bronches, Allergies et Sommeil, Hôpital Nord, AP-HM, Marseille, France
| | - Claus Bachert
- Upper Airways Research Laboratory, ENT Department, Ghent University Hospital, Ghent, Belgium.,Division of ENT Diseases, CLINTEC, Karolinska Institute, Stockholm, Sweden
| | - Amanda Gonçalves
- BioImaging Core, VIB Inflammation Research Center, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Hanne Van Gorp
- Immunoregulation Unit, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | | | | | | | - Hamida Hammad
- Immunoregulation Unit, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Savvas N Savvides
- Unit for Structural Biology, VIB Center for Inflammation Research, Ghent, Belgium. .,Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Immunoregulation Unit, VIB Center for Inflammation Research, Ghent, Belgium. .,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Department of Pulmonary Medicine, ErasmusMC, Rotterdam, Netherlands
| |
Collapse
|
165
|
Marone G, Granata F, Pucino V, Pecoraro A, Heffler E, Loffredo S, Scadding GW, Varricchi G. The Intriguing Role of Interleukin 13 in the Pathophysiology of Asthma. Front Pharmacol 2019; 10:1387. [PMID: 31866859 PMCID: PMC6908970 DOI: 10.3389/fphar.2019.01387] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/31/2019] [Indexed: 12/22/2022] Open
Abstract
Approximately 5–10% of asthmatic patients worldwide suffer from severe asthma. Experimental and clinical studies have demonstrated that IL-13 is an important cytokine in chronic airways inflammation. IL-13 is involved in Th2 inflammation and has been identified as a possible therapeutic target in the treatment of asthma. Two different human monoclonal antibodies (mAbs) anti-IL-13 (tralokinumab and lebrikizumab) block binding and signaling of IL-13 to its receptors, IL-13Rα1 and IL-13Rα2. Several randomized, double-blind, placebo-controlled multicenter studies have evaluated the safety and efficacy of tralokinumab and lebrikizumab in the treatment of adult patients with severe asthma, but all have failed to meet their primary endpoints. No serious adverse events related to the treatment with these anti-IL-13 mAbs have been reported in these studies. These negative clinical results contrast with positive findings from blocking IL-13 signaling in experimental models of asthma, raising doubts about the transferrable value of some models. Interestingly, dupilumab, a mAb which blocks both IL-4 and IL-13 signaling reduces exacerbation rates and improves lung function in severe asthmatics. These results suggest that IL-4 and IL-13 share some, but not all functional activities in airway inflammation. Tralokinumab might show efficacy in a highly selected cohort of asthmatics characterized by overexpression of IL-13.
Collapse
Affiliation(s)
- Giancarlo Marone
- Department of Public Health, University of Naples Federico II, Naples, Italy.,Azienda Ospedaliera Ospedali dei Colli, Monaldi Hospital Pharmacy, Naples, Italy
| | - Francescopaolo Granata
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, University of Naples Federico II, Naples, Italy
| | - Valentina Pucino
- College of Medical and Dental Sciences, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Antonio Pecoraro
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, United Kingdom
| | - Enrico Heffler
- Personalized Medicine, Asthma, and Allergy, Humanitas Clinical and Research Center, IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, University of Naples Federico II, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| | - Guy W Scadding
- Allergy and Clinical Immunology, Imperial College, National Heart and Lung Institute, London, United Kingdom
| | - Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, University of Naples Federico II, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| |
Collapse
|
166
|
Macmoondongtang modulates Th1-/Th2-related cytokines and alleviates asthma in a murine model. PLoS One 2019; 14:e0224517. [PMID: 31790411 PMCID: PMC6886797 DOI: 10.1371/journal.pone.0224517] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 10/15/2019] [Indexed: 12/31/2022] Open
Abstract
Objective Macmoondongtang has been used as a traditional medicine to treat pulmonary disease in Korea. However, the mechanism underlying its therapeutic effect has yet to be reported. In the present study, the role of macmoondongtang as a respiratory medicine, especially as an anti-asthmatic agent, has been attributed to the down-regulation of interleukin (IL)-4 and tumor necrosis factor (TNF)-α. Materials & methods BALB/c mice were divided into five groups: control, asthma-induced control, dexamethasone treatment, treatment with 150 mg/kg macmoondongtang, and treatment with 1500 mg/kg macmoondongtang. To evaluate the anti-asthmatic effect of macmoondongtang, we investigated its suppressive or inhibitory effects against typical asthmatic changes such as differential cell count in bronchioalveolar fluid (BALF), serum IgE levels, lung morphology, expression of Th1/Th2 cell transcription factors such as T-bet and GATA-3, and Th1-/Th2-/Th17-related cytokines such as interferon (IFN)-γ, IL-12p40, IL-4, -5, -13, TNF-α, and IL-6. The active ingredients in macmoondongtang were further analyzed. Results Macmoondongtang treatment down-regulated serum IgE level, a very important marker of hyper-responsiveness. It reversed typical morphological changes such as mucous hypersecretion, lung epithelial cell hyperplasia, and inflammatory cell infiltration near bronchioalveolar space and veins. Macmoondongtang significantly decreased neutrophil count in BALF, as well as reduced T-bet, IFN-γ, and TNF-α expression in the lung. It also showed a dose-dependent control of inflammatory cells in BALF, controlled the expression of IL-12, IL-4, and IL-5 genes in the lung, and the protein expression of IL12p40, GATA-3, IL-4, IL-5, and IL-13. The component analysis revealed glycyrrhizin and liquiritin as the active ingredients. Conclusions Macmoondongtang treatment alleviates asthma symptoms and modulate the Th1-/Th2- related cytokines. Glycyrrhizin and liquiritin could be the major the active therapeutic components.
Collapse
|
167
|
Maternal Obesity in Mice Exacerbates the Allergic Inflammatory Response in the Airways of Male Offspring. Nutrients 2019; 11:nu11122902. [PMID: 31805682 PMCID: PMC6950392 DOI: 10.3390/nu11122902] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 11/11/2019] [Accepted: 11/19/2019] [Indexed: 02/05/2023] Open
Abstract
It was previously demonstrated that non-allergen-sensitized rodents born to mothers exposed to a high-fat diet (HFD) spontaneously develop lower respiratory compliance and higher respiratory resistance. In the present study, we sought to determine if mice born to mothers consuming HFD would exhibit changes in inflammatory response and lung remodeling when subjected to ovalbumin (OVA) sensitization/challenge in adult life. Mice born to dams consuming either HFD or standard chow had increased bronchoalveolar lavage (BAL) levels of IL-1β, IL-4, IL-5, IL-10, IL-13, TNF-α and TGF-β1 after challenge with OVA. IL-4, IL-13, TNF-α and TGF-β1 levels were further increased in the offspring of HFD-fed mothers. Mice born to obese dams also had exacerbated values of leukocyte infiltration in lung parenchyma, eosinophil and neutrophil counts in BAL, mucus overproduction and collagen deposition. The programming induced by maternal obesity was accompanied by increased expression of miR-155 in peripheral-blood mononuclear cells and reduced miR-133b in trachea and lung tissue in adult life. Altogether, the present data support the unprecedented notion that the progeny of obese mice display exacerbated responses to sensitization/challenge with OVA, leading to the intensification of the morphological changes of lung remodeling. Such changes are likely to result from long-lasting changes in miR-155 and miR-133b expression.
Collapse
|
168
|
Interleukin-13 Stimulation Reveals the Cellular and Functional Plasticity of the Airway Epithelium. Ann Am Thorac Soc 2019; 15:S98-S102. [PMID: 29676620 DOI: 10.1513/annalsats.201711-868mg] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
About 50% of patients with asthma exhibit chronic airway inflammation driven by the type 2 cytokines interleukin (IL)-4, IL-5, and IL-13. These patients with type 2-high asthma experience more allergic symptoms, greater airway hyperresponsiveness, and more severe mucus obstruction than patients with type 2-low asthma. Mouse models of asthma have shown that much of the airway dysfunction in these models can be generated by IL-13 stimulation of the airway epithelium alone. Both in vivo mouse model studies and in vitro studies of human mucociliary airway epithelial cultures have shown that IL-13 induces cellular remodeling of the airway epithelium through proliferation-independent transdifferentiation processes. In both humans and mice, IL-13 stimulation of the airway epithelium results in generation of hypersecretory mucin 5AC (MUC5AC)-expressing mucus cells. Whereas club cells have been shown to be the source of these mucin 5AC-positive mucus cells in mice, the origin of these mucus cells in humans is unclear. In humans, chronic IL-13 stimulation appears to result in loss of ciliated cells. Moreover, IL-13 stimulation can block ciliated cell differentiation from human basal airway epithelial cells. Coincident with IL-13 cellular remodeling are reported decreases in mucociliary transport and ciliary beat frequency. These IL-13-mediated changes in mucociliary function are accompanied by disorganization of cilia, a decrease in the ratio of mucin 5B (MUC5B) to mucin 5AC, and mucus gel tethering to the epithelial surface by mucin 5AC. These airway epithelial responses to IL-13 are mediated by multiple transcription factors, including signal transducer and activator of transcription-6 (STAT6), SAM pointed domain-containing Ets transcription factor (SPDEF), Forkhead box A2 (FOXA2), and Forkhead box J1 (FOXJ1). In addition, analysis of RNA-sequencing data derived from airway epithelial cells shows how IL-13 stimulation promotes broad changes in gene expression across the transcriptome. These results reveal the plastic nature of airway epithelial cells that enables the epithelium to undergo remodeling and functional shifts in response to IL-13 stimulation. With use of new technology, future studies should lead to greater understanding of how IL-13 and other stimuli of disease bring about airway epithelial remodeling, which may aid in the development of therapies that ameliorate airway dysfunction in asthma and other diseases.
