101
|
Mechanistic Implications of Biomass-Derived Particulate Matter for Immunity and Immune Disorders. TOXICS 2021; 9:toxics9020018. [PMID: 33498426 PMCID: PMC7909393 DOI: 10.3390/toxics9020018] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/04/2021] [Accepted: 01/15/2021] [Indexed: 12/29/2022]
Abstract
Particulate matter (PM) is a major and the most harmful component of urban air pollution, which may adversely affect human health. PM exposure has been associated with several human diseases, notably respiratory and cardiovascular diseases. In particular, recent evidence suggests that exposure to biomass-derived PM associates with airway inflammation and can aggravate asthma and other allergic diseases. Defective or excess responsiveness in the immune system regulates distinct pathologies, such as infections, hypersensitivity, and malignancies. Therefore, PM-induced modulation of the immune system is crucial for understanding how it causes these diseases and highlighting key molecular mechanisms that can mitigate the underlying pathologies. Emerging evidence has revealed that immune responses to biomass-derived PM exposure are closely associated with the risk of diverse hypersensitivity disorders, including asthma, allergic rhinitis, atopic dermatitis, and allergen sensitization. Moreover, immunological alteration by PM accounts for increased susceptibility to infectious diseases, such as tuberculosis and coronavirus disease-2019 (COVID-19). Evidence-based understanding of the immunological effects of PM and the molecular machinery would provide novel insights into clinical interventions or prevention against acute and chronic environmental disorders induced by biomass-derived PM.
Collapse
|
102
|
Shenderov K, Collins SL, Powell JD, Horton MR. Immune dysregulation as a driver of idiopathic pulmonary fibrosis. J Clin Invest 2021; 131:143226. [PMID: 33463535 DOI: 10.1172/jci143226] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) affects hundreds of thousands of people worldwide, reducing their quality of life and leading to death from respiratory failure within years of diagnosis. Treatment options remain limited, with only two FDA-approved drugs available in the United States, neither of which reverse the lung damage caused by the disease or prolong the life of individuals with IPF. The only cure for IPF is lung transplantation. In this review, we discuss recent major advances in our understanding of the role of the immune system in IPF that have revealed immune dysregulation as a critical driver of disease pathophysiology. We also highlight ways in which an improved understanding of the immune system's role in IPF may enable the development of targeted immunomodulatory therapies that successfully halt or potentially even reverse lung fibrosis.
Collapse
Affiliation(s)
- Kevin Shenderov
- Department of Medicine, Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Samuel L Collins
- Department of Medicine, Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jonathan D Powell
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Maureen R Horton
- Department of Medicine, Division of Pulmonary and Critical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
103
|
Mo Y, Bae B, Kim J, Kim RL, Son K, Kang MJ, Lee CG, Cho SH, Kang HR. Therapeutic effect of atorvastatin on interleukin-13-induced lung pathology. ALLERGY ASTHMA & RESPIRATORY DISEASE 2021. [DOI: 10.4168/aard.2021.9.2.76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Yosep Mo
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Boram Bae
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Junghyun Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Ruth Lee Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Kyunghee Son
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Min-Jong Kang
- Pulmonary, Critical Care and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, USA
| | - Chun-Gen Lee
- Department of Molecular Microbiology and Immunology, Division of Biology and Medical Sciences, Brown University, Providence, RI, USA
| | - Sang-Heon Cho
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hye-Ryun Kang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
104
|
Darling NJ, Arthur JSC, Cohen P. Salt-inducible kinases are required for the IL-33-dependent secretion of cytokines and chemokines in mast cells. J Biol Chem 2021; 296:100428. [PMID: 33600797 PMCID: PMC7988334 DOI: 10.1016/j.jbc.2021.100428] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/28/2021] [Accepted: 02/12/2021] [Indexed: 02/07/2023] Open
Abstract
Cytokines and chemokines are important regulators of airway hyper-responsiveness, immune cell infiltration, and inflammation and are produced when mast cells are stimulated with interleukin-33 (IL-33). Here, we establish that the salt-inducible kinases (SIKs) are required for the IL-33-stimulated transcription of il13, gm-csf and tnf and hence the production of these cytokines. The IL-33-stimulated secretion of IL-13, granulocyte-macrophage colony stimulating factor, and tumor necrosis factor was strongly reduced in fetal liver-derived mast cells from mice expressing a kinase-inactive mutant of SIK3 and abolished in cells expressing kinase-inactive mutants of SIK2 and SIK3. The IL-33-dependent secretion of these cytokines and several chemokines was also abolished in SIK2/3 double knock-out bone marrow-derived mast cells (BMMC), reduced in SIK3 KO cells but little affected in BMMC expressing kinase-inactive mutants of SIK1 and SIK2 or lacking SIK2 expression. In SIK2 knock-out BMMC, the expression of SIK3 was greatly increased. Our studies identify essential roles for SIK2 and SIK3 in producing inflammatory mediators that trigger airway inflammation. The effects of SIKs were independent of IκB kinase β, IκB kinase β-mediated NF-κB-dependent gene transcription, and activation of the mitogen-activated protein kinase family members p38α and c-jun N-terminal kinases. Our results suggest that dual inhibitors of SIK2 and SIK3 may have therapeutic potential for the treatment of mast cell-driven diseases.
Collapse
Affiliation(s)
- Nicola J Darling
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, Angus, UK
| | - J Simon C Arthur
- Division of Cell Signalling and Immunology, University of Dundee, Dundee, Angus, UK
| | - Philip Cohen
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, Angus, UK.
| |
Collapse
|
105
|
IGF-1 Receptor Signaling Regulates Type II Pneumocyte Senescence and Resulting Macrophage Polarization in Lung Fibrosis. Int J Radiat Oncol Biol Phys 2020; 110:526-538. [PMID: 33385497 DOI: 10.1016/j.ijrobp.2020.12.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 11/22/2022]
Abstract
PURPOSE Type II pneumocyte (alveolar epithelial cells type II [AECII]) senescence has been implicated in the progression of lung fibrosis. The capacity of senescent cells to modulate pulmonary macrophages to drive fibrosis is unexplored. Insulin-like growth factor-1 receptor (IGF-1R) signaling has been implicated as a regulator of senescence and aging. METHODS AND MATERIALS Mice with an AECII-specific deletion of IGF-1R received thoracic irradiation (n ≥ 5 per condition), and the effect of IGF-1R deficiency on radiation-induced AECII senescence and macrophage polarization to an alternatively activated phenotype (M2) was investigated. IGF-1R signaling, macrophage polarization, and senescence were evaluated in surgically resected human lung (n = 63). RESULTS IGF-1R deficient mice demonstrated reduced AECII senescence (senescent AECII/field; intact: 7.25% ± 3.5% [mean ± SD], deficient: 2.75% ± 2.8%, P = .0001), reduced accumulation of M2 macrophages (intact: 24.7 ± 2.2 cells/field, deficient: 15.5 ± 1.2 cells/field, P = .0086), and fibrosis (hydroxyproline content; intact: 71.9 ± 21.7 μg/lung, deficient: 31.7 ± 7.9, P = .0485) after irradiation. Senescent AECII enhanced M2 polarization in a paracrine fashion (relative Arg1 mRNA, 0 Gy: 1.0 ± 0.4, 17.5 Gy: 7.34 ± 0.5, P < .0001). Evaluation of surgical samples from patients treated with chemoradiation demonstrated increased expression of IGF-1 (unirradiated: 10.2% ± 4.9% area, irradiated: 15.1% ± 11.5%, P = .0377), p21 (unirradiated: 0.013 ± 0.02 histoscore, irradiated: 0.084 ± 0.09 histoscore, P = .0002), IL-13 (unirradiated: 13.7% ± 2.8% area, irradiated: 21.7% ± 3.8%, P < .0001), and M2 macrophages in fibrotic regions relative to nonfibrotic regions (unirradiated: 11.4 ± 12.2 CD163 + cells/core, irradiated: 43.1 ± 40.9 cells/core, P = .0011), consistent with findings from animal models of lung fibrosis. CONCLUSIONS This study demonstrates that senescent AECII are necessary for the progression of pulmonary fibrosis and serve as a targetable, chronic stimuli for macrophage activation in fibrotic lung.
Collapse
|
106
|
Sohn KH, Baek MG, Choi SM, Bae B, Kim RY, Kim YC, Kim HY, Yi H, Kang HR. Alteration of Lung and Gut Microbiota in IL-13-Transgenic Mice Simulating Chronic Asthma. J Microbiol Biotechnol 2020; 30:1819-1826. [PMID: 33046682 PMCID: PMC9728179 DOI: 10.4014/jmb.2009.09019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022]
Abstract
Increasing evidence suggests a potential role of microbial colonization in the inception of chronic airway diseases. However, it is not clear whether the lung and gut microbiome dysbiosis is coincidental or a result of mutual interaction. In this study, we investigated the airway microbiome in interleukin 13 (IL-13)-rich lung environment and related alterations of the gut microbiome. IL-13- overexpressing transgenic (TG) mice presented enhanced eosinophilic inflammatory responses and mucus production, together with airway hyperresponsiveness and subepithelial fibrosis. While bronchoalveolar lavage fluid and cecum samples obtained from 10-week-old IL-13 TG mice and their C57BL/6 wild-type (WT) littermates showed no significant differences in alpha diversity of lung and gut microbiome, they presented altered beta diversity in both lung and gut microbiota in the IL-13 TG mice compared to the WT mice. Lung-specific IL-13 overexpression also altered the composition of the gut as well as the lung microbiome. In particular, IL-13 TG mice showed an increased proportion of Proteobacteria and Cyanobacteria and a decreased amount of Bacteroidetes in the lungs, and depletion of Firmicutes and Proteobacteria in the gut. The patterns of polymicrobial interaction within the lung microbiota were different between WT and IL-13 TG mice. For instance, in IL-13 TG mice, lung Mesorhizobium significantly affected the alpha diversity of both lung and gut microbiomes. In summary, chronic asthma-like pathologic changes can alter the lung microbiota and affect the gut microbiome. These findings suggest that the lung-gut microbial axis might actually work in asthma.
