1
|
Celik E, Kocacik Uygun D, Kaya MA, Gungoren MS, Keven A, Bingol A. Aspergillus-sensitized asthma in children. Pediatr Allergy Immunol 2024; 35:e14212. [PMID: 39099328 DOI: 10.1111/pai.14212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Asthma is the most common chronic respiratory disease in childhood. Aspergillus fumigatus sensitivity may be involved in the pathogenesis of asthma by leading to different clinical presentations. OBJECTIVE To investigate the demographic, clinical, laboratory, and radiological characteristics of A. fumigatus sensitivity in childhood asthma and identify associated risk factors and diagnostic parameters. METHODS A total of 259 children with asthma were included in the study, 7 (2.7%) with allergic bronchopulmonary aspergillosis (ABPA), 84 (32.4%) with A. fumigatus-sensitized asthma (Af-SA), and 168 (64.9%) with A. fumigatus-unsensitized asthma (Af-UA). RESULTS Aspergillus sensitivity was associated with early asthma onset and longer asthma duration. Total IgE level and asthma severity are highest in ABPA and higher in Af-SA. Absolute eosinophil count was higher, and FEV1 was lower in Af-SA and ABPA. Aspergillus fumigatus was associated with greater odds of being male (odds ratio [OR], 2.45), having atopic dermatitis (OR, 3.159), Alternaria sensitivity (OR, 10.37), and longer asthma duration (OR, 1.266). The best cut-off values for detecting A. fumigatus positivity were 363.5 IU/mL for total IgE and 455 cells/μL for absolute eosinophil count. In Af-SA compared to Af-UA, centrilobular nodules and peribronchial thickening were more common, and the bronchoarterial ratio was higher. CONCLUSIONS Aspergillus sensitivity is a strong allergic stimulus in asthma, leading to laboratory, structural, clinical, and functional consequences. Af-SA is a distinct asthma endotype independent of ABPA that is characterized by increased risk of severe clinical presentations and impaired lung function.
Collapse
Affiliation(s)
- Enes Celik
- Department of Pediatric Allergy-Immunology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Dilara Kocacik Uygun
- Department of Pediatric Allergy-Immunology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Mehmet Akif Kaya
- Department of Pediatric Allergy-Immunology, Akdeniz University School of Medicine, Antalya, Turkey
| | | | - Ayse Keven
- Department of Radiology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Aysen Bingol
- Department of Pediatric Allergy-Immunology, Akdeniz University School of Medicine, Antalya, Turkey
| |
Collapse
|
2
|
Asghar A, Khalid A, Baqar Z, Hussain N, Saleem MZ, Sairash, Rizwan K. An insights into emerging trends to control the threats of antimicrobial resistance (AMR): an address to public health risks. Arch Microbiol 2024; 206:72. [PMID: 38252323 DOI: 10.1007/s00203-023-03800-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024]
Abstract
Antimicrobial agents are used to treat microbial ailments, but increased use of antibiotics and exposure to infections in healthcare facilities and hospitals as well as the excessive and inappropriate use of antibiotics at the society level lead to the emergence of multidrug-resistant (MDR) bacteria. Antimicrobial resistance (AMR) is considered a public health concern and has rendered the treatment of different infections more challenging. The bacterial strains develop resistance against antimicrobial agents by limiting intracellular drug accumulation (increasing efflux or decreasing influx of antibiotics), modification and inactivation of drugs and its targets, enzymatic inhibition, and biofilm formation. However, the driving factors of AMR include the sociocultural and economic circumstances of a country, the use of falsified and substandard medicines, the use of antibiotics in farm animals, and food processing technologies. These factors make AMR one of the major menaces faced by mankind. In order to promote reciprocal learning, this article summarizes the current AMR situation in Pakistan and how it interacts with the health issues related to the COVID-19 pandemic. The COVID-19 pandemic aids in illuminating the possible long-term impacts of AMR, which are less immediate but not less severe since their measures and effects are equivalent. Impact on other sectors, including the health industry, the economy, and trade are also discussed. We conclude by summarizing the several approaches that could be used to address this issue.