Collapse
|
169
|
Abstract
The presentation, pathobiology, and prognosis of asthma are highly heterogeneous and challenging for clinicians to diagnose and treat. In addition to the adaptive immune response that underlies allergic inflammation, innate immune mechanisms are increasingly recognized to be critical mediators of the eosinophilic airway inflammation present in most patients with asthma. Efforts to classify patients by severity and immune response have identified a number of different clinical and immune phenotypes, indicating that the innate and adaptive immune responses are differentially active among patients with the disease. Advances in the detection of these subgroups using clinical characteristics and biomarkers have led to the successful development of targeted biologics. This has moved us to a more personalized approach to managing asthma. Here we review the emerging endotypes of asthma and the biologics that have been developed to treat them.
Collapse
Affiliation(s)
- Geoffrey Lowell Chupp
- Division of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut 06520, USA;
| | - Ravdeep Kaur
- Division of Allergy and Immunology, Yale School of Medicine, New Haven, Connecticut 06520, USA
| | - Anne Mainardi
- Division of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut 06520, USA;
| |
Collapse
|
170
|
Morrison CB, Markovetz MR, Ehre C. Mucus, mucins, and cystic fibrosis. Pediatr Pulmonol 2019; 54 Suppl 3:S84-S96. [PMID: 31715083 PMCID: PMC6853602 DOI: 10.1002/ppul.24530] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/06/2019] [Indexed: 02/06/2023]
Abstract
Cystic fibrosis (CF) is both the most common and most lethal genetic disease in the Caucasian population. CF is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene and is characterized by the accumulation of thick, adherent mucus plaques in multiple organs, of which the lungs, gastrointestinal tract and pancreatic ducts are the most commonly affected. A similar pathogenesis cascade is observed in all of these organs: loss of CFTR function leads to altered ion transport, consisting of decreased chloride and bicarbonate secretion via the CFTR channel and increased sodium absorption via epithelial sodium channel upregulation. Mucosa exposed to changes in ionic concentrations sustain severe pathophysiological consequences. Altered mucus biophysical properties and weakened innate defense mechanisms ensue, furthering the progression of the disease. Mucins, the high-molecular-weight glycoproteins responsible for the viscoelastic properties of the mucus, play a key role in the disease but the actual mechanism of mucus accumulation is still undetermined. Multiple hypotheses regarding the impact of CFTR malfunction on mucus have been proposed and are reviewed here. (a) Dehydration increases mucin monomer entanglement, (b) defective Ca2+ chelation compromises mucin expansion, (c) ionic changes alter mucin interactions, and (d) reactive oxygen species increase mucin crosslinking. Although one biochemical change may dominate, it is likely that all of these mechanisms play some role in the progression of CF disease. This article discusses recent findings on the initial cause(s) of aberrant mucus properties in CF and examines therapeutic approaches aimed at correcting mucus properties.
Collapse
Affiliation(s)
- Cameron Bradley Morrison
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Matthew Raymond Markovetz
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Camille Ehre
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Division of Pediatric Pulmonology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
171
|
Caminati M, Polk B, Rosenwasser LJ. What have recent advances in therapy taught us about severe asthma disease mechanisms? Expert Rev Clin Immunol 2019; 15:1145-1153. [PMID: 31549894 DOI: 10.1080/1744666x.2020.1672536] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Introduction: Severe asthma still represents a worldwide challenge. The need for further treatment options has stimulated basic and pharmacological research to focus on the immune and inflammatory background of asthma. The new biologic drugs express the considerable advances in the field and besides providing a revolutionary treatment option for severe asthma, contribute themselves to better understand the pathophysiologic mechanisms they address, paving the way to new potential targets.Areas covered: A selective search on PubMed and Medline was performed, including the evidence on immunology of severe asthma published up to May 2019 by focusing on the immunological effects of biologic drugs underlying their clinical outcomes.Expert opinion: The recent pharmacological research in the field of biologics has represented an exceptional opportunity for exploring severe asthma mechanisms. However, some points deserve to be addressed by further investigation. Although in the absence of safety warnings so far, interfering with the immune system may raise some safety concerns, especially in the long-term use. Particularly when interacting with epithelial and innate immunity the selection of candidates probably deserves special caution. Also, whether biologics exert a true disease-modifying effect is not completely clear. As a direct practical implication, the optimal treatment duration is still controversial.
Collapse
Affiliation(s)
- Marco Caminati
- Asthma Center and Allergy Unit, Verona University Hospital, Verona, Italy.,Department of Medicine, University of Verona, Verona, Italy
| | - Brooke Polk
- Wash U School of Medicine, St Louis, MO, USA
| | | |
Collapse
|
172
|
Somogyi V, Chaudhuri N, Torrisi SE, Kahn N, Müller V, Kreuter M. The therapy of idiopathic pulmonary fibrosis: what is next? Eur Respir Rev 2019; 28:190021. [PMID: 31484664 PMCID: PMC9488691 DOI: 10.1183/16000617.0021-2019] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrosing interstitial lung disease, characterised by progressive scarring of the lung and associated with a high burden of disease and early death. The pathophysiological understanding, clinical diagnostics and therapy of IPF have significantly evolved in recent years. While the recent introduction of the two antifibrotic drugs pirfenidone and nintedanib led to a significant reduction in lung function decline, there is still no cure for IPF; thus, new therapeutic approaches are needed. Currently, several clinical phase I-III trials are focusing on novel therapeutic targets. Furthermore, new approaches in nonpharmacological treatments in palliative care, pulmonary rehabilitation, lung transplantation, management of comorbidities and acute exacerbations aim to improve symptom control and quality of life. Here we summarise new therapeutic attempts and potential future approaches to treat this devastating disease.
Collapse
Affiliation(s)
- Vivien Somogyi
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
- Dept of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Nazia Chaudhuri
- Manchester University NHS Foundation Trust, Wythenshawe Hospital, Manchester, UK
| | - Sebastiano Emanuele Torrisi
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
- Regional Referral Centre for Rare Lung Diseases, University Hospital "Policlinico", Dept of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Nicolas Kahn
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| | - Veronika Müller
- Dept of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Michael Kreuter
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
173
|
Farcas MT, Stefaniak AB, Knepp AK, Bowers L, Mandler WK, Kashon M, Jackson SR, Stueckle TA, Sisler JD, Friend SA, Qi C, Hammond DR, Thomas TA, Matheson J, Castranova V, Qian Y. Acrylonitrile butadiene styrene (ABS) and polycarbonate (PC) filaments three-dimensional (3-D) printer emissions-induced cell toxicity. Toxicol Lett 2019; 317:1-12. [PMID: 31562913 DOI: 10.1016/j.toxlet.2019.09.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/30/2019] [Accepted: 09/14/2019] [Indexed: 10/26/2022]
Abstract
During extrusion of some polymers, fused filament fabrication (FFF) 3-D printers emit billions of particles per minute and numerous organic compounds. The scope of this study was to evaluate FFF 3-D printer emission-induced toxicity in human small airway epithelial cells (SAEC). Emissions were generated from a commercially available 3-D printer inside a chamber, while operating for 1.5 h with acrylonitrile butadiene styrene (ABS) or polycarbonate (PC) filaments, and collected in cell culture medium. Characterization of the culture medium revealed that repeat print runs with an identical filament yield various amounts of particles and organic compounds. Mean particle sizes in cell culture medium were 201 ± 18 nm and 202 ± 8 nm for PC and ABS, respectively. At 24 h post-exposure, both PC and ABS emissions induced a dose dependent significant cytotoxicity, oxidative stress, apoptosis, necrosis, and production of pro-inflammatory cytokines and chemokines in SAEC. Though the emissions may not completely represent all possible exposure scenarios, this study indicate that the FFF could induce toxicological effects. Further studies are needed to quantify the detected chemicals in the emissions and their corresponding toxicological effects.
Collapse
Affiliation(s)
- Mariana T Farcas
- Pathology and Physiology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA; Pharmaceutical and Pharmacological Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, 26505, USA.
| | - Aleksandr B Stefaniak
- Field Studies Branch, Respiratory Health Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA.
| | - Alycia K Knepp
- Field Studies Branch, Respiratory Health Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA.
| | - Lauren Bowers
- Field Studies Branch, Respiratory Health Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA.
| | - William K Mandler
- Pathology and Physiology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA.
| | - Michael Kashon
- Biostatistics and Epidemiology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA.
| | - Stephen R Jackson
- Exposure Assessment Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA.
| | - Todd A Stueckle
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA.
| | - Jenifer D Sisler
- Pathology and Physiology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA.
| | - Sherri A Friend
- Pathology and Physiology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA.
| | - Chaolong Qi
- Engineering and Physical Hazards Branch, Division of Applied Research & Technology, National Institute for Occupational Safety and Health, Cincinnati, OH, USA.
| | - Duane R Hammond
- Engineering and Physical Hazards Branch, Division of Applied Research & Technology, National Institute for Occupational Safety and Health, Cincinnati, OH, USA.
| | - Treye A Thomas
- Office of Hazard Identification and Reduction, U.S. Consumer Product Safety Commission, Rockville, MD, USA.
| | - Joanna Matheson
- Office of Hazard Identification and Reduction, U.S. Consumer Product Safety Commission, Rockville, MD, USA.
| | - Vincent Castranova
- Pharmaceutical and Pharmacological Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, 26505, USA.
| | - Yong Qian
- Pathology and Physiology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA.
| |
Collapse
|
174
|
Abstract
Fibrosis is the abnormal deposition of extracellular matrix, which can lead to organ dysfunction, morbidity, and death. The disease burden caused by fibrosis is substantial, and there are currently no therapies that can prevent or reverse fibrosis. Metabolic alterations are increasingly recognized as an important pathogenic process that underlies fibrosis across many organ types. As a result, metabolically targeted therapies could become important strategies for fibrosis reduction. Indeed, some of the pathways targeted by antifibrotic drugs in development - such as the activation of transforming growth factor-β and the deposition of extracellular matrix - have metabolic implications. This Review summarizes the evidence to date and describes novel opportunities for the discovery and development of drugs for metabolic reprogramming, their associated challenges, and their utility in reducing fibrosis. Fibrotic therapies are potentially relevant to numerous common diseases such as cirrhosis, non-alcoholic steatohepatitis, chronic renal disease, heart failure, diabetes, idiopathic pulmonary fibrosis, and scleroderma.