Collapse
Affiliation(s)
- Kyoung-Hee Sohn
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea,Division of Pulmonology, Allergy and Critical Care, Department of Internal Medicine, Kyung Hee University Medical Center, Seoul 0447, Republic of Korea
| | - Min-gyung Baek
- Department of Public Health Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Sung-Mi Choi
- Department of Public Health Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Boram Bae
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea
| | - Ruth Yuldam Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea
| | - Young-Chan Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea
| | - Hye-Young Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea,Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 08826, Republic of Korea
| | - Hana Yi
- Department of Public Health Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea,School of Biosystems and Biomedical Sciences, Korea University, Seoul 02841, Republic of Korea,Corresponding authors H.Yi Phone: +82-2-3290-5644 Fax: +82-2-940-2849 E-mail:
| | - Hye-Ryun Kang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea,Division of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul 0882, Republic of Korea,H-R.Kang Phone: 82-2-2072-0820 Fax: 82-2-742-3291 E-mail:
| |
Collapse
|
107
|
Jiang L, Li Y, Wang F, Zhang X, Zhao R. Protective Effect of S-Allyl Cysteine Against Neonatal Asthmatic Rats. Dose Response 2020; 18:1559325820982189. [PMID: 33488313 PMCID: PMC7768841 DOI: 10.1177/1559325820982189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/16/2020] [Accepted: 11/26/2020] [Indexed: 11/22/2022] Open
Abstract
S-Allyl cysteine (SAC), an organic compound and a natural constituent of Allium sativum, commonly known as garlic have been consumed in routine foods are known to possess various biological activities. Nevertheless, scientific evidence on the protective effect of SAC against neonatal asthmatic rats is not available. Hence, the present study aimed at investigating the anti-asthmatic activity of SAC in neonatal asthmatic rats using Wistar rats. The study conducted in 4 groups consists of normal control rats, asthma-induced, asthma animals administered with SAC (25 mg/kg), and SAC control. At the end of the experimental period, inflammatory cells in bronchoalveolar lavage fluid (BALF), inflammatory markers, fibrinogen level, activated partial thromboplastin time, coagulation factor activity, and histopathology were elucidated. The current investigation exhibits that SAC significantly reduced the total leukocytes, with restored fibrinogen level, and activated partial thromboplastin time. In addition, the levels of inflammatory cytokines such as TNF-α (tumor necrosis factor- α), IL-6 (Interleukin 6), and IL-1β have also attenuated in SAC treated animals. Furthermore, the mRNA expression levels of COX2 (cyclooxygenase-2), MCP-1 (monocyte chemoattractant protein-1), RANTES (regulated upon activation, normal T cell expressed and secreted), and eotaxin were reduced in SAC treated animals. Treatment of rats with SAC significantly reduced inflammation and eosinophil infiltration in the lungs. These results suggest that SAC exert protection in neonatal asthmatic rats suffering from acute or chronic inflammation by inducing anti-inflammatory and cell-protective responses.
Collapse
Affiliation(s)
- Li Jiang
- Department of Pediatrics, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuning Li
- Department of Pediatrics, The First Hospital of Lanzhou University, Lanzhou, China
| | - Fang Wang
- Department of Pediatrics, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xindao Zhang
- Department of Pediatrics, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ruiping Zhao
- Department of Pediatrics, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
108
|
Chatzileontiadou DSM, Sloane H, Nguyen AT, Gras S, Grant EJ. The Many Faces of CD4 + T Cells: Immunological and Structural Characteristics. Int J Mol Sci 2020; 22:E73. [PMID: 33374787 PMCID: PMC7796221 DOI: 10.3390/ijms22010073] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 12/14/2022] Open
Abstract
As a major arm of the cellular immune response, CD4+ T cells are important in the control and clearance of infections. Primarily described as helpers, CD4+ T cells play an integral role in the development and activation of B cells and CD8+ T cells. CD4+ T cells are incredibly heterogeneous, and can be divided into six main lineages based on distinct profiles, namely T helper 1, 2, 17 and 22 (Th1, Th2, Th17, Th22), regulatory T cells (Treg) and T follicular helper cells (Tfh). Recent advances in structural biology have allowed for a detailed characterisation of the molecular mechanisms that drive CD4+ T cell recognition. In this review, we discuss the defining features of the main human CD4+ T cell lineages and their role in immunity, as well as their structural characteristics underlying their detection of pathogens.
Collapse
Affiliation(s)
- Demetra S. M. Chatzileontiadou
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (D.S.M.C.); (H.S.); (A.T.N.); (S.G.)
| | - Hannah Sloane
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (D.S.M.C.); (H.S.); (A.T.N.); (S.G.)
| | - Andrea T. Nguyen
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (D.S.M.C.); (H.S.); (A.T.N.); (S.G.)
| | - Stephanie Gras
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (D.S.M.C.); (H.S.); (A.T.N.); (S.G.)
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, VIC 3800, Australia
| | - Emma J. Grant
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (D.S.M.C.); (H.S.); (A.T.N.); (S.G.)
| |
Collapse
|
109
|
Tereschenko SY, Smolnikova MV, Kasparov EV, Shakhtshneider EV, Malinchik MA, Konopleva OS, Smirnova SV. Role of IL13 genetic polymorphism in the development of bronchial asthma in children. MEDICAL IMMUNOLOGY (RUSSIA) 2020. [DOI: 10.15789/1563-0625-roi-1986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Bronchial asthma is a multifactorial disease, with both environmental factors and genetic predisposal affecting its development. A number of gene associations have been obtained between polymorphisms of cytokine genes produced by different types of immune cells and asthma development. Interleukin-13 is involved in allergic inflammation, increased bronchial hypersensitivity, regulation of eosinophil levels and IgE production by B cells, thus making it promising for studying IL13 gene polymorphisms in bronchial asthma coupled to development of the disease. The aim of this study was to investigate possible association between asthma and IL13 rs1800925 polymorphism in the children of Caucasian origin in Eastern Siberia. Four groups of patients with asthma were examined (mean age 12.8±1.2 years): with a controlled (n = 95) and uncontrolled course (n = 107), with severe (n = 71) and moderate severity (n = 131) diseases. The control group consisted of healthy individuals: children (n = 33) and adults (n = 102). DNA was isolated with sorbent method; genotyping was carried out using RT-PCR using specific oligonucleotide primers and fluorescent TaqMan probes. The allele and genotype frequencies were compared by the χ-square test using an online calculator. The odds ratio (OR) with a 95% confidence interval (CI) was performed to link genetic markers with pathological phenotypes. The CT IL13 rs1800925 genotype was shown to be associated with moderate asthma and cases of uncontrollable clinical course, whereas the TT genotype was associated with severe asthma. Thus, rs1800925 polymorphism of IL13 gene (the T* variant is known to be associated with increased IL-13 expression) may be associated with bronchial asthma in children. Our data are consistent with results of other authors. E.g., Liu Z. et al. revealed an association between rs1800925 IL13 and the risk of developing asthma in children, with CT and TT genotypes being more common in the patient group. Radhakrishnan A. et al., was studied rs1800925 IL13 in adult population of Malaysia and found that the T* allele frequency in the group of patients significantly exceeds the frequency of this allele in the control group. Thus, the results of our study showed that IL13 rs1800925 polymorphism is associated with bronchial asthma in children, especially, with level of its control and severity of the disease.
Collapse
Affiliation(s)
- S. Yu. Tereschenko
- Research Institute of Medical Problems of the North, Krasnoyarsk Scientific Center, Siberian Branch, Russian Academy of Sciences, Krasnoyarsk
| | - M. V. Smolnikova
- Research Institute of Medical Problems of the North, Krasnoyarsk Scientific Center, Siberian Branch, Russian Academy of Sciences, Krasnoyarsk
| | - E. V. Kasparov
- Research Institute of Medical Problems of the North, Krasnoyarsk Scientific Center, Siberian Branch, Russian Academy of Sciences, Krasnoyarsk
| | - E. V. Shakhtshneider
- Institute of Internal and Preventive Medicine, Branch of Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences
| | - M. A. Malinchik
- Research Institute of Medical Problems of the North, Krasnoyarsk Scientific Center, Siberian Branch, Russian Academy of Sciences, Krasnoyarsk
| | - O. S. Konopleva
- Research Institute of Medical Problems of the North, Krasnoyarsk Scientific Center, Siberian Branch, Russian Academy of Sciences, Krasnoyarsk; Krasnoyarsk State V. Voino-Yasenetsky Medical University
| | - S. V. Smirnova
- Research Institute of Medical Problems of the North, Krasnoyarsk Scientific Center, Siberian Branch, Russian Academy of Sciences, Krasnoyarsk
| |
Collapse
|
110
|
He L, Feng QQ, Zhang Q, Zhang B, Wu SS, Gong JH. Protective role of overexpressed MUC5AC against fibrosis in MHV-68-induced combined pulmonary fibrosis and emphysema mouse model. J Med Virol 2020; 92:3726-3735. [PMID: 32557739 DOI: 10.1002/jmv.26094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/19/2020] [Accepted: 05/28/2020] [Indexed: 11/05/2022]
Abstract
Mucins have long been regarded to play a role as a barrier to prevent mucosal infections; however, some studies report that overexpression of mucins induces obstruction and inflammation of airways. We investigated whether the secretion of overexpressed mucin, mucin5ac (MUC5AC), could improve protection against pathogens. To examine the possible roles of mucin hypersecretion in augmenting host defense against disease-promoting muco-obstructive lung disease, a mouse model that overexpressed MUC5AC was generated. We had previously proved that murine gammaherpesvirus-68 (MHV-68) infection could induce emphysema in mice, which later developed into combined pulmonary fibrosis and emphysema (CPFE). We further explored whether increased MUC5AC secretion could provide benefits against MHV-68 induced fibrosis. We initially developed a pcDNA3.1-MUC5AC mouse model. Next, the experimental mice were randomly divided into five groups: normal control, pcDNA3.1 control, pcDNA3.1-MUC5AC, CPFE, and pcDNA3.1- MUC5AC + CPFE. Morphometric analysis of each group was performed by hematoxylin and eosin staining and Masson trichrome staining. MUC5AC levels in lung tissues were analyzed by immunohistochemical staining, real-time polymerase chain reaction, and Western blot analysis. The airway inflammation was determined by differential cell counts of bronchoalveolar lavage fluid (BALF) and measurement of cytokines and chemokines in BALF by enzyme-linked immunosorbent assay. MUC5AC hypersecretion alone was not sufficient to drive goblet cell metaplasia to induce obvious mucus plugging and airway inflammation. However, MUC5AC overexpression served as a protective barrier against MHV-68 virus infection in vivo. Infectivity of MHV-68 was decreased in the pcDNA3.1-MUC5AC + CPFE group compared with that in CPFE group. Meanwhile, a reduction of MHV-68 virus attenuated the expressions of chemokine (C-C motif) ligand 2 (CCL2), chemokine (C-X-C motif) ligand 5 (CXCL5), interleukin-13 (IL-13), and transforming growth factor-β1 (TGF-β1), and weakened airway inflammation and fibrosis in the pcDNA3.1-MUC5AC + CPFE group. Overexpression of MUC5AC appears to exhibit a protective role against MHV-68 infection in mice with emphysema that subsequently developed into CPFE and to further decrease airway inflammation and fibrosis induced by MHV-68 by decreasing the expressions of CCL2, CXCL5, IL-13, and TGF-β1.