Collapse
Affiliation(s)
- Ayesha Asghar
- School of Biochemistry and Biotechnology, University of the Punjab, Quaid-E-Azam Campus, Lahore, Pakistan
| | - Aneeza Khalid
- School of Biochemistry and Biotechnology, University of the Punjab, Quaid-E-Azam Campus, Lahore, Pakistan
| | - Zulqarnain Baqar
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Nazim Hussain
- Centre for Applied Molecular Biology (CAMB), University of the Punjab, Quaid-E-Azam Campus, Lahore, Pakistan.
| | - Muhammad Zafar Saleem
- Centre for Applied Molecular Biology (CAMB), University of the Punjab, Quaid-E-Azam Campus, Lahore, Pakistan
| | - Sairash
- Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Komal Rizwan
- Department of Chemistry, University of Sahiwal, Sahiwal, 57000, Pakistan.
| |
Collapse
|
3
|
Wilson NG, Hernandez-Leyva A, Rosen AL, Jaeger N, McDonough RT, Santiago-Borges J, Lint MA, Rosen TR, Tomera CP, Bacharier LB, Swamidass SJ, Kau AL. The gut microbiota of people with asthma influences lung inflammation in gnotobiotic mice. iScience 2023; 26:105991. [PMID: 36824270 PMCID: PMC9941210 DOI: 10.1016/j.isci.2023.105991] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/28/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
The gut microbiota in early childhood is linked to asthma risk, but may continue to affect older patients with asthma. Here, we profile the gut microbiota of 38 children (19 asthma, median age 8) and 57 adults (17 asthma, median age 28) by 16S rRNA sequencing and find individuals with asthma harbored compositional differences from healthy controls in both adults and children. We develop a model to aid the design of mechanistic experiments in gnotobiotic mice and show enterotoxigenic Bacteroides fragilis (ETBF) is more prevalent in the gut microbiota of patients with asthma compared to healthy controls. In mice, ETBF, modulated by community context, can increase oxidative stress in the lungs during allergic airway inflammation (AAI). Our results provide evidence that ETBF affects the phenotype of airway inflammation in a subset of patients with asthma which suggests that therapies targeting the gut microbiota may be helpful tools for asthma control.
Collapse
Affiliation(s)
- Naomi G. Wilson
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ariel Hernandez-Leyva
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anne L. Rosen
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Natalia Jaeger
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ryan T. McDonough
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jesus Santiago-Borges
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael A. Lint
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thomas R. Rosen
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christopher P. Tomera
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Leonard B. Bacharier
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Monroe Carell Jr Children’s Hospital at Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - S. Joshua Swamidass
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew L. Kau
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
4
|
Asri AK, Liu T, Tsai HJ, Lee HY, Pan WC, Wu CD, Wang JY. Residential greenness and air pollution's association with nasal microbiota among asthmatic children. ENVIRONMENTAL RESEARCH 2023; 219:115095. [PMID: 36535395 DOI: 10.1016/j.envres.2022.115095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/05/2022] [Accepted: 12/15/2022] [Indexed: 06/17/2023]
Abstract
Both greenness and air pollution have widely been linked with asthma. However, the potential mechanism has rarely been investigated. This study aimed to identify the association between residential greenness and air pollution (fine particulate matter [PM2.5]; nitrogen dioxide [NO2]; ozone [O3]) with nasal microbiota among asthmatic children during the recovery phase. The normalized difference vegetation index was used to assess the extent of residential greenness. Spatiotemporal air pollution variation was estimated using an integrated hybrid kriging-LUR with the XG-Boost algorithm. These exposures were measured in 250-m intervals for four incremental buffer ranges. Nasal microbiota was collected from 47 children during the recovery phase. A generalized additive model controlled for various covariates was applied to evaluate the exposure-outcome association. The lag-time effect of greenness and air pollution related to the nasal microbiota also was examined. A significant negative association was observed between short-term exposure to air pollution and nasal bacterial diversity, as a one-unit increment in PM2.5 or O3 significantly decreased the observed species (PM2.5: -0.59, 95%CI -1.13, -0.05 and O3: -0.93, 95%CI -1.54, -0.32) and species richness (PM2.5: -0.64, 95%CI -1.25, -0.02 and O3: -0.68, 95%CI -1.43, -0.07). Considering the lag-time effect, we found a significant positive association between greenness and both the observed species and species richness. In addition, we identified a significant negative association for all pollutants with the observed species richness. These findings add to the evidence base of the links between nasal microbiota and air pollution and greenness. This study establishes a foundation for future studies of how environmental exposure plays a role in nasal microbiota, which in turn may affect the development of asthma.