Collapse
|
175
|
Lightwood D, Tservistas M, Zehentleitner M, Sarkar K, Turner A, Bracher M, Smith B, Lamour S, Bourne T, Shaw S, Gozzard N, Palframan RT. Efficacy of an Inhaled IL-13 Antibody Fragment in a Model of Chronic Asthma. Am J Respir Crit Care Med 2019; 198:610-619. [PMID: 29883204 DOI: 10.1164/rccm.201712-2382oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
RATIONALE IL-13 is an important cytokine implicated in the pathogenesis of allergic asthma and is an attractive target for an inhaled therapeutic. OBJECTIVE To investigate the efficacy of CDP7766, a nebulized inhaled anti-IL-13 monoclonal antibody Fab fragment, in a model of allergic asthma in cynomolgus macaques naturally sensitized to Ascaris suum. METHODS CDP7766 was nebulized using a vibrating-membrane nebulizer on the basis of eFlow technology. The aerosol generated was analyzed to determine the particle size profile and the biophysical and functional properties of CDP7766. Nebulized CDP7766 (0.1-60 mg/animal, once daily for 5 d) was delivered via the inhaled route. MEASUREMENTS AND MAIN RESULTS The investigational eFlow nebulizer used in this study generated a respirable aerosol of CDP7766 with no evidence of degradation, loss of potency, aggregation, or formation of particulates. Inhaled CDP7766 was well tolerated in the model (no adverse effects related to local irritation) and significantly inhibited BAL allergen-induced cytokine and chemokine upregulation (60 mg vs. vehicle: eotaxin-3, P < 0.0008; MIP [macrophage inflammatory protein]-1β, IL-8, IFN-γ, P ≤ 0.01). CDP7766 significantly inhibited the increase in pulmonary resistance stimulated by inhaled allergen, measured 15 minutes and 24 hours after allergen challenge. CONCLUSION Inhaled CDP7766 potently inhibited the function of IL-13 generated during the airway response to inhaled allergen in cynomolgus macaques, demonstrating the potential of inhaled anti-IL-13 therapeutics for the treatment of allergic asthma.
Collapse
Affiliation(s)
| | | | | | | | - Alison Turner
- 1 UCB Pharma, Slough, Berkshire, United Kingdom; and
| | | | - Bryan Smith
- 1 UCB Pharma, Slough, Berkshire, United Kingdom; and
| | | | - Tim Bourne
- 1 UCB Pharma, Slough, Berkshire, United Kingdom; and
| | - Stevan Shaw
- 1 UCB Pharma, Slough, Berkshire, United Kingdom; and
| | - Neil Gozzard
- 1 UCB Pharma, Slough, Berkshire, United Kingdom; and
| | | |
Collapse
|
176
|
Wu DM, Zheng ZH, Wang S, Wen X, Han XR, Wang YJ, Shen M, Fan SH, Zhang ZF, Shan Q, Li MQ, Hu B, Zheng YL, Chen GQ, Lu J. Retracted: The role of HOTAIR-induced downregulation of microRNA-126 and interleukin-13 in the development of bronchial hyperresponsiveness in neonates. J Cell Physiol 2019; 234:16400-16411. [PMID: 30790266 DOI: 10.1002/jcp.28309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/18/2019] [Accepted: 01/24/2019] [Indexed: 01/24/2023]
Abstract
Long noncoding RNAs, including HOTAIR, are involved in the pathogenesis of a wide range of diseases. This study aimed to explore the mechanism underlying the involvement of HOTAIR in neonatal bronchial hyperresponsiveness (BHR). A total of 105 newborns were recruited in this study to collect their peripheral blood mononuclear cell and serum samples, which were then divided into different genotype groups based on the genotypes of rs4759314, rs874945, and rs7958904. The real-time polymerase chain reaction, western blot analysis, computational analyses, and luciferase assays were performed to establish the regulatory relationships between the HOTAIR, microRNA-126 (miR-126), and interleukin-13 (IL-13). The level of HOTAIR, miR-126, and IL-13 among rs4759314 AA, AG, and GG groups, as well as among rs874945 GG, AG, and AA groups was similar. However, the level of HOTAIR was increased in the rs7958904 GG group, accompanied by a decreased level of miR-126 and IL-13. In addition, the level of airway responsiveness was comparable among rs4759314 AA, AG, and GG groups, as well as among rs874945 GG, AG, and AA groups. However, the airway responsiveness in the groups rs7958904 CG and CC was much stronger than that of the GG group. We also demonstrated that, by directly binding to miR-126, HOTAIR reduced the expression of miR-126, which in turn decreased the expression of IL-13. In summary, we demonstrated the role of HOTAIR-induced downregulation of miR-126 and IL-13 in the development of BHR in neonates.
Collapse
Affiliation(s)
- Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu, School of Life Science, College of Health Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Zi-Hui Zheng
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu, School of Life Science, College of Health Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu, School of Life Science, College of Health Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Xin-Rui Han
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu, School of Life Science, College of Health Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Yong-Jian Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu, School of Life Science, College of Health Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Min Shen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu, School of Life Science, College of Health Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Shao-Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu, School of Life Science, College of Health Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Zi-Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu, School of Life Science, College of Health Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu, School of Life Science, College of Health Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Meng-Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu, School of Life Science, College of Health Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu, School of Life Science, College of Health Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu, School of Life Science, College of Health Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| | - Gui-Quan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu, School of Life Science, College of Health Sciences, Jiangsu Normal University, Xuzhou, China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, China
| |
Collapse
|
177
|
Zanoni M, Cortesi M, Zamagni A, Tesei A. The Role of Mesenchymal Stem Cells in Radiation-Induced Lung Fibrosis. Int J Mol Sci 2019; 20:E3876. [PMID: 31398940 PMCID: PMC6719901 DOI: 10.3390/ijms20163876] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023] Open
Abstract
Radiation therapy is one of the most important treatment modalities for thoracic tumors. Despite significant advances in radiation techniques, radiation-induced lung injury (RILI) still occurs in up to 30% of patients undergoing thoracic radiotherapy, and therefore remains the main dose-limiting obstacle. RILI is a potentially lethal clinical complication of radiotherapy that has 2 main stages: an acute stage defined as radiation pneumonitis, and a late stage defined as radiation-induced lung fibrosis. Patients who develop lung fibrosis have a reduced quality of life with progressive and irreversible organ malfunction. Currently, the most effective intervention for the treatment of lung fibrosis is lung transplantation, but the lack of available lungs and transplantation-related complications severely limits the success of this procedure. Over the last few decades, advances have been reported in the use of mesenchymal stem cells (MSCs) for lung tissue repair and regeneration. MSCs not only replace damaged lung epithelial cells but also promote tissue repair through the secretion of anti-inflammatory and anti-fibrotic factors. Here, we present an overview of MSC-based therapy for radiation-induced lung fibrosis, focusing in particular on the molecular mechanisms involved and describing the most recent preclinical and clinical studies carried out in the field.
Collapse
Affiliation(s)
- Michele Zanoni
- Bioscience Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Michela Cortesi
- Bioscience Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Alice Zamagni
- Bioscience Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Anna Tesei
- Bioscience Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| |
Collapse
|
178
|
Bok SH, Seo JH, Bae CS, Kang B, Cho SS, Park DH. Allium hookeri root extract regulates asthmatic changes through immunological modulation of Th1/Th2‑related factors in an ovalbumin‑induced asthma mouse model. Mol Med Rep 2019; 20:3215-3223. [PMID: 31432168 PMCID: PMC6755185 DOI: 10.3892/mmr.2019.10560] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 06/26/2019] [Indexed: 12/30/2022] Open
Abstract
In 2013, WHO estimated that approximately 235 million people suffered from asthma worldwide. Asthma is a hyper responsive disorder, which is related to an imbalance between the T-helper type 1 and 2 cells (henceforth, Th1 and Th2, respectively). Allium hookeri is a plant that is widely used for culinary purposes and also in traditional Asian medicine. The present study was conducted to elucidate the anti-asthmatic effects and mechanism of action of A. hookeri root extracts (AHRE) in an ovalbumin (OVA)-induced asthma mouse model. The mice were divided into five groups, namely, the control, the OVA-treated group, the dexamethasone-treated group, the 30 mg/kg AHRE-treated group, and the 300 mg/kg AHRE-treated group. The total WBC count and the differential cell count in the bronchoalveolar fluid, the level of serum IgE, the histopathological changes in the lung, and changes in the cell surface molecules, the asthma-related cytokine levels, and Th cell transcription factors were evaluated. AHRE significantly ameliorated asthmatic changes, such as the total WBC count, eosinophil count, and the level of IgE; in addition, it reduced mucus hypersecretion, epithelial hyperplasia, and eosinophil infiltration in the lungs. AHRE significantly inhibited the expression of CD68+ cells and MHC class II+ molecules, Th1 cell transcription factor (T-bet) activation, Th2 cell transcription factor (GATA-3) activation, and TNF-α in the lung tissue. Furthermore, it suppressed cell surface molecules, such as CD4+and CD8+; Th1-related cytokines, such as IFN-γ and IL-12p40; Th2-related cytokines, such as IL-4 and IL-5; and Th17-related cytokines, such as IL-6 and TNF-α, in a dose-dependent manner. Thus, AHRE may be considered a promising anti-asthmatic drug.