Collapse
Affiliation(s)
- Li He
- Department of Respiratory and Critical Care Medicine, Jingzhou Hospital of Tongji Medical College, Huazhong University of Science and Technology, Jingzhou, Hubei, China
- Department of Respiratory and Critical Care Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qian-Qian Feng
- Department of Respiratory and Critical Care Medicine, Jingzhou Hospital of Tongji Medical College, Huazhong University of Science and Technology, Jingzhou, Hubei, China
| | - Qiao Zhang
- Department of Respiratory and Critical Care Medicine, Jingzhou Hospital of Tongji Medical College, Huazhong University of Science and Technology, Jingzhou, Hubei, China
| | - Bo Zhang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Respiratory and Critical Care Medicine, Wuhan Fourth Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Si-Si Wu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of ICU, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jian-Hua Gong
- Department of Respiratory and Critical Care Medicine, Jingzhou Hospital of Tongji Medical College, Huazhong University of Science and Technology, Jingzhou, Hubei, China
| |
Collapse
|
111
|
Austin CD, Gonzalez Edick M, Ferrando RE, Solon M, Baca M, Mesh K, Bradding P, Gauvreau GM, Sumino K, FitzGerald JM, Israel E, Bjermer L, Bourdin A, Arron JR, Choy DF, Olsson JK, Abreu F, Howard M, Wong K, Cai F, Peng K, Putnam WS, Holweg CT, Matthews JG, Kraft M, Woodruff PG. A randomized, placebo-controlled trial evaluating effects of lebrikizumab on airway eosinophilic inflammation and remodelling in uncontrolled asthma (CLAVIER). Clin Exp Allergy 2020; 50:1342-1351. [PMID: 32909660 PMCID: PMC7756263 DOI: 10.1111/cea.13731] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/16/2020] [Accepted: 08/31/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND The anti-interleukin 13 (IL-13) monoclonal antibody lebrikizumab improves lung function in patients with moderate-to-severe uncontrolled asthma, but its effects on airway inflammation and remodelling are unknown. CLAVIER was designed to assess lebrikizumab's effect on eosinophilic inflammation and remodelling. OBJECTIVE To report safety and efficacy results from enrolled participants with available data from CLAVIER. METHODS We performed bronchoscopy on patients with uncontrolled asthma before and after 12 weeks of randomized double-blinded treatment with lebrikizumab (n = 31) or placebo (n = 33). The pre-specified primary end-point was relative change in airway subepithelial eosinophils per mm2 of basement membrane (cells/mm2 ). Pre-specified secondary and exploratory outcomes included change in IL-13-associated biomarkers and measures of airway remodelling. RESULTS There was a baseline imbalance in tissue eosinophils and high variability between treatment groups. There was no discernible change in adjusted mean subepithelial eosinophils/mm2 in response to lebrikizumab (95% CI, -82.5%, 97.5%). As previously observed, FEV1 increased after lebrikizumab treatment. Moreover, subepithelial collagen thickness decreased 21.5% after lebrikizumab treatment (95% CI, -32.9%, -10.2%), and fractional exhaled nitric oxide, CCL26 and SERPINB2 mRNA expression in bronchial tissues also reduced. Lebrikizumab was well tolerated, with a safety profile consistent with other lebrikizumab asthma studies. CONCLUSIONS & CLINICAL RELEVANCE We did not observe reduced tissue eosinophil numbers in association with lebrikizumab treatment. However, in pre-specified exploratory analyses, lebrikizumab treatment was associated with reduced degree of subepithelial fibrosis, a feature of airway remodelling, as well as improved lung function and reduced key pharmacodynamic biomarkers in bronchial tissues. These results reinforce the importance of IL-13 in airway pathobiology and suggest that neutralization of IL-13 may reduce asthmatic airway remodelling. CLINICAL TRIAL REGISTRATION NCT02099656.
Collapse
Affiliation(s)
| | | | - Ronald E. Ferrando
- Genentech, Inc.South San FranciscoCAUSA
- Present address:
Stemcentrx/AbbVie, Inc.South San FranciscoCAUSA
| | | | | | | | - Peter Bradding
- University of Leicester and Glenfield HospitalLeicesterUK
| | | | - Kaharu Sumino
- Washington University School of Medicine in St. LouisSt LouisMOUSA
| | | | | | | | | | | | | | | | | | | | - Kit Wong
- Genentech, Inc.South San FranciscoCAUSA
| | - Fang Cai
- Genentech, Inc.South San FranciscoCAUSA
| | - Kun Peng
- Genentech, Inc.South San FranciscoCAUSA
| | | | | | - John G. Matthews
- Genentech, Inc.South San FranciscoCAUSA
- Present address:
23andMeMountain ViewCAUSA
| | - Monica Kraft
- University of Arizona College of MedicineTucsonAZUSA
| | | | | |
Collapse
|
112
|
Role of Cytokines in EGPA and the Possibility of Treatment with an Anti-IL-5 Antibody. J Clin Med 2020; 9:jcm9123890. [PMID: 33265990 PMCID: PMC7760889 DOI: 10.3390/jcm9123890] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 01/22/2023] Open
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA) is a type of systemic vasculitis with eosinophilia in the peripheral blood, which is preceded by bronchial asthma or allergic disease. EGPA is pathologically characterized by microangiopathy granulomatosis vasculitis. Vasculitis can be exacerbated and cause central nervous system and cardiovascular disorders and gastrointestinal perforation. Histological examination reveals eosinophil infiltration and granulomas in lesions in areas such as the lung, nervous system, and skin. Laboratory tests show inflammatory findings such as C-reactive protein (CRP) elevation, increased eosinophils, elevated serum IgE, and elevated myeloperoxidase-anti-neutrophil cytoplasmic antibodies (MPO-ANCA). MPO-ANCA is positive in approximately 40-70% of cases of this disease. EGPA is a necrotizing vasculitis that affects small- and medium-sized blood vessels; however, it differs from other types of ANCA-related vasculitis (such as microscopic polyangiitis and granulomatosis) because it is preceded by bronchial asthma and eosinophilia in the blood and tissues. Treatment with immunosuppressive agents such as steroids or cyclophosphamide depends on the Five Factor Score, which predicts the prognosis and severity of the condition. If the effect of appropriate treatment with steroids is insufficient, the anti-interleukin-5 antibody mepolizumab can be administered. The combination of mepolizumab with standard treatment leads to a significantly longer duration of remission, a higher proportion of patients who achieve sustained remission, and less steroid use than with a placebo.
Collapse
|
113
|
Kariyawasam HH. Chronic rhinosinusitis with nasal polyps: mechanistic insights from targeting IL-4 and IL-13 via IL-4Rα inhibition with dupilumab. Expert Rev Clin Immunol 2020; 16:1115-1125. [PMID: 33148074 DOI: 10.1080/1744666x.2021.1847083] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Introduction: Chronic rhinosinusitis with nasal polyps (CRSwNP) is a complex immunological upper airway disease . CRSwNP, particularly in Caucasians, often has a more distinct T2 inflammatory endotype. IL-4 and IL-13 are key upstream cytokines that help establish and sustain T2 inflammation as well as strongly influencing tissue remodeling. They have a shared signaling receptor IL-4Rα. An attractive and novel therapeutic approach is by way of blocking IL-4 and IL-13 simultaneously via inhibiting IL-4Rα. Dupilumab is a murine derived fully human monoclonal inhibitory antibody directed against IL-4Rα which thereby prevents IL-4/IL-13 cell signaling. Following successful Phase 3 studies dupilumab has become the first licensed biologic for treating CRSwNP. Areas covered: This review covers the essential immunology of CRSwNP in the context of IL-4 and IL-13 signaling via IL-4Rα. The potential mechanisms by which therapeutic improvements occur with dupilumab are evaluated. IL-4, IL-13, dupilumab and rhinosinusitis were used as the search terms in PubMed and Google Scholar through to August 2020. Expert commentary: Dupilumab has the potential to transform the care for patients with CRSwNP. It is essential that further studies are conducted promptly to identify disease-specific biomarkers and clinical traits to guide clinicians on best patient selection thereby ensuring optimal dupilumab outcomes.
Collapse
Affiliation(s)
- Harsha H Kariyawasam
- Rhinology Section, Specialist Allergy and Clinical Immunology, Royal National ENT Hospital, London University College London Hospital NHS Foundation Trust, University College London , London, UK
| |
Collapse
|
114
|
Laulajainen‐Hongisto A, Lyly A, Hanif T, Dhaygude K, Kankainen M, Renkonen R, Donner K, Mattila P, Jartti T, Bousquet J, Kauppi P, Toppila‐Salmi S. Genomics of asthma, allergy and chronic rhinosinusitis: novel concepts and relevance in airway mucosa. Clin Transl Allergy 2020; 10:45. [PMID: 33133517 PMCID: PMC7592594 DOI: 10.1186/s13601-020-00347-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/25/2020] [Indexed: 12/14/2022] Open
Abstract
Genome wide association studies (GWASs) have revealed several airway disease-associated risk loci. Their role in the onset of asthma, allergic rhinitis (AR) or chronic rhinosinusitis (CRS), however, is not yet fully understood. The aim of this review is to evaluate the airway relevance of loci and genes identified in GWAS studies. GWASs were searched from databases, and a list of loci associating significantly (p < 10-8) with asthma, AR and CRS was created. This yielded a total of 267 significantly asthma/AR-associated loci from 31 GWASs. No significant CRS -associated loci were found in this search. A total of 170 protein coding genes were connected to these loci. Of these, 76/170 (44%) showed bronchial epithelial protein expression in stained microscopic figures of Human Protein Atlas (HPA), and 61/170 (36%) had a literature report of having airway epithelial function. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) annotation analyses were performed, and 19 functional protein categories were found as significantly (p < 0.05) enriched among these genes. These were related to cytokine production, cell activation and adaptive immune response, and all were strongly connected in network analysis. We also identified 15 protein pathways that were significantly (p < 0.05) enriched in these genes, related to T-helper cell differentiation, virus infection, JAK-STAT signaling pathway, and asthma. A third of GWAS-level risk loci genes of asthma or AR seemed to have airway epithelial functions according to our database and literature searches. In addition, many of the risk loci genes were immunity related. Some risk loci genes also related to metabolism, neuro-musculoskeletal or other functions. Functions overlapped and formed a strong network in our pathway analyses and are worth future studies of biomarker and therapeutics.
Collapse
Affiliation(s)
- Anu Laulajainen‐Hongisto
- Department of Otorhinolaryngology–Head and Neck SurgeryUniversity of Helsinki and Helsinki University HospitalP.O.Box 263Kasarmikatu 11‐1300029 HUSHelsinkiFinland
- Laboratory of Cellular and Molecular ImmunologyInstitute of Microbiology of the Czech Academy of SciencesPragueCzech Republic
| | - Annina Lyly
- Department of Otorhinolaryngology–Head and Neck SurgeryUniversity of Helsinki and Helsinki University HospitalP.O.Box 263Kasarmikatu 11‐1300029 HUSHelsinkiFinland
- Skin and Allergy HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | | | | | - Matti Kankainen
- HUS Diagnostic CenterHelsinki University HospitalHelsinkiFinland
- Hematology Research Unit HelsinkiDepartment of HematologyHelsinki University Hospital Comprehensive Cancer CenterHelsinkiFinland
- Translational Immunology Research Program and Department of Clinical ChemistryUniversity of HelsinkiHelsinkiFinland
| | - Risto Renkonen
- Haartman InstituteUniversity of HelsinkiHelsinkiFinland
- HUS Diagnostic CenterHelsinki University HospitalHelsinkiFinland
| | - Kati Donner
- Hematology Research Unit HelsinkiDepartment of HematologyHelsinki University Hospital Comprehensive Cancer CenterHelsinkiFinland
| | - Pirkko Mattila
- Haartman InstituteUniversity of HelsinkiHelsinkiFinland
- Hematology Research Unit HelsinkiDepartment of HematologyHelsinki University Hospital Comprehensive Cancer CenterHelsinkiFinland
| | - Tuomas Jartti
- Department of Pediatrics and Adolescent MedicineTurku University Hospital and University of TurkuTurkuFinland
| | - Jean Bousquet
- Université MontpellierMontpellierFrance
- MACVIA‐FranceMontpellierFrance
- Corporate Member of Freie Universität BerlinHumboldt‐Universität Zu BerlinBerlin Institute of HealthComprehensive Allergy CenterDepartment of Dermatology and AllergyCharité–Universitätsmedizin BerlinBerlinGermany
| | - Paula Kauppi
- Skin and Allergy HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Sanna Toppila‐Salmi
- Skin and Allergy HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
- Haartman InstituteUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
115
|
Comparative efficacy of glucocorticoid receptor agonists on Th2 cell function and attenuation by progesterone. BMC Immunol 2020; 21:54. [PMID: 33076829 PMCID: PMC7574173 DOI: 10.1186/s12865-020-00383-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/05/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Corticosteroids (CS)s suppress cytokine production and induce apoptosis of inflammatory cells. Prednisone and dexamethasone are oral CSs prescribed for treating asthma exacerbations. While prednisone is more commonly prescribed, dexamethasone is long acting and a more potent glucocorticoid receptor (GR) agonist. It can be administered as a one or two dose regime, unlike the five to seven days required for prednisone, a feature that increases compliance. We compared the relative ability of these two oral CSs to suppress type 2 inflammation. Since progesterone has affinity for the GR and women are more likely to relapse following an asthma exacerbation, we assessed its influence on CS action. RESULTS Dexamethasone suppressed the level of IL-5 and IL-13 mRNA within Th2 cells with ~ 10-fold higher potency than prednisolone (the active form of prednisone). Dexamethasone induced a higher proportion of apoptotic and dying cells than prednisolone, at all concentrations examined. Addition of progesterone reduced the capacity of both CS to drive cell death, though dexamethasone maintained significantly more killing activity. Progesterone blunted dexamethasone-induction of FKBP5 mRNA, indicating that the mechanism of action was by interference of the CS:GR complex. CONCLUSIONS Dexamethasone is both more potent and effective than prednisolone in suppressing type 2 cytokine levels and mediating apoptosis. Progesterone attenuated these anti-inflammatory effects, indicating its potential influence on CS responses in vivo. Collectively, our data suggest that when oral CS is required, dexamethasone may be better able to control type 2 inflammation, eliminate Th2 cells and ultimately lead to improved long-term outcomes. Further research in asthmatics is needed.
Collapse
|
116
|
Lee SY, Kim MH, Kim SH, Ahn T, Kim SW, Kwak YS, Cho IH, Nah SY, Cho SS, Park KM, Park DH, Bae CS. Korean Red Ginseng affects ovalbumin-induced asthma by modulating IL-12, IL-4, and IL-6 levels and the NF-κB/COX-2 and PGE 2 pathways. J Ginseng Res 2020; 45:482-489. [PMID: 34295208 PMCID: PMC8282494 DOI: 10.1016/j.jgr.2020.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 09/16/2020] [Accepted: 10/14/2020] [Indexed: 11/29/2022] Open
Abstract
Background Asthma is an incurable hyper-responsive disease of the pulmonary system that is caused by various allergens, including indoor and outdoor stimulators. According to the Global Asthma Network, 339 million people suffered from asthma in 2018, with particularly severe forms in children. Numerous treatments for asthma are available; however, they are frequently associated with adverse effects such as growth retardation, neurological disorders (e.g., catatonia, poor concentration, and insomnia), and physiological disorders (e.g., immunosuppression, hypertension, hyperglycemia, and osteoporosis). Methods Korean Red Ginseng has long been used to treat numerous diseases in many countries, and we investigated the anti-asthmatic effects and mechanisms of action of Korean Red Ginseng. Eighty-four BALB/c mice were assigned to 6 treatment groups: control, ovalbumin-induced asthma group, dexamethasone treatment group, and 3 groups treated with Korean Red Ginseng water extract (KRGWE) at 5, 25, or 50 mg/kg/day for 5 days. Anti-asthmatic effects of KRGWE were assessed based on biological changes, such as white blood cell counts and differential counts in the bronchoalveolar lavage fluid, serum IgE levels, and histopathological changes in the lungs, and by examining anti-asthmatic mechanisms, such as the cytokines associated with Th1, Th2, and Treg cells and inflammation pathways. Results KRGWE affected ovalbumin-induced changes, such as increased white blood cell counts, increased IgE levels, and morphological changes (mucous hypersecretion, epithelial cell hyperplasia, inflammatory cell infiltration) by downregulating cytokines such as IL-12, IL-4, and IL-6 via GATA-3 inactivation and suppression of inflammation via NF-κB/COX-2 and PGE2 pathways. Conclusion KRGWE is a promising drug for asthma treatment.
Collapse
Affiliation(s)
- Soon-Young Lee
- College of Korean Medicine, Dongshin University, Naju, Jeonnam, Republic of Korea
| | - Min-Hee Kim
- College of Agriculture and Life Science, Chonnam National University, Gwangju, Republic of Korea
| | - Seung-Hyun Kim
- College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Taeho Ahn
- College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Sung-Won Kim
- Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Yi-Seong Kwak
- Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Ik-Hyun Cho
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - Seung-Sik Cho
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Jeonnam, Republic of Korea
| | - Kyung Mok Park
- College of Korean Medicine, Dongshin University, Naju, Jeonnam, Republic of Korea
| | - Dae-Hun Park
- College of Korean Medicine, Dongshin University, Naju, Jeonnam, Republic of Korea
- Corresponding author. College of Korean Medicine, Dongshin University, 67 Donsghindae-gil, Naju, Jeonnam, 58245, Republic of Korea.
| | - Chun-Sik Bae
- College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
- Corresponding author. College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
117
|
He CH, Lee CG, Ma B, Kamle S, Choi AMK, Elias JA. N-Glycosylation Regulates Chitinase 3-like-1 and IL-13 Ligand Binding to IL-13 Receptor α2. Am J Respir Cell Mol Biol 2020; 63:386-395. [PMID: 32402213 DOI: 10.1165/rcmb.2019-0446oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Chitinase 3-like-1 (Chi3l1) and IL-13 are both ligands of IL-13 receptor α2 (IL-13Rα2). The binding of the former activates mitogen-activated protein kinase, AKT, and Wnt/β-catenin signaling, and plays important roles in innate and adaptive immunity, cellular apoptosis, oxidative injury, allergic inflammation, tumor metastasis and wound healing, fibrosis, and repair in the lung. In contrast, the latter binding is largely a decoy event that diminishes the effects of IL-13. Here, we demonstrate that IL-13Rα2 N-glycosylation is a critical determinant of which ligand binds. Structure-function evaluations demonstrated that Chi3l1-IL-13Rα2 binding was increased when sites of N-glycosylation are mutated, and studies with tunicamycin and Peptide:N-glycosidase F (PNGase F) demonstrated that Chi3l1-IL-13Rα2 binding and signaling were increased when N-glycosylation was diminished. In contrast, structure-function experiments demonstrated that IL-13 binding to IL-13Rα2 was dependent on each of the four sites of N-glycosylation in IL-13Rα2, and experiments with tunicamycin and PNGase F demonstrated that IL-13-IL-13Rα2 binding was decreased when IL-13Rα2 N-glycosylation was diminished. Studies with primary lung epithelial cells also demonstrated that Chi3l1 inhibited, whereas IL-13 stimulated, N-glycosylation as evidenced by the ability of Chi3l1 to inhibit and IL-13 to stimulate the subunits of the oligosaccharide complex A and B (STT3A and STT3B). These studies demonstrate that N-glycosylation is a critical determinant of Chi3l1 and IL-13 binding to IL-13Rα2, and highlight the ability of Chi3l1 and IL-13 to alter key elements of the N-glycosylation apparatus in a manner that would augment their respective binding.