Collapse
Affiliation(s)
- So-Hyeon Bok
- College of Oriental Medicine, Dongshin University, Naju, Jeonnam 58245, Republic of Korea
| | - Ji-Hye Seo
- College of Oriental Medicine, Dongshin University, Naju, Jeonnam 58245, Republic of Korea
| | - Chun-Sik Bae
- College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Bossng Kang
- Department of Emergency Medicine, College of Medicine, Hanyang University, Guri, Gyunggi 11923, Republic of Korea
| | - Seung Sik Cho
- College of Pharmacy, Mokpo National University, Muan, Jeonnam 58554, Republic of Korea
| | - Dae-Hun Park
- College of Oriental Medicine, Dongshin University, Naju, Jeonnam 58245, Republic of Korea
| |
Collapse
|
179
|
Gazzinelli-Guimaraes PH, de Queiroz Prado R, Ricciardi A, Bonne-Année S, Sciurba J, Karmele EP, Fujiwara RT, Nutman TB. Allergen presensitization drives an eosinophil-dependent arrest in lung-specific helminth development. J Clin Invest 2019; 129:3686-3701. [PMID: 31380805 DOI: 10.1172/jci127963] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022] Open
Abstract
This study investigates the relationship between helminth infection and allergic sensitization by assessing the influence of preexisting allergy on the outcome of helminth infections, rather than the more traditional approach in which the helminth infection precedes the onset of allergy. Here we used a murine model of house dust mite-induced (HDM-induced) allergic inflammation followed by Ascaris infection to demonstrate that allergic sensitization drives an eosinophil-rich pulmonary type 2 immune response (Th2 cells, M2 macrophages, type 2 innate lymphoid cells, IL-33, IL-4, IL-13, and mucus) that directly hinders larval development and reduces markedly the parasite burden in the lungs. This effect is dependent on the presence of eosinophils, as eosinophil-deficient mice were unable to limit parasite development or numbers. In vivo administration of neutralizing antibodies against CD4 prior to HDM sensitization significantly reduced eosinophils in the lungs, resulting in the reversal of the HDM-induced Ascaris larval killing. Our data suggest that HDM allergic sensitization drives a response that mimics a primary Ascaris infection, such that CD4+ Th2-mediated eosinophil-dependent helminth larval killing in the lung tissue occurs. This study provides insight into the mechanisms underlying tissue-specific responses that drive a protective response against the early stages of the helminths prior to their establishing long-lasting infections in the host.
Collapse
Affiliation(s)
- Pedro H Gazzinelli-Guimaraes
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Rafael de Queiroz Prado
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Alessandra Ricciardi
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Sandra Bonne-Année
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Joshua Sciurba
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Erik P Karmele
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA.,Institute for Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Ricardo T Fujiwara
- Department of Parasitology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thomas B Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
180
|
Chung EJ, Reedy JL, Kwon S, Patil S, Valle L, White AO, Citrin DE. 12-Lipoxygenase is a Critical Mediator of Type II Pneumocyte Senescence, Macrophage Polarization and Pulmonary Fibrosis after Irradiation. Radiat Res 2019; 192:367-379. [PMID: 31373871 PMCID: PMC6816027 DOI: 10.1667/rr15356.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Radiation-induced pulmonary fibrosis (RIPF) is a chronic, progressive complication of therapeutic irradiation of the thorax. It has been suggested that senescence of type II pneumocytes (AECIIs), an alveolar stem cell, plays a role in the development of RIPF through loss of replicative reserve and via senescent AECII-driven release of proinflammatory and profibrotic cytokines. Within this context, we hypothesized that arachidonate 12-lipoxygenase (12-LOX) is a critical mediator of AECII senescence and RIPF. Treatment of wild-type AECIIs with 12S-hydroxyeicosateraenoic acid (12S-HETE), a downstream product of 12-LOX, was sufficient to induce senescence in a NADPH oxidase 4 (NOX4)-dependent manner. Mice deficient in 12-LOX exhibited reduced AECII senescence, pulmonary collagen accumulation and accumulation of alternatively activated (M2) macrophages after thoracic irradiation (5 × 6 Gy) compared to wild-type mice. Conditioned media from irradiated or 12S-HETE-treated primary pneumocytes contained elevated levels of IL-4 and IL-13 compared to untreated pneumocytes. Primary macrophages treated with conditioned media from irradiated AECII demonstrated preferential M2 type polarization when AECIIs were derived from wild-type mice compared to 12-LOX-deficient mice. Together, these data identified 12-LOX as a critical component of RIPF and a therapeutic target for radiation-induced lung injury.
Collapse
Affiliation(s)
- Eun Joo Chung
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Jessica L. Reedy
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Seokjoo Kwon
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Shilpa Patil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Luca Valle
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Ayla O. White
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Deborah E. Citrin
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
181
|
Yang J, Ramirez Moral I, van 't Veer C, de Vos AF, de Beer R, Roelofs JJTH, Morgan BP, van der Poll T. Complement factor C5 inhibition reduces type 2 responses without affecting group 2 innate lymphoid cells in a house dust mite induced murine asthma model. Respir Res 2019; 20:165. [PMID: 31340811 PMCID: PMC6657208 DOI: 10.1186/s12931-019-1136-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/16/2019] [Indexed: 01/13/2023] Open
Abstract
Background Complement factor C5 can either aggravate or attenuate the T-helper type 2 (TH2) immune response and airway hyperresponsiveness (AHR) in murine models of allergic asthma. The effect of C5 during the effector phase of allergen-induced asthma is ill-defined. Objectives We aimed to determine the effect of C5 blockade during the effector phase on the pulmonary TH2 response and AHR in a house dust mite (HDM) driven murine asthma model. Methods BALB/c mice were sensitized and challenged repeatedly with HDM via the airways to induce allergic lung inflammation. Sensitized mice received twice weekly injections with a blocking anti-C5 or control antibody 24 h before the first challenge. Results HDM challenge in sensitized mice resulted in elevated C5a levels in bronchoalveolar lavage fluid. Anti-C5 administered to sensitized mice prior to the first HDM challenge prevented this rise in C5a, but did not influence the influx of eosinophils or neutrophils. While anti-C5 did not impact the recruitment of CD4 T cells upon HDM challenge, it reduced the proportion of TH2 cells recruited to the airways, attenuated IL-4 release by regional lymph nodes restimulated with HDM ex vivo and mitigated the plasma IgE response. Anti-C5 did not affect innate lymphoid cell (ILC) proliferation or group 2 ILC (ILC2) differentiation. Anti-C5 attenuated HDM induced AHR in the absence of an effect on lung histopathology, mucus production or vascular leak. Conclusions Generation of C5a during the effector phase of HDM induced allergic lung inflammation contributes to TH2 cell differentiation and AHR without impacting ILC2 cells. Electronic supplementary material The online version of this article (10.1186/s12931-019-1136-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jack Yang
- Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - Ivan Ramirez Moral
- Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Cornelis van 't Veer
- Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Alex F de Vos
- Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Regina de Beer
- Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - B Paul Morgan
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Division of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
182
|
Gottlow M, Svensson DJ, Lipkovich I, Huhn M, Bowen K, Wessman P, Colice G. Application of structured statistical analyses to identify a biomarker predictive of enhanced tralokinumab efficacy in phase III clinical trials for severe, uncontrolled asthma. BMC Pulm Med 2019; 19:129. [PMID: 31315668 PMCID: PMC6637533 DOI: 10.1186/s12890-019-0889-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 07/02/2019] [Indexed: 11/10/2022] Open
Abstract
Background Tralokinumab is an anti–interleukin (IL)-13 monoclonal antibody investigated for the treatment of severe, uncontrolled asthma in two Phase III clinical trials, STRATOS 1 and 2. The STRATOS 1 biomarker analysis plan was developed to identify biomarker(s) indicative of IL-13 activation likely to predict tralokinumab efficacy and define a population in which there was an enhanced treatment effect; this defined population was then tested in STRATOS 2. Methods The biomarkers considered were blood eosinophil counts, fractional exhaled nitric oxide (FeNO), serum dipeptidyl peptidase-4, serum periostin and total serum immunoglobulin E. Tralokinumab efficacy was measured as the reduction in annualised asthma exacerbation rate (AAER) compared with placebo (primary endpoint measure of STRATOS 1 and 2). The biomarker analysis plan included negative binomial and generalised additive models, and the Subgroup Identification based on Differential Effect Search (SIDES) algorithm, supported by robustness and sensitivity checks. Effects on the key secondary endpoints of STRATOS 1 and 2, which included changes from baseline in standard measures of asthma outcomes, were also investigated. Prior to the STRATOS 1 read-out, numerous simulations of the methodology were performed with hypothetical data. Results FeNO and periostin were identified as the only biomarkers potentially predictive of treatment effect, with cut-offs chosen by the SIDES algorithm of > 32.3 ppb and > 27.4 ng/ml, respectively. The FeNO > 32.3 ppb subgroup was associated with greater AAER reductions and improvements in key secondary endpoints compared with the periostin > 27.4 ng/ml subgroup. Upon further evaluation of AAER reductions at different FeNO cut-offs, ≥37 ppb was chosen as the best cut-off for predicting tralokinumab efficacy. Discussion A rigorous statistical approach incorporating multiple methods was used to investigate the predictive properties of five potential biomarkers and to identify a participant subgroup that demonstrated an enhanced tralokinumab treatment effect. Using STRATOS 1 data, our analyses identified FeNO at a cut-off of ≥37 ppb as the best assessed biomarker for predicting enhanced treatment effect to be tested in STRATOS 2. Our findings were inconclusive, which reflects the complexity of subgroup identification in the severe asthma population. Trial registration STRATOS 1 and 2 are registered on ClinicalTrials.gov (NCT02161757 registered on June 12, 2014, and NCT02194699 registered on July 18, 2014). Electronic supplementary material The online version of this article (10.1186/s12890-019-0889-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mattis Gottlow
- Biometrics and Information Sciences, AstraZeneca, Pepparedsleden 1, SE-431 83, Mölndal, Sweden
| | - David J Svensson
- Biometrics and Information Sciences, AstraZeneca, Pepparedsleden 1, SE-431 83, Mölndal, Sweden
| | - Ilya Lipkovich
- IQVIA, 4820 Emperor Blvd, Durham, NC, 27703, USA.,Present Address: Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, 46285, USA
| | - Monika Huhn
- Biometrics and Information Sciences, AstraZeneca, Pepparedsleden 1, SE-431 83, Mölndal, Sweden
| | - Karin Bowen
- Biometrics and Information Sciences, AstraZeneca, One MedImmune Way, Gaithersburg, MD, 20878, USA
| | - Peter Wessman
- Biometrics and Information Sciences, AstraZeneca, Pepparedsleden 1, SE-431 83, Mölndal, Sweden
| | - Gene Colice
- Global Medicines Development, AstraZeneca, One MedImmune Way, Gaithersburg, MD, 20878, USA.