Collapse
Affiliation(s)
- Chuan Hua He
- Department of Molecular Microbiology and Immunology and
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology and
| | - Bing Ma
- Department of Molecular Microbiology and Immunology and
| | | | - Augustine M K Choi
- Department of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology and.,Department of Medicine, Brown University, Providence, Rhode Island; and
| |
Collapse
|
118
|
Song MK, Kim DI, Lee K. Kathon Induces Fibrotic Inflammation in Lungs: The First Animal Study Revealing a Causal Relationship between Humidifier Disinfectant Exposure and Eosinophil and Th2-Mediated Fibrosis Induction. Molecules 2020; 25:molecules25204684. [PMID: 33066398 PMCID: PMC7587358 DOI: 10.3390/molecules25204684] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 12/21/2022] Open
Abstract
Currently available toxicity data on humidifier disinfectants are primarily limited to polyhexamethylene guanidine phosphate-induced lung fibrosis. We, therefore, investigated whether the sterilizer component Kathon, which is a mixture of chloromethylisothiazolinone and methylisothiazolinone, induces fibrotic lung injury following direct lung exposure in an animal model. Mice were intratracheally instilled with either the vehicle or Kathon. Differential cell counts, cytokine analysis, and histological analysis of lung tissue were then performed to characterize the injury features, and we investigated whether Kathon altered fibrosis-related gene expression in lung tissues via RNA-Seq and bioinformatics. Cell counting showed that Kathon exposure increased the proportion of macrophages, eosinophils, and neutrophils. Moreover, T helper 2 (Th2) cytokine levels in the bronchoalveolar lavage were significantly increased in the Kathon groups. Histopathological analysis revealed increased perivascular/alveolar inflammation, eosinophilic cells, mucous cell hyperplasia, and pulmonary fibrosis following Kathon exposure. Additionally, Kathon exposure modulated the expression of genes related to fibrotic inflammation, including the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway, extracellular signal regulated kinase (ERK)1 and ERK2 cascade, extracellular matrix (ECM)-receptor interaction pathway, transforming growth factor beta receptor signaling pathway, cellular response to tumor necrosis factor, and collagen fibril organization. Our results suggest that Kathon exposure is associated with fibrotic lung injury via a Th2-dependent pathway and is thus a possible risk factor for fibrosis.
Collapse
Affiliation(s)
- Mi-Kyung Song
- National Center for Efficacy Evaluation of Respiratory Disease Product, Korea Institute of Toxicology, 30 Baehak1-gil, Jongeup, Jeollabuk-do 56212, Korea; (M.-K.S.); (D.I.K.)
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Korea
| | - Dong Im Kim
- National Center for Efficacy Evaluation of Respiratory Disease Product, Korea Institute of Toxicology, 30 Baehak1-gil, Jongeup, Jeollabuk-do 56212, Korea; (M.-K.S.); (D.I.K.)
| | - Kyuhong Lee
- National Center for Efficacy Evaluation of Respiratory Disease Product, Korea Institute of Toxicology, 30 Baehak1-gil, Jongeup, Jeollabuk-do 56212, Korea; (M.-K.S.); (D.I.K.)
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Korea
- Correspondence: or ; Tel.: +82-63-570-8740
| |
Collapse
|
119
|
Nemeth J, Schundner A, Quast K, Winkelmann VE, Frick M. A Novel Fibroblast Reporter Cell Line for in vitro Studies of Pulmonary Fibrosis. Front Physiol 2020; 11:567675. [PMID: 33162897 PMCID: PMC7582034 DOI: 10.3389/fphys.2020.567675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal disease of the lower respiratory tract with restricted therapeutic options. Repetitive injury of the bronchoalveolar epithelium leads to activation of pulmonary fibroblasts, differentiation into myofibroblasts and excessive extracellular matrix (ECM) deposition resulting in aberrant wound repair. However, detailed molecular and cellular mechanisms underlying initiation and progression of fibrotic changes are still elusive. Here, we report the generation of a representative fibroblast reporter cell line (10-4A BFP ) to study pathophysiological mechanisms of IPF in high throughput or high resolution in vitro live cell assays. To this end, we immortalized primary fibroblasts isolated from the distal lung of Sprague-Dawley rats. Molecular and transcriptomic characterization identified clone 10-4A as a matrix fibroblast subpopulation. Mechanical or chemical stimulation induced a reversible fibrotic state comparable to effects observed in primary isolated fibroblasts. Finally, we generated a reporter cell line (10-4A BFP ) to express nuclear blue fluorescent protein (BFP) under the promotor of the myofibroblast marker alpha smooth muscle actin (Acta2) using CRISPR/Cas9 technology. We evaluated the suitability of 10-4A BFP as reporter tool in plate reader assays. In summary, the 10-4A BFP cell line provides a novel tool to study fibrotic processes in vitro to gain new insights into the cellular and molecular processes involved in fibrosis formation and propagation.
Collapse
Affiliation(s)
- Julia Nemeth
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | | - Karsten Quast
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
120
|
Song M, Lee SY, Kim M, Park S, Park J, Kwon Y, Park DH. Saururus chinensis-controlled allergic pulmonary disease through NF-κB/COX-2 and PGE 2 pathways. PeerJ 2020; 8:e10043. [PMID: 33024647 PMCID: PMC7520084 DOI: 10.7717/peerj.10043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/04/2020] [Indexed: 12/28/2022] Open
Abstract
Saururus chinensis is a perennial herb found in the northeastern regions of Asia, including Korea, China, and Japan, and is used in traditional medicine. Studies have identified the four major constituents in Saururus chinensis water extract (LHF618®) as miquelianin (11.75 ± 0.092 mg/g), rutin (1.20 ± 0.008 mg/g), quercitrin (2.38 ± 0.389 mg/g), and quercetin (0.068 ± 0.017 mg/g). Saururus chinensis can improve the symptoms of ovalbumin- or fine dust-induced allergic pulmonary disease by suppressing the effects of WBCs and neutrophils in BALF and IgE in the serum. Saururus chinensis dose-dependently recovered morphological changes such as mucous hyper secretion (from 2.7 ± 0.46 to 0.6 ± 0.65), pulmonary epithelial cell hyperplasia (from 2.4 ± 0.55 to 0.7 ± 0.67), and inflammatory cell infiltration (from 2.3 ± 0.45 to 0.6 ± 0.43), and effectively controlled cDNA levels and protein levels of IL-13. It inhibited NF-κB translocation and COX-2 protein synthesis and suppressed the expression of PGE2. Our results show that Saururus chinensis controlled allergic pulmonary disease via the anti-inflammatory pathways, NF-κB/COX-2 and PGE2. Saururus chinensis may be a promising drug candidate against fine dust-induced allergic pulmonary disease.
Collapse
Affiliation(s)
- MiKyung Song
- Bio Technology R&D Center, WiLab Co., Ltd., Seoul, South Korea
| | - Soon-Young Lee
- Department of Nursing, Dongshin University, Naju, South Korea
| | - Minhee Kim
- Department of Forestry, Chonnam National University, Gwangji, South Korea
| | - Sangwoug Park
- Bio Technology R&D Center, WiLab Co., Ltd., Seoul, South Korea
| | - Juyeon Park
- Bio Technology R&D Center, WiLab Co., Ltd., Seoul, South Korea
| | - Yongbum Kwon
- Bio Technology R&D Center, WiLab Co., Ltd., Seoul, South Korea
| | - Dae-Hun Park
- Department of Nursing, Dongshin University, Naju, South Korea
| |
Collapse
|
121
|
Bidirectional interaction of airway epithelial remodeling and inflammation in asthma. Clin Sci (Lond) 2020; 134:1063-1079. [PMID: 32369100 DOI: 10.1042/cs20191309] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/28/2020] [Accepted: 04/20/2020] [Indexed: 12/18/2022]
Abstract
Asthma is a chronic disease of the airways that has long been viewed predominately as an inflammatory condition. Accordingly, current therapeutic interventions focus primarily on resolving inflammation. However, the mainstay of asthma therapy neither fully improves lung function nor prevents disease exacerbations, suggesting involvement of other factors. An emerging concept now holds that airway remodeling, another major pathological feature of asthma, is as important as inflammation in asthma pathogenesis. Structural changes associated with asthma include disrupted epithelial integrity, subepithelial fibrosis, goblet cell hyperplasia/metaplasia, smooth muscle hypertrophy/hyperplasia, and enhanced vascularity. These alterations are hypothesized to contribute to airway hyperresponsiveness, airway obstruction, airflow limitation, and progressive decline of lung function in asthmatic individuals. Consequently, targeting inflammation alone does not suffice to provide optimal clinical benefits. Here we review asthmatic airway remodeling, focusing on airway epithelium, which is critical to maintaining a healthy respiratory system, and is the primary defense against inhaled irritants. In asthma, airway epithelium is both a mediator and target of inflammation, manifesting remodeling and resulting obstruction among its downstream effects. We also highlight the potential benefits of therapeutically targeting airway structural alterations. Since pathological tissue remodeling is likewise observed in other injury- and inflammation-prone tissues and organs, our discussion may have implications beyond asthma and lung disease.
Collapse
|
122
|
Worrell JC, Walsh SM, Fabre A, Kane R, Hinz B, Keane MP. CXCR3A promotes the secretion of the antifibrotic decoy receptor sIL-13Rα2 by pulmonary fibroblasts. Am J Physiol Cell Physiol 2020; 319:C1059-C1069. [PMID: 33026833 DOI: 10.1152/ajpcell.00076.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
CXC chemokine receptor 3 (CXCR3) A and its IFN-inducible ligands CXCL9 and CXCL10 regulate vascular remodeling and fibroblast motility. IL-13 is a profibrotic cytokine implicated in the pathogenesis of inflammatory and fibroproliferative conditions. Previous work from our laboratory has shown that CXCR3A is negatively regulated by IL-13 and is necessary for the basal regulation of the IL-13 receptor subunit IL-13Rα2. This study investigates the regulation of fibroblast phenotype, function, and downstream IL-13 signaling by CXCR3A in vitro. CXCR3A was overexpressed via transient transfection. CXCR3A-/- lung fibroblasts were isolated for functional analysis. Additionally, the contribution of CXCR3A to tissue remodeling following acute lung injury was assessed in vivo with wild-type (WT) and CXCR3-/- mice challenged with IL-13. CXCR3 and IL-13Rα2 displayed a reciprocal relationship after stimulation with either IL-13 or CXCR3 ligands. CXCR3A reduced expression of fibroblast activation makers, soluble collagen production, and proliferation. CXCR3A enhanced the basal expression of pERK1/2 while inducing IL-13-mediated downregulation of NF-κB-p65. CXCR3A-/- pulmonary fibroblasts were increasingly proliferative and displayed reduced contractility and α-smooth muscle actin expression. IL-13 challenge regulated expression of the CXCR3 ligands and soluble IL-13Rα2 levels in lungs and bronchoalveolar lavage fluid (BALF) of WT mice; this response was absent in CXCR3-/- mice. Alveolar macrophage accumulation and expression of genes involved in lung remodeling was increased in CXCR3-/- mice. We conclude that CXCR3A is a central antifibrotic factor in pulmonary fibroblasts, limiting fibroblast activation and reducing extracellular matrix (ECM) production. Therefore, targeting of CXCR3A may be a novel approach to regulating fibroblast activity in lung fibrosis and remodeling.
Collapse
Affiliation(s)
- Julie C Worrell
- St. Vincent's University Hospital and School of Medicine, University College Dublin and UCD Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland
| | - Sinead M Walsh
- St. Vincent's University Hospital and School of Medicine, University College Dublin and UCD Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland.,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Aurélie Fabre
- St. Vincent's University Hospital and School of Medicine, University College Dublin and UCD Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland.,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.,UCD Conway Research Pathology Core Technology, University College Dublin, Dublin, Ireland
| | - Rosemary Kane
- St. Vincent's University Hospital and School of Medicine, University College Dublin and UCD Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Michael P Keane
- St. Vincent's University Hospital and School of Medicine, University College Dublin and UCD Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland.,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
123
|
Socheongryongtang Modulates Asthma-Related Changes via Modulation of TNF-α and T-bet as well as IFN-γ in an Asthma Murine Model. Processes (Basel) 2020. [DOI: 10.3390/pr8091167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In 2017 the World Health Organization (WHO) reported that 235 million people suffered from asthma, and that 383,000 deaths were due to asthma in 2015. Asthma cannot be completely eradicated and the medications for asthma are associated with many adverse effects. Socheongryongtang is one of the prescriptions which has traditionally been used for the treatment of pulmonary disease, but the anti-asthmatic mechanism is unclear. To investigate the anti-asthmatic mechanism of socheongryongtang, BALB/c mice were divided into five groups: control, asthma-induced control, dexamethasone treatment, and 150 mg/kg or 1500 mg/kg socheongryongtang treatment and several biomarkers were analyzed, such as white blood cell (WBC) and differential counts in broncheoalveolar fluid (BALF), immunoglobulin E (IgE) in serum, and morphological changes/helper T cell-related cytokines/transcription factor in the lung. The therapeutic ingredients were also analyzed. Socheongryongtang inhibited the neutrophils differentiation in BALF, controlled interleukin (IL)-12p40 releasing, down-regulated not only GATA-3 and helper 2 T (Th2) cell transcription factors but also IL-4, and also decreased the level of tumor necrosis factor (TNF)-α in the lung. In addition, through high-performance liquid chromatography (HPLC) analysis, we confirmed that the therapeutic ingredients in socheongryongtang were paeoniflorin, liquiritin, and glycyrrhizin. The oral intake of 7.3 g of socheongryongtang is beneficial for suppressing the possibility of the occurrence of asthma via modulation of TNF-α and T-bet as well as IFN-γ.
Collapse
|
124
|
Schuler CF, Malinczak C, Best SKK, Morris SB, Rasky AJ, Ptaschinski C, Lukacs NW, Fonseca W. Inhibition of uric acid or IL-1β ameliorates respiratory syncytial virus immunopathology and development of asthma. Allergy 2020; 75:2279-2293. [PMID: 32277487 DOI: 10.1111/all.14310] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 03/04/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) affects most infants early in life and is associated with increased asthma risk. The specific mechanism remains unknown. OBJECTIVE To investigate the role of uric acid (UA) and IL-1β in RSV immunopathology and asthma predisposition. METHODS Tracheal aspirates from human infants with and without RSV were collected and analyzed for pro-IL-1β mRNA and protein to establish a correlation in human disease. Neonatal mouse models of RSV were employed, wherein mice infected at 6-7 days of life were analyzed at 8 days postinfection, 5 weeks postinfection, or after a chronic cockroach allergen asthma model. A xanthine oxidase inhibitor or IL-1 receptor antagonist was administered during RSV infection. RESULTS Human tracheal aspirates from RSV-infected infants showed elevated pro-IL-1β mRNA and protein. Inhibition of UA or IL-1β during neonatal murine RSV infection decreased mucus production, reduced cellular infiltrates to the lung (especially ILC2s), and decreased type 2 immune responses. Inhibition of either UA or IL-1β during RSV infection led to chronic reductions in pulmonary immune cell composition and reduced type 2 immune responses and reduced similar responses after challenge with cockroach antigen. CONCLUSIONS Inhibiting UA and IL-1β during RSV infection ameliorates RSV immunopathology, reduces the consequences of allergen-induced asthma, and presents new therapeutic targets to reduce early-life viral-induced asthma development.
Collapse
Affiliation(s)
- Charles F. Schuler
- Division of Allergy and Clinical Immunology Department of Internal Medicine University of Michigan Ann Arbor MI USA
- Mary H. Weiser Food Allergy Center University of Michigan Ann Arbor MI USA
| | | | | | - Susan B. Morris
- Department of Pathology University of Michigan Ann Arbor MI USA
| | - Andrew J. Rasky
- Department of Pathology University of Michigan Ann Arbor MI USA
| | - Catherine Ptaschinski
- Mary H. Weiser Food Allergy Center University of Michigan Ann Arbor MI USA
- Department of Pathology University of Michigan Ann Arbor MI USA
| | - Nicholas W. Lukacs
- Mary H. Weiser Food Allergy Center University of Michigan Ann Arbor MI USA
- Department of Pathology University of Michigan Ann Arbor MI USA
| | - Wendy Fonseca
- Department of Pathology University of Michigan Ann Arbor MI USA
| |
Collapse
|
125
|
Asano T, Ohbayashi H, Ariga M, Furuta O, Kudo S, Ono J, Izuhara K. Serum periostin reflects dynamic hyperinflation in patients with asthma. ERJ Open Res 2020; 6:00347-2019. [PMID: 32714962 PMCID: PMC7369445 DOI: 10.1183/23120541.00347-2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/30/2020] [Indexed: 12/26/2022] Open
Abstract
Introduction Dynamic hyperinflation (DH) is sometimes observed and is associated with impaired daily life activities of asthma. We assessed the relationship between DH and asthma biomarkers (blood eosinophil, fractional exhaled nitric oxide (F eNO) and serum periostin) in patients with asthma. Methods Fifty patients with stable asthma were prospectively recruited and underwent blood test, F eNO measurement, spirometry and metronome-paced tachypnoea (MPT) test to assess DH. In MPT tests, inspiratory capacity (IC) was measured at baseline and after 30 s of MPT with breathing frequencies of 20, 30 and 40 breaths·min-1. DH was assessed by the decline of IC from baseline, and maximal IC reduction ≥10% was considered as positive DH. Results Thirty patients (60%) showed positive DH. Patients with positive DH showed higher serum periostin levels (107.0±30.7 ng·mL-1) than patients with negative DH (89.7±23.7) (p=0.04). Patients in Global Initiative for Asthma treatment steps 4-5 (n=19) showed higher serum periostin levels (p=0.01) and more severe IC reduction after MPT (p<0.0001) than patients in steps 1-3 (n=31). Maximal IC reduction after MPT was significantly correlated with asthma control test score (r=-0.28, p=0.05), forced expiratory volume in 1 s (r=-0.56, p<0.0001), and serum periostin levels (r=0.41, p=0.003). Conclusion Serum periostin may have the possibility to reflect DH in patients with stable asthma.