| |
Collapse
|
183
|
Immunopathology of Airway Surface Liquid Dehydration Disease. J Immunol Res 2019; 2019:2180409. [PMID: 31396541 PMCID: PMC6664684 DOI: 10.1155/2019/2180409] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/29/2019] [Accepted: 05/26/2019] [Indexed: 12/30/2022] Open
Abstract
The primary purpose of pulmonary ventilation is to supply oxygen (O2) for sustained aerobic respiration in multicellular organisms. However, a plethora of abiotic insults and airborne pathogens present in the environment are occasionally introduced into the airspaces during inhalation, which could be detrimental to the structural integrity and functioning of the respiratory system. Multiple layers of host defense act in concert to eliminate unwanted constituents from the airspaces. In particular, the mucociliary escalator provides an effective mechanism for the continuous removal of inhaled insults including pathogens. Defects in the functioning of the mucociliary escalator compromise the mucociliary clearance (MCC) of inhaled pathogens, which favors microbial lung infection. Defective MCC is often associated with airway mucoobstruction, increased occurrence of respiratory infections, and progressive decrease in lung function in mucoobstructive lung diseases including cystic fibrosis (CF). In this disease, a mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene results in dehydration of the airway surface liquid (ASL) layer. Several mice models of Cftr mutation have been developed; however, none of these models recapitulate human CF-like mucoobstructive lung disease. As an alternative, the Scnn1b transgenic (Scnn1b-Tg+) mouse model overexpressing a transgene encoding sodium channel nonvoltage-gated 1, beta subunit (Scnn1b) in airway club cells is available. The Scnn1b-Tg+ mouse model exhibits airway surface liquid (ASL) dehydration, impaired MCC, increased mucus production, and early spontaneous pulmonary bacterial infections. High morbidity and mortality among mucoobstructive disease patients, high economic and health burden, and lack of scientific understanding of the progression of mucoobstruction warrants in-depth investigation of the cause of mucoobstruction in mucoobstructive disease models. In this review, we will summarize published literature on the Scnn1b-Tg+ mouse and analyze various unanswered questions on the initiation and progression of mucobstruction and bacterial infections.
Collapse
|
184
|
Huang Z, Cao Y, Zhou M, Qi X, Fu B, Mou Y, Wu G, Xie J, Zhao J, Xiong W. Hsa_circ_0005519 increases IL-13/IL-6 by regulating hsa-let-7a-5p in CD4 + T cells to affect asthma. Clin Exp Allergy 2019; 49:1116-1127. [PMID: 31148290 DOI: 10.1111/cea.13445] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 04/30/2019] [Accepted: 05/13/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND Circular RNAs (circRNAs) are a class of non-coding RNAs that could serve as novel biomarkers for the diagnosis and treatment of diseases. We hypothesized that circRNAs of CD4+ T cells are involved in asthma. OBJECTIVE In this study, we investigated the circRNA expression profile and the possible mechanism by which hsa_circ_0005519 participates in asthma. METHODS The expression profiles of circRNAs in CD4+ T cells were revealed by circRNA microarray. Hsa_circ_0005519 expression in CD4+ T cells was confirmed in asthmatic patients (n = 65) and healthy subjects (n = 30). Hsa-let-7a-5p, the target of hsa_circ_0005519, was predicted by online algorithms and verified by a dual-luciferase reporter assay. Correlation assays between the expression of hsa_circ_0005519 and hsa-let-7a-5p, the mRNA levels of interleukin (IL)-13 and IL-6 in CD4+ T cells, and the clinical characteristics of asthmatic patients were performed. The role of hsa_circ_0005519 in proinflammatory cytokine expression was investigated in CD4+ T cells from asthmatic patients in vitro. Hsa_circ_0005519 expression in PBMCs was determined in another cohort including 30 asthmatic patients and 24 controls. Correlation assays of hsa_circ_0005519 expressions between CD4+ T cells and PBMCs were performed. RESULTS Hsa_circ_0005519 was up-regulated and negatively correlated with hsa-let-7a-5p expression in CD4+ T cells of asthmatic patients. Both the fraction of exhaled nitric oxide (FeNO) and the peripheral blood eosinophil ratio were positively correlated with hsa_circ_0005519 expression in CD4+ T cells. These outcomes were also different in asthmatic patients with low vs high hsa_circ_0005519 levels. Hsa_circ_0005519 expressions between CD4+ T cells and PBMCs were concordant in asthmatic patients. Mechanistically, hsa_circ_0005519 might bind to hsa-let-7a-5p and relieve suppression for IL-13/IL-6 in CD4+ T cells. CONCLUSIONS AND CLINICAL RELEVANCE Our data suggest that hsa_circ_0005519 may induce IL-13 and IL-6 expression by regulating hsa-let-7a-5p in CD4+ T cells to affect asthma. And hsa_circ_0005519 may be a potential biomarker of asthma.
Collapse
Affiliation(s)
- Zhenli Huang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Yong Cao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Min Zhou
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Xuefei Qi
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Bohua Fu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Yong Mou
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Guorao Wu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Jianping Zhao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Weining Xiong
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| |
Collapse
|
185
|
Antonioli L, Blandizzi C, Pacher P, Haskó G. The Purinergic System as a Pharmacological Target for the Treatment of Immune-Mediated Inflammatory Diseases. Pharmacol Rev 2019; 71:345-382. [PMID: 31235653 PMCID: PMC6592405 DOI: 10.1124/pr.117.014878] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Immune-mediated inflammatory diseases (IMIDs) encompass a wide range of seemingly unrelated conditions, such as multiple sclerosis, rheumatoid arthritis, psoriasis, inflammatory bowel diseases, asthma, chronic obstructive pulmonary disease, and systemic lupus erythematosus. Despite differing etiologies, these diseases share common inflammatory pathways, which lead to damage in primary target organs and frequently to a plethora of systemic effects as well. The purinergic signaling complex comprising extracellular nucleotides and nucleosides and their receptors, the P2 and P1 purinergic receptors, respectively, as well as catabolic enzymes and nucleoside transporters is a major regulatory system in the body. The purinergic signaling complex can regulate the development and course of IMIDs. Here we provide a comprehensive review on the role of purinergic signaling in controlling immunity, inflammation, and organ function in IMIDs. In addition, we discuss the possible therapeutic applications of drugs acting on purinergic pathways, which have been entering clinical development, to manage patients suffering from IMIDs.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (L.A., C.B.); Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (P.P.); and Department of Anesthesiology, Columbia University, New York, New York (G.H.)
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (L.A., C.B.); Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (P.P.); and Department of Anesthesiology, Columbia University, New York, New York (G.H.)
| | - Pál Pacher
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (L.A., C.B.); Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (P.P.); and Department of Anesthesiology, Columbia University, New York, New York (G.H.)
| | - György Haskó
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (L.A., C.B.); Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (P.P.); and Department of Anesthesiology, Columbia University, New York, New York (G.H.)
| |
Collapse
|
186
|
Abstract
Asthma is a genetically and phenotypically complex disease that has a major impact on global health. Signs and symptoms of asthma are caused by the obstruction of airflow through the airways. The epithelium that lines the airways plays a major role in maintaining airway patency and in host defense. The epithelium initiates responses to inhaled or aspirated substances, including allergens, viruses, and bacteria, and epithelial-derived cytokines are important in the recruitment and activation of immune cells in the airway. Changes in the structure and function of the airway epithelium are a prominent feature of asthma. Approximately half of individuals with asthma have evidence of active type 2 immune responses in the airway. In these individuals, epithelial cytokines promote type 2 responses, and responses to type 2 cytokines result in increased epithelial mucus production and other effects that cause airway obstruction. Recent work also implicates other epithelial responses, including interleukin-17, interferon and ER stress responses, that may contribute to asthma pathogenesis and provide new targets for therapy.
Collapse
Affiliation(s)
- Luke R Bonser
- Lung Biology Center, University of California San Francisco, San Francisco, CA, United States
| | - David J Erle
- Lung Biology Center, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
187
|
Schmidt H, Braubach P, Schilpp C, Lochbaum R, Neuland K, Thompson K, Jonigk D, Frick M, Dietl P, Wittekindt OH. IL-13 Impairs Tight Junctions in Airway Epithelia. Int J Mol Sci 2019; 20:ijms20133222. [PMID: 31262043 PMCID: PMC6651493 DOI: 10.3390/ijms20133222] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 06/27/2019] [Indexed: 12/12/2022] Open
Abstract
Interleukin-13 (IL-13) drives symptoms in asthma with high levels of T-helper type 2 cells (Th2-cells). Since tight junctions (TJ) constitute the epithelial diffusion barrier, we investigated the effect of IL-13 on TJ in human tracheal epithelial cells. We observed that IL-13 increases paracellular permeability, changes claudin expression pattern and induces intracellular aggregation of the TJ proteins zonlua occludens protein 1, as well as claudins. Furthermore, IL-13 treatment increases expression of ubiquitin conjugating E2 enzyme UBE2Z. Co-localization and proximity ligation assays further showed that ubiquitin and the proteasomal marker PSMA5 co-localize with TJ proteins in IL-13 treated cells, showing that TJ proteins are ubiquitinated following IL-13 exposure. UBE2Z upregulation occurs within the first day after IL-13 exposure. Proteasomal aggregation of ubiquitinated TJ proteins starts three days after IL-13 exposure and transepithelial electrical resistance (TEER) decrease follows the time course of TJ-protein aggregation. Inhibition of JAK/STAT signaling abolishes IL-13 induced effects. Our data suggest that that IL-13 induces ubiquitination and proteasomal aggregation of TJ proteins via JAK/STAT dependent expression of UBE2Z, resulting in opening of TJs. This may contribute to barrier disturbances in pulmonary epithelia and lung damage of patients with inflammatory lung diseases.