Collapse
Affiliation(s)
- Takamitsu Asano
- Dept of Allergy and Respiratory Medicine, Tohno Chuo Clinic, Gifu, Japan
| | - Hiroyuki Ohbayashi
- Dept of Allergy and Respiratory Medicine, Tohno Chuo Clinic, Gifu, Japan
| | - Mitsue Ariga
- Dept of Allergy and Respiratory Medicine, Tohno Chuo Clinic, Gifu, Japan
| | - Osamu Furuta
- Dept of Allergy and Respiratory Medicine, Tohno Chuo Clinic, Gifu, Japan
| | - Sahori Kudo
- Dept of Allergy and Respiratory Medicine, Tohno Chuo Clinic, Gifu, Japan
| | - Junya Ono
- The Shino-Test Corporation, Sagamihara, Japan
| | - Kenji Izuhara
- Dept of Biomolecular Sciences, Saga Medical School, Saga, Japan
| |
Collapse
|
126
|
Gandhi GR, Vasconcelos ABS, Haran GH, Calisto VKDS, Jothi G, Quintans JDSS, Cuevas LE, Narain N, Júnior LJQ, Cipolotti R, Gurgel RQ. Essential oils and its bioactive compounds modulating cytokines: A systematic review on anti-asthmatic and immunomodulatory properties. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 73:152854. [PMID: 31036393 DOI: 10.1016/j.phymed.2019.152854] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 05/06/2023]
Abstract
BACKGROUND Asthma, the main inflammatory chronic condition affecting the respiratory system, is characterized by hyperresponsiveness and reversible airway obstruction, recruitment of inflammatory cells and excessive production of mucus. Cytokines as biochemical messengers of immune cells, play an important role in the regulation of allergic inflammatory and infectious airway processes. Essential oils of plant origin are complex mixtures of volatile and semi volatile organic compounds that determine the specific aroma of plants and are categorized by their biological activities. PURPOSE We reviewed whether essential oils and their bioactive compounds of plant origin could modulate cytokines' immune responses and improve asthma therapy in experimental systems in vitro and in vivo. METHODS Electronic and manual search of articles in English available from inception up to November 2018 reporting the immunomodulatory activity of essential oils and their bioactive compounds for the management of asthma. We used PubMed, EMBASE, Scopus and Web of Science. Publications reporting preclinical experiments where cytokines were examined to evaluate the consequence of anti-asthmatic therapy were included. RESULTS 914 publications were identified and 13 were included in the systematic review. Four articles described the role of essential oils and their bioactive compounds on bronchial asthma using cell lines; nine in vivo studies evaluated the anti-inflammatory efficacy and immunomodulating effects of essential oil and their secondary metabolites on cytokines production and inflammatory responses. The most important immunopharmacological mechanisms reported were the regulation of cytokine production, inhibition of reactive oxygen species accumulation, inactivation of eosinophil migration and remodeling of the airways and lung tissue, modulation of FOXP3 gene expression, regulation of inflammatory cells in the airways and decreasing inflammatory mediator expression levels. CONCLUSION Plant derived essential oils and related active compounds have potential therapeutic activity for the treatment of asthma by modulating the release of pro-inflammatory (TNF-α, IL-1β, IL-8), Th17 (IL-17), anti-inflammatory (IL-10), Th1 (IFN-γ, IL-2, IL-12) and Th2 (IL-4, IL-5, IL-6, IL-13) cytokines and the suppression of inflammatory cell accumulation.
Collapse
Affiliation(s)
- Gopalsamy Rajiv Gandhi
- Division of Paediatrics, Department of Medicine, Federal University of Sergipe, Rua Cláudio Batista, s/n, Cidade Nova, Aracaju, 49.100-000 Sergipe, Brazil; Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, 49.100-000 Sergipe, Brazil.
| | | | - Govindasamy Hari Haran
- Department of Biochemistry, Srimad Andavan Arts and Science College (Autonomous), Tiruchirappalli, 620005 Tamil Nadu, India
| | - Valdete Kaliane da Silva Calisto
- Division of Paediatrics, Department of Medicine, Federal University of Sergipe, Rua Cláudio Batista, s/n, Cidade Nova, Aracaju, 49.100-000 Sergipe, Brazil
| | - Gnanasekaran Jothi
- Department of Biochemistry, Srimad Andavan Arts and Science College (Autonomous), Tiruchirappalli, 620005 Tamil Nadu, India
| | - Jullyana de Souza Siqueira Quintans
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, 49.100-000 Sergipe, Brazil
| | - Luis Eduardo Cuevas
- Liverpool School of Tropical Medicine, Pembroke Place Liverpool, Liverpool, UK
| | - Narendra Narain
- Laboratory of Flavor and Chromatographic Analysis, Federal University of Sergipe, São Cristóvão, Aracaju, Sergipe 49.100-000, Brazil
| | - Lucindo José Quintans Júnior
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, 49.100-000 Sergipe, Brazil
| | - Rosana Cipolotti
- Division of Paediatrics, Department of Medicine, Federal University of Sergipe, Rua Cláudio Batista, s/n, Cidade Nova, Aracaju, 49.100-000 Sergipe, Brazil
| | - Ricardo Queiroz Gurgel
- Division of Paediatrics, Department of Medicine, Federal University of Sergipe, Rua Cláudio Batista, s/n, Cidade Nova, Aracaju, 49.100-000 Sergipe, Brazil.
| |
Collapse
|
127
|
Usui-Kawanishi F, Takahashi M, Sakai H, Suto W, Kai Y, Chiba Y, Hiraishi K, Kurahara LH, Hori M, Inoue R. Implications of immune-inflammatory responses in smooth muscle dysfunction and disease. J Smooth Muscle Res 2020; 55:81-107. [PMID: 32023567 PMCID: PMC6997890 DOI: 10.1540/jsmr.55.81] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In the past few decades, solid evidence has been accumulated for the pivotal significance
of immunoinflammatory processes in the initiation, progression, and exacerbation of many
diseases and disorders. This groundbreaking view came from original works by Ross who
first described that excessive inflammatory-fibroproliferative response to various forms
of insult to the endothelium and smooth muscle of the artery wall is essential for the
pathogenesis of atherosclerosis (Ross, Nature 1993; 362(6423): 801–9). It is now widely
recognized that both innate and adaptive immune reactions are avidly involved in the
inflammation-related remodeling of many tissues and organs. When this state persists,
irreversible fibrogenic changes would occur often culminating in fatal insufficiencies of
many vital parenchymal organs such as liver, lung, heart, kidney and intestines. Thus,
inflammatory diseases are becoming the common life-threatening risk for and urgent concern
about the public health in developed countries (Wynn et al., Nature Medicine 2012; 18(7):
1028–40). Considering this timeliness, we organized a special symposium entitled
“Implications of immune/inflammatory responses in smooth muscle dysfunction and disease”
in the 58th annual meeting of the Japan Society of Smooth Muscle Research. This symposium
report will provide detailed synopses of topics presented in this symposium; (1) the role
of inflammasome in atherosclerosis and abdominal aortic aneurysms by Fumitake
Usui-Kawanishi and Masafumi Takahashi; (2) Mechanisms underlying the pathogenesis of
hyper-contractility of bronchial smooth muscle in allergic asthma by Hiroyasu Sakai,
Wataru Suto, Yuki Kai and Yoshihiko Chiba; (3) Vascular remodeling in pulmonary arterial
hypertension by Keizo Hiraishi, Lin Hai Kurahara and Ryuji Inoue.
Collapse
Affiliation(s)
- Fumitake Usui-Kawanishi
- Division of Biopharmaceutical Engineering, Department of Pharmaceutical Engineering, Toyoma Prefectural University, 5180 Kurokawa, Imizu-shi, Toyama 939-0398, Japan.,Division of Inflammation Research, Center of Molecular Medicine, Jichi Medical University, 3311-159 Yakushiji, Shimono-shi, Tochigi 329-0498, Japan
| | - Masafumi Takahashi
- Division of Inflammation Research, Center of Molecular Medicine, Jichi Medical University, 3311-159 Yakushiji, Shimono-shi, Tochigi 329-0498, Japan
| | - Hiroyasu Sakai
- Department of Analytical Pathophysiology, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Wataru Suto
- Department of Physiology and Molecular Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Yuki Kai
- Department of Analytical Pathophysiology, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Yoshihiko Chiba
- Department of Physiology and Molecular Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Keizo Hiraishi
- Department of Physiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Lin Hai Kurahara
- Department of Physiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.,Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, 1750-1 Ido, Miki-machi, Kida-gun, Kagawa 761-0793, Japan
| | - Masatoshi Hori
- Department of Veterinary Pharmacology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Ryuji Inoue
- Department of Physiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| |
Collapse
|
128
|
Mendoza DP, Kohli P, Nance JW, Singh R, Cho J, Griffith J, Harris RS, Kelly VJ, Luster AD, Medoff B, Digumarthy SR. Lung parenchymal and airway changes on CT imaging following allergen challenge and bronchoalveolar lavage in atopic and asthmatic subjects. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:862. [PMID: 32793706 DOI: 10.21037/atm-20-1719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Computed tomography (CT) imaging findings in the lungs in the setting of an acute allergic response and following bronchoalveolar lavage (BAL) are not well established. Our goals are to characterize the pulmonary CT findings of acute allergic response in both asthmatic and non-asthmatic subjects and, secondarily, to characterize the pulmonary imaging findings following BAL. Methods In this prospective observational (cohort) study, we identified atopic, asthmatic (AA) and atopic, non-asthmatic (ANA) subjects. CT of the chest was performed following BAL and instillation of an allergen (AL) and of an inert diluent (DL). Two radiologists analyzed the CT examinations for airway and parenchymal changes. Results We had a cohort of 20 atopic subjects (AA=10, ANA=10; F=11, M=9; median age: 23.5 years, range: 18-48 years). Compared to diluent instillation and BAL, allergen instillation resulted in more significant bronchial wall thickening (AL=70%, DL=0%, BAL=0%, P<0.01), consolidations (AL=55%, DL=0%, BAL=15%, P<0.05), and septal thickening (AL=35%, DL=0%, BAL=0%, P<0.01). When present, consolidations tended to be more common in asthmatic subjects compared to non-asthmatics following instillation of the allergen, although this did not reach statistical significance (AA=80% vs. ANA=30%; P=0.07). BAL, on the other hand, resulted in more ground-glass opacities (BAL=15/20, 75% vs. AL=2/20, 10%, vs. DL=0/20, 0%; P<0.01). Conclusions Acute allergic response in the lungs can result in significant bronchial wall thickening, septal thickening, and consolidations in those with atopy, particularly those with asthma. Localized ground-glass opacities may be expected following BAL, and care should be taken so as to not misinterpret these as significant pathology.