Collapse
Affiliation(s)
- Hanna Schmidt
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Peter Braubach
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 130625 Hannover, Germany
- German Center of Lung Research (DZL), Partnersite BREATH, 306245 Hannover, Germany
| | - Carolin Schilpp
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Robin Lochbaum
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Kathrin Neuland
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Kristin Thompson
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 130625 Hannover, Germany
- German Center of Lung Research (DZL), Partnersite BREATH, 306245 Hannover, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Paul Dietl
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Oliver H Wittekindt
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany.
| |
Collapse
|
188
|
Beng H, Su H, Wang S, Kuai Y, Hu J, Zhang R, Liu F, Tan W. Differential effects of inhaled R- and S-terbutaline in ovalbumin-induced asthmatic mice. Int Immunopharmacol 2019; 73:581-589. [PMID: 31234092 DOI: 10.1016/j.intimp.2019.04.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/05/2019] [Accepted: 04/17/2019] [Indexed: 12/15/2022]
Abstract
Inhaled terbutaline is commercially available β2-agonist which consists of equivalent amount of R- and S-enantiomer. In this study, we aimed to investigate the effects of single enantiomers of terbutaline and its racemate in an ovalbumin (OVA)-induced mouse model of asthma via. seven days inhalation and the potential mechanisms involved. In a standard experimental asthma model, BALB/c mice were sensitized and challenged with OVA. R-terbutaline (R-ter), S-terbutaline (S-ter) or racemic terbutaline (rac-ter) was given via. nose-only inhalation for one week. Airway responsiveness to methacholine was measured by the plethysmography in conscious mice. Eosinophils counts in blood and bronchoalveolar (BAL) fluid were determined. The OVA-sIgE in plasma and inflammatory cytokines and mediators in BAL fluid or lung tissue were analyzed by ELISA, qRT-PCR or western blotting. Airway inflammation and remodeling were evaluated with hematoxylin and eosin (HE), periodic acid-Schiff (PAS), and Masson staining. Drug distribution and deposition after inhalation were determined by LC-MS/MS. Our data showed that R-ter efficiently ameliorated asthma responses, including airway hyperresponsiveness, eosinophils influx and IL-5 in BALF, plasma OVA-sIgE and significantly reduced pulmonary inflammation, peribronchial smooth muscle layer thickness, goblet cell hyperplasia, and deposition of collagen fibers, as well as downregulation of p38 MAPK phosphorylation and NF-κB expression. Racemic mixture exhibited diminished effects while S-ter enhanced airway responsiveness to methacholine and exerted pro-asthmatic effects.
Collapse
Affiliation(s)
- Huimin Beng
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Hao Su
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Shanping Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yihe Kuai
- Institute of Biomedical & Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Junhua Hu
- Institute of Biomedical & Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Rui Zhang
- Institute of Biomedical & Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Fei Liu
- Institute of Biomedical & Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Wen Tan
- Institute of Biomedical & Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
189
|
Saeki M, Nishimura T, Kitamura N, Hiroi T, Mori A, Kaminuma O. Potential Mechanisms of T Cell-Mediated and Eosinophil-Independent Bronchial Hyperresponsiveness. Int J Mol Sci 2019; 20:ijms20122980. [PMID: 31216735 PMCID: PMC6627885 DOI: 10.3390/ijms20122980] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022] Open
Abstract
Bronchial asthma is a chronic disease characterized by reversible airway obstruction, mucus production, and bronchial hyperresponsiveness (BHR). Although Th2 cell-mediated eosinophilic inflammation is an important disease mechanism in the majority of patients with bronchial asthma, recent studies suggest the possible development of Th2-independent airway inflammation and BHR. These non-Th2 endotype patients seem to consist of multiple subgroups, and often do not respond to inhaled corticosteroids. Therefore, to understand the pathogenesis of asthma, it is important to characterize these non-Th2 subgroups. Recently, we demonstrated that Th9 cells induce eosinophil infiltration and eosinophil-independent BHR, and Th9 cells-mediated BHR may be resistant to glucocorticoid. In this review, we summarize the contribution of several T cell subsets in the development of bronchial asthma and introduce our recent study demonstrating Th9 cell-mediated and eosinophil-independent BHR.
Collapse
Affiliation(s)
- Mayumi Saeki
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan.
| | - Tomoe Nishimura
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan.
| | - Noriko Kitamura
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan.
| | - Takachika Hiroi
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan.
| | - Akio Mori
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan.
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization, Sagamihara National Hospital, Kanagawa 252-0392, Japan.
| | - Osamu Kaminuma
- Allergy and Immunology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan.
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization, Sagamihara National Hospital, Kanagawa 252-0392, Japan.
- Department of Disease Model, Research Institute of Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-0037, Japan.
- Center for Life Science Research, University of Yamanashi, Yamanashi 400-8510, Japan.
| |
Collapse
|
190
|
Giuffrida P, Caprioli F, Facciotti F, Di Sabatino A. The role of interleukin-13 in chronic inflammatory intestinal disorders. Autoimmun Rev 2019; 18:549-555. [DOI: 10.1016/j.autrev.2019.03.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 01/04/2019] [Indexed: 12/17/2022]
|
191
|
Ayakannu R, Abdullah NA, Radhakrishnan AK, Lechimi Raj V, Liam CK. Relationship between various cytokines implicated in asthma. Hum Immunol 2019; 80:755-763. [PMID: 31054782 DOI: 10.1016/j.humimm.2019.04.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 04/18/2019] [Accepted: 04/26/2019] [Indexed: 01/01/2023]
Abstract
Asthma is a complex disorder involving immunologic, environmental, genetic and other factors. Today, asthma is the most common disease encountered in clinical medicine in both children and adults worldwide. Asthma is characterized by increased responsiveness of the tracheobronchial tree resulting in chronic swelling and inflammation of the airways recognized to be controlled by the T-helper 2 (Th2) lymphocytes, which secrete cytokines to increase the production of IgE by B cells. There are many cytokines implicated in the development of the chronic inflammatory processes that are often observed in asthma. Ultimately, these cytokines cause the release of mediators such as histamine and leukotrienes (LT), which in turn promote airway remodeling, bronchial hyperresponsiveness and bronchoconstriction. The CD4+ T-lymphocytes from the airways of asthmatics express a panel of cytokines that represent the Th2 cells. The knowledge derived from numerous experimental and clinical studies have allowed physicians and scientists to understand the normal functions of these cytokines and their roles in the pathogenesis of asthma. The main focus of this review is to accentuate the relationship between various cytokines implicated in human asthma. However, some key findings from animal models will be highlighted to support the discoveries from clinical studies.
Collapse
Affiliation(s)
- Rathimalar Ayakannu
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - N A Abdullah
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Ammu K Radhakrishnan
- Jeffrey Cheah School of Medicine, Monash University Malaysia, Jalan Lagoon, 47500 Bandar Sunway, Selangor, Malaysia
| | - Vijaya Lechimi Raj
- Department of Pharmacology, Faculty of Medicine, MAHSA University, Bandar Saujana Putra, Selangor, Malaysia
| | - C K Liam
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
192
|
Adolescents with urinary stones have elevated urine levels of inflammatory mediators. Urolithiasis 2019; 47:461-466. [PMID: 30993354 DOI: 10.1007/s00240-019-01133-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/03/2019] [Indexed: 10/27/2022]
Abstract
Urinary stones are increasing in children, primarily during adolescence. Although urinary stones are often viewed in the context of intermittent stone events, increasing evidence indicates that stones are a metabolic process associated with chronic kidney disease and cardiovascular disease. These aforementioned stone-associated conditions may have pediatric origins. To compare urine inflammatory markers in otherwise healthy stone forming children versus matched controls. Urine samples were collected from 12 adolescents with urinary stones along with 15 controls. The levels of 30 urine cytokines were measured using a Mesoscale 30-Plex Human Cytokine panel and normalized to urine creatinine levels. Macrophage inflammatory protein 1β and interleukin 13 levels were significantly elevated in the urine of the stone forming adolescents compared to controls. Interleukin 17A was elevated in the urine of controls. This study indicates that urine levels of cytokines involved in chronic inflammation and fibrosis are elevated in urinary stone formers as early as adolescence. Because stone formers are at risk for chronic kidney disease, macrophage inflammatory protein 1β and interleukin 13 represent investigative targets.
Collapse
|
193
|
Lee SY, Bae CS, Seo NS, Na CS, Yoo HY, Oh DS, Bae MS, Kwon MS, Cho SS, Park DH. Camellia japonica oil suppressed asthma occurrence via GATA-3 & IL-4 pathway and its effective and major component is oleic acid. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 57:84-94. [PMID: 30668326 DOI: 10.1016/j.phymed.2018.12.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 11/03/2018] [Accepted: 12/09/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND In December 2016, WHO released a report stating that in 2015 there were 383,000 deaths caused by asthma and 235 million people suffering from asthma. As there are many adverse effects associated with the currently-used asthma drugs, new anti-asthmatic drugs need to be developed. PURPOSE In order to find new drug candidates with safe and low side effects, the anti-asthmatic function and mechanism of C. japonica oil were evaluated, and its active ingredients were analyzed for use in an ovalbumin asthma murine model. STUDY DESIGN AND METHODS The study consisted of six groups: control; ovalbumin group; and dexamethasone group as a positive control; and 10, 100, and 500 mg/kg C. japonica oil treatment groups. In order to measure the anti-asthmatic effect of C. japonica oil, WBC and differential cell count in BALF, IgE in serum, morphological changes in pulmonary system, and gene and protein levels such as IFN-γ, IL-12p40, IL-4, IL-5, IL-6, TNF-α, and IL-6 were all evaluated. RESULTS C. japonica oil had an anti-asthmatic effect and significantly controlled eosinophil in BALF, Th2-related factors such as GATA-3 that is Th2 cell transcription factor, IL-4, IL-5, and IL-13, and TNF-α in the lung. It also dose-dependently modulated inflammatory cells, T-bet, IL-12p40, and IL-6. Oleci acid was the major gradient (52.89%) in C. japonica oil and also had anti-asthmatic effects such as the downregulation of inflammatory cells, WBC, and eosinophil in BALF, IgE in serum, and morphological changes in the lung. CONCLUSION We concluded that C. japonica oil is a new anti-asthmatic drug candidate and that oleic acid is the major anti-asthmatic ingredient in C. japonica oil.
Collapse
Affiliation(s)
- Soon-Young Lee
- College of Oriental Medicine, Dongshin University, Naju, Jeonnam 58245, South Korea
| | - Chun-Sik Bae
- College of Veterinary Medicine, Chonnam National University, Gwangju 61186, South Korea
| | - Nam-Sook Seo
- College of Oriental Medicine, Dongshin University, Naju, Jeonnam 58245, South Korea
| | - Chang-Su Na
- College of Oriental Medicine, Dongshin University, Naju, Jeonnam 58245, South Korea
| | - Hah Young Yoo
- Department of Biotechnology, Sangmyung University, Seoul 03016, South Korea
| | - Deuk-Sil Oh
- Jeollanam-do Forest Resource Research Institute, Naju, Jeonnam 58213, South Korea
| | - Min-Suk Bae
- College of Engineering, Mokpo National University, Muan, Jeonnam 58554, South Korea
| | - Myung-Sang Kwon
- College of Veterinary Medicine, Kangwon National University, Chuncheon, Gangwon 24341, South Korea
| | - Seung-Sik Cho
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan, Jeonnam 58554, South Korea.
| | - Dae-Hun Park
- College of Oriental Medicine, Dongshin University, Naju, Jeonnam 58245, South Korea.
| |
Collapse
|
194
|
Perkins TN, Oczypok EA, Dutz RE, Donnell ML, Myerburg MM, Oury TD. The receptor for advanced glycation end products is a critical mediator of type 2 cytokine signaling in the lungs. J Allergy Clin Immunol 2019; 144:796-808.e12. [PMID: 30940519 DOI: 10.1016/j.jaci.2019.03.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 03/05/2019] [Accepted: 03/18/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND Asthma is estimated to effect more than 300 million persons worldwide, leading to nearly 250,000 deaths annually. The majority of patients with mild-to-severe asthma have what is deemed "type-2 high" asthma, which is driven by the prototypical type 2 cytokines IL-4, IL-5, and IL-13. Studies have indicated that the receptor for advanced glycation end products (RAGE) is a critical molecule in the pathogenesis of experimental asthma/allergic airway inflammation. More specifically, RAGE expressed on stromal cells, rather than hematopoietic cells, is critical to induction of asthma/allergic airway inflammation by driving type 2 inflammatory responses. However, the role of RAGE in directly mediating type 2 cytokine signaling has never been investigated. OBJECTIVE The goal of this study was to test the hypothesis that RAGE mediates type 2 cytokine-induced signal transduction, airway inflammation, and mucus metaplasia in the lungs. METHODS Wild-type (WT) and RAGE knockout (RAGE-/-) mice, were intranasally administered rIL-5/rIL-13 or rIL-4 alone, and signal transducer and activator of transcription 6 (STAT6) signaling, airway inflammation, and mucus metaplasia were assessed. A RAGE small-molecule antagonist was used to determine the effects of pharmacologically inhibiting RAGE on type 2 cytokine-induced effects. RESULTS Administration of type 2 cytokines induced pronounced airway inflammation and mucus metaplasia in WT mice, which was nearly completely abrogated in RAGE-/- mice. In addition, treatment with a RAGE-specific antagonist diminished the effects of type 2 cytokines in WT mice and in primary human bronchial epithelial cell cultures. Genetic ablation or pharmacologic inhibition of RAGE blocks the effects of IL-13 and IL-4 by inhibiting sustained STAT6 activation and downstream target gene expression in mice and in human bronchial epithelial cells. CONCLUSIONS This study is the first to indicate that RAGE is a critical component of type 2 cytokine signal transduction mechanisms, which is a driving force behind type 2-high asthma.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Pathology, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa; Department of Pediatrics, Division of Pulmonary, Allergy, and Clinical Immunology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pa.
| | - Elizabeth A Oczypok
- Department of Medicine, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Regina E Dutz
- Department of Pathology, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Mason L Donnell
- Department of Pathology, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Michael M Myerburg
- Department of Medicine, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh, School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pa.
| |
Collapse
|
195
|
Sveen LR, Timmerhaus G, Krasnov A, Takle H, Handeland S, Ytteborg E. Wound healing in post-smolt Atlantic salmon (Salmo salar L.). Sci Rep 2019; 9:3565. [PMID: 30837496 PMCID: PMC6400935 DOI: 10.1038/s41598-019-39080-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 01/15/2019] [Indexed: 12/17/2022] Open
Abstract
Skin biopsies (5 mm) taken from behind the dorsal fin on Atlantic salmon post-smolts were followed over a 2 month period. The healing process was dominated by hemostasis, acute inflammation, and epidermal repair the first 14 days post wounding (dpw), as shown through imaging, histological evaluation, and transcriptomics. Most of the immune genes showed decreased expression after two weeks, approaching the levels of intact skin, as also reflected in sections where reduced inflammation in the wound bed was observed. Transcriptional events suggest recruitment of lymphocytes to the wound site during the acute phase, with activation of humoral responses from 14 dpw and onward. From the histology, a more adherent mucus was observed that correlated with altered transcription of glycosyltransferases. This may indicate different properties and functions of the mucus during the wound healing process. Wound contraction started between 14 and 36 dpw. The occurrence of these events was concurrent with granulation tissue formation, melanocyte migration and up-regulation of genes involved in extracellular matrix formation. The presented description of the wound healing processes in Atlantic salmon gives insight into comparative ulcerative biology in mammals and fish and provides both novel and updated knowledge that can be applied for improved best operational practices for fish welfare in aquaculture.
Collapse
Affiliation(s)
- Lene Rydal Sveen
- University of Bergen, Postboks 7800, 5020, Bergen, Norway. .,Nofima, Osloveien 1, 1430, Ås, Norway.
| | | | | | - Harald Takle
- Cermaq Group AS, Dronning Eufemias gate 16,0102, Oslo, Norway
| | | | | |
Collapse
|
196
|
Fahy N, Menzel U, Alini M, Stoddart MJ. Shear and Dynamic Compression Modulates the Inflammatory Phenotype of Human Monocytes in vitro. Front Immunol 2019; 10:383. [PMID: 30891042 PMCID: PMC6411641 DOI: 10.3389/fimmu.2019.00383] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/14/2019] [Indexed: 11/13/2022] Open
Abstract
Monocytes and their derived macrophages are found at the site of remodeling tissue, such as fracture hematoma, that is exposed to mechanical forces and have been previously implicated in the reparative response. However, the mechanoresponsive of monocytes and macrophages to skeletal tissue-associated mechanical forces and their subsequent contribution to skeletal repair remains unclear. The aim of this study was to investigate the potential of skeletal tissue-associated loading conditions to modulate human monocyte activation and phenotype. Primary human monocytes or the human monocyte reporter cell line, THP1-Blue, were encapsulated in agarose and exposed to a combination of shear and compressive loading for 1 h a day for 3 consecutive days. Exposure of monocytes to mechanical loading conditions increased their pro-inflammatory gene and protein expression. Exposure of undifferentiated monocytes to mechanical loading conditions significantly upregulated gene expression levels of interleukin(IL)-6 and IL-8 compared to free swelling controls. Additionally, multiaxial loading of unstimulated monocytes resulted in increased protein secretion of TNF-α (17.1 ± 8.9 vs. 8 ± 7.4 pg/ml) and MIP-1α (636.8 ± 471.1 vs. 124.1 ± 40.1 pg/ml), as well as IL-13 (42.1 ± 19.8 vs. 21.7 ± 13.6) compared monocytes cultured under free-swelling conditions. This modulatory effect was observed irrespective of previous activation with the M1/pro-inflammatory differentiation stimuli lipopolysaccharide and interferon-γ or the M2/anti-inflammatory differentiation factor interleukin-4. Furthermore, mechanical shear and compression were found to differentially regulate nitric oxide synthase 2 (NOS2) and IL-12B gene expression as well as inflammatory protein production by THP1-Blue monocytes. The findings of this study indicate that human monocytes are responsive to mechanical stimuli, with a modulatory effect of shear and compressive loading observed toward pro-inflammatory mediator production. This may play a role in healing pathways that are mechanically regulated. An in depth understanding of the impact of skeletal tissue-associated mechanical loading on monocyte behavior may identify novel targets to maximize inflammation-mediated repair mechanisms.
Collapse
Affiliation(s)
- Niamh Fahy
- AO Research Institute Davos, Davos, Switzerland
| | | | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | | |
Collapse
|
197
|
Bui TT, Kwon DA, Choi DW, Jung SY, Lee SY, Piao CH, Hyeon E, Fan Y, Yeon SH, Son RH, Shon DH, Song CH, Shin HS, Chai OH. Rosae multiflorae fructus extract and its four active components alleviate ovalbumin-induced allergic inflammatory responses via regulation of Th1/Th2 imbalance in BALB/c rhinitis mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 55:238-248. [PMID: 30668435 DOI: 10.1016/j.phymed.2018.06.044] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 05/29/2018] [Accepted: 06/19/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Rosae Multiflorae fructus has potent antioxidative, analgesic, and anti-inflammatory properties. PURPOSE We investigated the immunomodulatory effect of Rosae Multiflorae fructus extract (RMFE) on allergic inflammation in an allergic rhinitis (AR) mouse model. METHODS Mice were sensitized and intranasally challenged with ovalbumin (OVA), the Th1/Th2-related cytokines and histopathology were examinated after RMFE treatments. Primary cell culture from spleen and NALT was performed to evaluate RMFE effect on Th1/Th2 responses. Four active components of RMFE were determined using HPLC and then tested the inhibition on Th2 response. RESULTS Oral administration of RMFE inhibited the accumulation of eosinophils in nasal lavage fluid (NALF) and the nasal mucosa, goblet cells in the nasal epithelium, and mast cells in the respiratory region of the nasal cavity. Thus, the swelling of the nasal epithelium, nasal-associated lymphoid tissue (NALT), and lung tissue were ameliorated. Furthermore, the RMFE suppressed Th2-related cytokines, such as IL-4, IL-5, and IL-13 in NALF, NALT, and splenocytes, whereas the Th1-associated cytokine IL-12 was up-regulated by RMFE. We also revealed the active components of RMFE, such as ellagic acid, hyperoside, isoquercitrin, and miquelianin. They may inhibit IL-4 secretion in allergic responses. CONCLUSION RMFE may have therapeutic potential for treating AR by modulating the relationships between Th1/Th2 responses.