Collapse
Affiliation(s)
- Dexter P Mendoza
- Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, Boston, MA, USA
| | - Puja Kohli
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - John W Nance
- Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, Boston, MA, USA
| | - Ramandeep Singh
- Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, Boston, MA, USA
| | - Josalyn Cho
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jason Griffith
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - R Scott Harris
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Vanessa J Kelly
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Andrew D Luster
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Boston, MA, USA
| | - Benjamin Medoff
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Subba R Digumarthy
- Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
129
|
Fonseca W, Malinczak CA, Schuler CF, Best SK, Rasky AJ, Morris SB, Cui TX, Popova AP, Lukacs NW. Uric acid pathway activation during respiratory virus infection promotes Th2 immune response via innate cytokine production and ILC2 accumulation. Mucosal Immunol 2020; 13:691-701. [PMID: 32047272 PMCID: PMC7316593 DOI: 10.1038/s41385-020-0264-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 01/05/2020] [Accepted: 01/23/2020] [Indexed: 02/07/2023]
Abstract
Respiratory syncytial virus (RSV) infects a majority of infants and can cause severe disease leading to increased risk to develop asthma later in life. In the present studies we detected high levels of uric acid pathway components during RSV infection and examined whether they altered the pathogenesis of RSV infection. Inhibition of uric acid (UA) pathway activation during RSV infection in airway epithelial cells using XOI decreased the expression of IL-33, thymic stromal lymphopoietin (TSLP), and CCL2. In addition, treatment of RSV infected bone marrow-derived macrophages with XOI decreased production of IL-1β. Thus, UA activation of different cell populations contributes different innate immune mediators that promote immunopathogenesis. When mice were treated with XOI or interleukin-1 receptor antagonist (IL1-ra) during RSV infection decreased pulmonary mucus was observed along with significantly reduced numbers of ILC2 and macrophages, accompanied by decreased IL-33 in bronchoalveolar lavage of the treated mice. These findings provide mechanistic insight into the development of RSV immunopathology and indicate that xanthine metabolites and UA are key immunoregulator molecules during RSV infection. Moreover, these findings suggest uric acid and IL-1β as possible therapeutic targets to attenuate severe RSV disease.
Collapse
Affiliation(s)
- Wendy Fonseca
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA 48109
| | | | - Charles F. Schuler
- Department of Internal Medicine, Division of Allergy and Clinical Immunology, University of Michigan, Ann Arbor, MI, USA 48109
| | - Shannon K.K. Best
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA 48109
| | - Andrew J Rasky
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA 48109
| | - Susan B Morris
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA 48109
| | - Tracy X. Cui
- Division of Pediatric Pulmonology, University of Michigan, Ann Arbor, MI, USA 48109
| | - Antonia P. Popova
- Division of Pediatric Pulmonology, University of Michigan, Ann Arbor, MI, USA 48109
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA. .,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
130
|
Mortazavi Moghaddam SG, Namaei MH, Eslami Manoochehri R, Zardast M. The sequential assay of interleukin-10 and 13 serum levels in relation to radiographic changes during pulmonary tuberculosis treatment. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2020; 25:63. [PMID: 33088300 PMCID: PMC7554419 DOI: 10.4103/jrms.jrms_116_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 11/15/2019] [Accepted: 02/26/2020] [Indexed: 11/22/2022]
Abstract
Background: We evaluated the sequential changes of interleukin (IL)-10 and IL-13 serum levels with tuberculosis (TB)-related radiographic changes during pulmonary TB (PTB) treatment. Materials and Methods: In this cross-sectional study during two consecutive years, forty cases with PTB were recorded, and finally, 24 cases were completed the study. Serum levels of IL-10 and IL-13 were measured on admission time, and 6 months later. Furthermore, chest radiography was performed on admission and 6 months later in the treatment course. Results: Radiography at the baseline indicated pulmonary infiltration in all patients (n = 24). Fifteen (62.5%) cases had abnormal and 9 (37.5%) cases had normal radiography at the end of 6 months treatment course. IL-10 and IL-13 upregulated during the treatment time course, and their relationship with radiographic changes shifted from negative (r = −0.14 and P = 0.71) on admission to positive (r = 0.80 and P < 0.001) at the end of 6 months treatment course in normal radiography group. IL-10 level at the start of the treatment was 121.90 ± 88.81 in patients with normal and 82.68 ± 41.50 in patients with abnormal radiography (P = 0.31). Conclusion: Sequential increase in IL-10 and IL-13 during PTB treatment course may have a role in clearing the TB-related radiographic infiltration and preventing scar formation.
Collapse
Affiliation(s)
- Sayyed Gholamreza Mortazavi Moghaddam
- Department of Internal Medicine, Division of Pulmonary, School of Medicine, Vali-e-Asr Hospital, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Hasan Namaei
- Infectius Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Reza Eslami Manoochehri
- Department of Internal Medicine, School of Medicine, Vali-e-Asr Hospital, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahmood Zardast
- Department of Pathology, School of Medicine, Vali-e-Asr Hospital, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
131
|
Dy ABC, Ledford JG. IL-13Rα2 Glycosylation Holds the Dance Card for Partnering with IL-13. Am J Respir Cell Mol Biol 2020; 63:277-278. [PMID: 32459972 PMCID: PMC7462345 DOI: 10.1165/rcmb.2020-0165ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Alane B C Dy
- Department of Cellular and Molecular Medicine University of Arizona Tucson, Arizona
| | - Julie G Ledford
- Department of Cellular and Molecular Medicine University of Arizona Tucson, Arizona
| |
Collapse
|
132
|
Hashem T, Kammala AK, Thaxton K, Griffin RM, Mullany K, Panettieri RA, Subramanian H, Das R. CD2 Regulates Pathogenesis of Asthma Induced by House Dust Mice Extract. Front Immunol 2020; 11:881. [PMID: 32477356 PMCID: PMC7235426 DOI: 10.3389/fimmu.2020.00881] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/16/2020] [Indexed: 01/13/2023] Open
Abstract
Characteristic of allergic asthma, CD4+Th2 lymphocytes secrete Th2 cytokines, interleukin (IL)-4, IL-13, and IL-5 that mediate the inflammatory immune response. Surface expression of CD2 and its ligand, CD58, is increased on the monocytes and eosinophils of asthma patients, which correlate with elevated serum IgE levels, suggesting that CD2 may contribute to allergic airway inflammation. Using a murine model of asthma, we observed that house dust mice extract (HDME)-exposed Balb/c mice have increased airway hyperresponsiveness (AHR), lung inflammation, goblet cell hyperplasia, and elevated levels of Th2 cytokines in the lungs, as well as increased serum IgE levels as compared to the control mice. In contrast, with the exception of serum IgE levels, all the other parameters were significantly reduced in HDME-treated Cd2 -/- mice. Interestingly, Il13 but not Il4 or Il5 gene expression in the lungs was dramatically decreased in HDME-exposed Cd2 -/- mice. Of note, the gene expression of IL-13 downstream targets (Muc5b and Muc5ac) and high affinity IL-13Rα2 were upregulated in the lungs of HDME-exposed Balb/c mice but were significantly reduced in HDME-exposed Cd2 -/- mice. Consistently, gene expression of microRNAs regulating mucin production, inflammation, airway smooth muscle cell proliferation and IL-13 transcripts were increased in the lungs of HDME-exposed Cd2 -/- mice. Given the established role of IL-13 in promoting goblet cell hyperplasia, lung inflammation and AHR in allergic asthma, our studies reveal a unique role for CD2 in the regulation of Th2-associated allergic asthma.
Collapse
Affiliation(s)
- Tanwir Hashem
- Department of Physiology, College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Ananth K Kammala
- College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Kanedra Thaxton
- Department of Physiology, College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Ryan M Griffin
- Department of Physiology, College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Kellie Mullany
- Department of Chemical Engineering and Material Science, College of Engineering, Michigan State University, East Lansing, MI, United States
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, New Brunswick, NJ, United States
| | - Hariharan Subramanian
- College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Rupali Das
- College of Human Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
133
|
Kim TM, Paudel KR, Kim DW. Eriobotrya japonica leaf extract attenuates airway inflammation in ovalbumin-induced mice model of asthma. JOURNAL OF ETHNOPHARMACOLOGY 2020; 253:112082. [PMID: 31310829 DOI: 10.1016/j.jep.2019.112082] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 06/25/2019] [Accepted: 07/11/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Eriobotrya japonica leaves has a very long history of medicinal use as an anti-inflammatory and antitussive agent for bronchial inflammation. AIM OF THE STUDY The aim of this study was to evaluate the anti-inflammatory activities of Eriobotrya japonica (EJ) leaf water extract in an ovalbumin (OVA)-induced murine asthma model and human tracheal smooth muscle cell (HTSMC). MATERIALS AND METHODS Mice were sensitized by intra peritoneal OVA and challenged with nebulized OVA. EJ extract was administered orally at various dose. Bronchoalveolar lavage fluid (BALF) was quantified for nitric oxide (NO), eosinophil peroxidase (EPO), interleukin (IL)-4, IL-13 level and immunoglobulin (Ig) E was quantified in serum. Lung tissue sections were stained with hematoxylin and eosin for assessment of inflammatory cell infiltration whereas mucus production and goblet cell hyperplasia were studied by periodic acid schiff staining. Western blot was done for analysis of pERK1/2 expression and NFκB translocation in HTSMC whereas iNOS and COX-2 expression in RAW264.7 cell. RESULTS EJ significantly reduced the levels of BALF's NO, EPO, MMPs, IL-4, IL-13, and serum IgE. It also decreases inflammatory cell infiltration and mucus production. EJ also attenuated the proliferation of HTSMC, inhibits overexpression of ERK 1/2 and translocation of NFκB in HTSMC as well as iNOS and COX-2 expression in RAW 264.7 cell. CONCLUSION Present study suggest that, EJ effectively protects against allergic airway inflammation thus possessing potential therapeutic option for allergic asthma management.
Collapse
Affiliation(s)
- Tae-Muk Kim
- Department of Oriental Medicine Resources, Mokpo National University, Muan-gun, 58554, Republic of Korea
| | - Keshav Raj Paudel
- Department of Oriental Medicine Resources, Mokpo National University, Muan-gun, 58554, Republic of Korea
| | - Dong-Wook Kim
- Department of Oriental Medicine Resources, Mokpo National University, Muan-gun, 58554, Republic of Korea.
| |
Collapse
|
134
|
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: 7.3] [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
|
135
|
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: 7.8] [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
|
136
|
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: 4.8] [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
|
137
|
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: 34] [Impact Index Per Article: 8.5] [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
|
138
|
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: 8] [Impact Index Per Article: 2.0] [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
|
139
|
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: 1.5] [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
|
140
|
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: 4.8] [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
|
141
|
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: 0.3] [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
|
142
|
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: 11] [Impact Index Per Article: 2.8] [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
|
143
|
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: 5.5] [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
|
144
|
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. [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] [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
|
145
|
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: 3.0] [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
|
146
|
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: 1.6] [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
|
147
|
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: 180] [Impact Index Per Article: 36.0] [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
|
148
|
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: 96] [Impact Index Per Article: 19.2] [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
|
149
|
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: 1.4] [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
|
150
|
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: 1.6] [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
|