Collapse
Affiliation(s)
- Thi Tho Bui
- Department of Anatomy, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea; Faculty of Biology & Environmental Science, University of Education, The University of Danang, Danang 555940, Vietnam
| | - Da-Ae Kwon
- Research Division of Food Functionality, Korea Food Research Institute, Wanju-gun, 55365, Republic of Korea
| | - Dae Woon Choi
- Food Biotechnology Program, Korea University of Science and Technology, Daejeon 305-350, Republic of Korea
| | - Sun Young Jung
- Food Biotechnology Program, Korea University of Science and Technology, Daejeon 305-350, Republic of Korea
| | - So-Young Lee
- Research Division of Food Functionality, Korea Food Research Institute, Wanju-gun, 55365, Republic of Korea.; Food Biotechnology Program, Korea University of Science and Technology, Daejeon 305-350, Republic of Korea
| | - Chun Hua Piao
- Department of Anatomy, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Eunjin Hyeon
- Department of Anatomy, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Yanjing Fan
- Department of Anatomy, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Sung Hum Yeon
- R&D Center, Huons. Co. Ltd., College of Pharmacy, Hanyang University, Sangnok-gu, Ansan-si, Kyeonggi-do 15588, Republic of Korea
| | - Rak-Ho Son
- R&D Center, Huons. Co. Ltd., College of Pharmacy, Hanyang University, Sangnok-gu, Ansan-si, Kyeonggi-do 15588, Republic of Korea
| | - Dong-Hwa Shon
- Research Division of Food Functionality, Korea Food Research Institute, Wanju-gun, 55365, Republic of Korea
| | - Chang Ho Song
- Department of Anatomy, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea; Institute for Medical Sciences, Chonbuk National University, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Hee Soon Shin
- Research Division of Food Functionality, Korea Food Research Institute, Wanju-gun, 55365, Republic of Korea.; Food Biotechnology Program, Korea University of Science and Technology, Daejeon 305-350, Republic of Korea..
| | - Ok Hee Chai
- Department of Anatomy, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea; Institute for Medical Sciences, Chonbuk National University, Jeonju, Jeonbuk 54896, Republic of Korea..
| |
Collapse
|
198
|
Alagha K, Bourdin A, Vernisse C, Garulli C, Tummino C, Charriot J, Vachier I, Suehs C, Chanez P, Gras D. Goblet cell hyperplasia as a feature of neutrophilic asthma. Clin Exp Allergy 2019; 49:781-788. [PMID: 30710420 DOI: 10.1111/cea.13359] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Goblet cell hyperplasia (GCH) is a pathological finding classically reported across asthma severity levels and usually associated with smoking. Multiple biological mechanisms may contribute to excessive mucus production. OBJECTIVE We aimed to decipher the clinical meanings and biological pathways related to GCH in non-smokers with asthma. METHODS Cough and sputum assessment questionnaire (CASA-Q) responses at entry and 1 year later were compared to clinical and functional outcomes in 59 asthmatic patients. GCH was assessed through periodic-acid shift (PAS) staining on endobronchial biopsies obtained at entry in a subset of 32 patients. RESULTS Periodic-acid shift-staining analysis revealed a double wave distribution discriminating patients with (>10% of the epithelial area) or without GCH. CASA-Q scores were mostly driven by overall asthma severity (P < 0.0001). CASA-Q scores remained stable at 1 year and were independently associated with BAL eosinophil content irrespective of the presence of GCH. GCH was unrelated to the presence of bronchiectasis at CT, GERD or chronic rhinosinusitis, but correlated well with neutrophilic inflammatory patterns observed upon BAL cellular analysis (P = 0.002 at multivariate analysis). BALF bacterial loads were unrelated to GCH or to CASA-Q. CONCLUSIONS AND CLINICAL RELEVANCE Goblet cell hyperplasia is disconnected from chronic cough and sputum when assessed by a specific questionnaire. GCH is related to neutrophilic asthma whereas symptoms are related to airway eosinophilia. The clinical counterpart of GCH is unlikely assessed by the CASA-Q.
Collapse
Affiliation(s)
- Khuder Alagha
- Département de Pneumologie et Addictologie, University of Montpellier, Montpellier, France.,Aix Marseille University, INSERM, INRA, C2VN, Marseille, France
| | - Arnaud Bourdin
- Département de Pneumologie et Addictologie, University of Montpellier, Montpellier, France.,PhyMedExp, Hôpital Arnaud de Villeneuve, INSERM U1046, CNRS, UMR 9214, University of Montpellier, Montpellier, France.,CHU Montpellier, Montpellier, France
| | - Charlotte Vernisse
- PhyMedExp, Hôpital Arnaud de Villeneuve, INSERM U1046, CNRS, UMR 9214, University of Montpellier, Montpellier, France.,CHU Montpellier, Montpellier, France
| | - Céline Garulli
- Aix Marseille University, INSERM, INRA, C2VN, Marseille, France
| | - Céline Tummino
- Clinique des Bronches, Allergies et Sommeil, Hôpital Nord, AP-HM, Aix Marseille Université Marseille, Marseille, France
| | - Jérémy Charriot
- Département de Pneumologie et Addictologie, University of Montpellier, Montpellier, France
| | - Isabelle Vachier
- Département de Pneumologie et Addictologie, University of Montpellier, Montpellier, France
| | - Carey Suehs
- Département de Pneumologie et Addictologie, University of Montpellier, Montpellier, France
| | - Pascal Chanez
- Aix Marseille University, INSERM, INRA, C2VN, Marseille, France.,Clinique des Bronches, Allergies et Sommeil, Hôpital Nord, AP-HM, Aix Marseille Université Marseille, Marseille, France
| | - Delphine Gras
- Aix Marseille University, INSERM, INRA, C2VN, Marseille, France
| |
Collapse
|
199
|
Gurram RK, Zhu J. Orchestration between ILC2s and Th2 cells in shaping type 2 immune responses. Cell Mol Immunol 2019; 16:225-235. [PMID: 30792500 DOI: 10.1038/s41423-019-0210-8] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/31/2019] [Indexed: 01/06/2023] Open
Abstract
The type 2 immune response is critical for host defense against large parasites such as helminths. On the other hand, dysregulation of the type 2 immune response may cause immunopathological conditions, including asthma, atopic dermatitis, rhinitis, and anaphylaxis. Thus, a balanced type 2 immune response must be achieved to mount effective protection against invading pathogens while avoiding immunopathology. The classical model of type 2 immunity mainly involves the differentiation of type 2 T helper (Th2) cells and the production of distinct type 2 cytokines, including interleukin-4 (IL-4), IL-5, and IL-13. Group 2 innate lymphoid cells (ILC2s) were recently recognized as another important source of type 2 cytokines. Although eosinophils, mast cells, and basophils can also express type 2 cytokines and participate in type 2 immune responses to various degrees, the production of type 2 cytokines by the lymphoid lineages, Th2 cells, and ILC2s in particular is the central event during the type 2 immune response. In this review, we discuss recent advances in our understanding of how ILC2s and Th2 cells orchestrate type 2 immune responses through direct and indirect interactions.
Collapse
Affiliation(s)
- Rama Krishna Gurram
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
200
|
He M, Ichinose T, Yoshida S, Nishikawa M, Sun G, Shibamoto T. Role of iron and oxidative stress in the exacerbation of allergic inflammation in murine lungs caused by urban particulate matter <2.5 μm and desert dust. J Appl Toxicol 2019; 39:855-867. [PMID: 30698282 DOI: 10.1002/jat.3773] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 12/19/2018] [Indexed: 01/13/2023]
Abstract
Simultaneous exposure of lipopolysaccharide (LPS) and urban particulate matter <2.5 μm (PM2.5) or desert dust exacerbated murine asthma. In the present study, the role of iron (Fe) contained in particles and oxidative stress was investigated using Fe chelator deferoxamine (DFO) and oxidative stress scavenger N-acetylcysteine (NAC) in a murine asthma model exacerbated by LPS + PM2.5 or LPS + Asian sand dust (ASD). When BALB/c mice were intratracheally challenged with ovalbumin (OVA) + LPS and either urban PM2.5 or ASD, LPS + PM2.5 and LPS + ASD caused exacerbation of OVA-induced lung eosinophilia along with T-helper 2 cytokine and eosinophil-relevant chemokine production in bronchoalveolar lavage fluid as well as the production of OVA-specific IgE in serum. LPS + PM2.5 with NAC tended to reduce the lung eosinophilia compared to the LPS + PM2.5 host, whereas LPS + PM2.5 with DFO did not reduce them. LPS + ASD with NAC moderately reduced the lung eosinophilia compared to the LPS + ASD host. LPS + ASD with DFO drastically reduced the lung eosinophilia compared to the LPS + ASD host. The concentration of Fe in urban PM2.5 and ASD were almost the same. However, the concentrations of trace metals Pb, Cu, As, Ni, Cr, Mo, Sb, Co, Se and Cd were greater in PM2.5 than in ASD. These results suggested that Fe and oxidative stress are at least partly involved in lung eosinophilia exacerbation caused by LPS + ASD. However, trace metals (except Fe) might also be involved in lung eosinophilia exacerbated by LPS + PM2.5.
Collapse
Affiliation(s)
- Miao He
- Key Laboratory of Environmental Health Damage Research and Assessment, Liaoning Province; Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Takamichi Ichinose
- Department of Health Sciences, Oita University of Nursing and Health Sciences, Oita, 870-1201, Japan
| | - Seiichi Yoshida
- Department of Health Sciences, Oita University of Nursing and Health Sciences, Oita, 870-1201, Japan
| | - Masataka Nishikawa
- Environmental Chemistry Division, National Institute for Environmental Studies, Ibaraki, 305-8506, Japan
| | - Guifan Sun
- Key Laboratory of Environmental Health Damage Research and Assessment, Liaoning Province; Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, China
| | - Takayuki Shibamoto
- Department of Environmental Toxicology, University of California, Davis, CA, 95616, USA
| |
Collapse
|