Collapse
Affiliation(s)
- Aji Kusumaning Asri
- Department of Geomatics, National Cheng Kung University, Tainan, 701, Taiwan.
| | - Tsunglin Liu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan.
| | - Hui-Ju Tsai
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, 350, Taiwan.
| | - Hsiao-Yun Lee
- Department of Leisure Industry and Health Promotion, National Taipei University of Nursing and Health Sciences, Taipei, 112, Taiwan.
| | - Wen-Chi Pan
- Institute of Environmental and Occupational Health Sciences, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan.
| | - Chih-Da Wu
- Department of Geomatics, National Cheng Kung University, Tainan, 701, Taiwan; National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, 35053, Taiwan.
| | - Jiu-Yao Wang
- Department of Pediatrics, National Cheng Kung University, Tainan, 701, Taiwan; Allergy, Immunology, and Microbiome (A.I.M.) Research Center, China Medical University, Taichung, 404, Taiwan.
| |
Collapse
|
5
|
Tuazon JA, Kilburg-Basnyat B, Oldfield LM, Wiscovitch-Russo R, Dunigan-Russell K, Fedulov AV, Oestreich KJ, Gowdy KM. Emerging Insights into the Impact of Air Pollution on Immune-Mediated Asthma Pathogenesis. Curr Allergy Asthma Rep 2022; 22:77-92. [PMID: 35394608 PMCID: PMC9246904 DOI: 10.1007/s11882-022-01034-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Increases in ambient levels of air pollutants have been linked to lung inflammation and remodeling, processes that lead to the development and exacerbation of allergic asthma. Conventional research has focused on the role of CD4+ T helper 2 (TH2) cells in the pathogenesis of air pollution-induced asthma. However, much work in the past decade has uncovered an array of air pollution-induced non-TH2 immune mechanisms that contribute to allergic airway inflammation and disease. RECENT FINDINGS In this article, we review current research demonstrating the connection between common air pollutants and their downstream effects on non-TH2 immune responses emerging as key players in asthma, including PRRs, ILCs, and non-TH2 T cell subsets. We also discuss the proposed mechanisms by which air pollution increases immune-mediated asthma risk, including pre-existing genetic risk, epigenetic alterations in immune cells, and perturbation of the composition and function of the lung and gut microbiomes. Together, these studies reveal the multifaceted impacts of various air pollutants on innate and adaptive immune functions via genetic, epigenetic, and microbiome-based mechanisms that facilitate the induction and worsening of asthma.
Collapse
Affiliation(s)
- J A Tuazon
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, 43210, USA
| | - B Kilburg-Basnyat
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC, 27858, USA
| | - L M Oldfield
- Department of Synthetic Biology and Bioenergy, J. Craig Venter Institute, Rockville, MD, 20850, USA
- Department of Synthetic Genomics, Replay Holdings LLC, San Diego, 92121, USA
| | - R Wiscovitch-Russo
- Department of Synthetic Biology and Bioenergy, J. Craig Venter Institute, Rockville, MD, 20850, USA
| | - K Dunigan-Russell
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, 43210, USA
| | - A V Fedulov
- Division of Surgical Research, Department of Surgery, Alpert Medical School, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
| | - K J Oestreich
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, The James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - K M Gowdy
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, 43210, USA.
| |
Collapse
|
6
|
Quan L, Ren G, Liu L, Huang W, Li M. Circular RNA circ_0002594 regulates PDGF-BB-induced proliferation and migration of human airway smooth muscle cells via sponging miR-139-5p/TRIM8 in asthma. Autoimmunity 2022; 55:339-350. [PMID: 35470728 DOI: 10.1080/08916934.2022.2062596] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Asthma is a chronic respiratory disease characterised by the contraction of smooth muscle and remodelling of the airway wall, which is correlated with increased airway smooth muscle mass. Circular RNA (circRNA) circ_0002594 has been reported as a pro-inflammatory factor in allergic asthma. Therefore, this study is designed to explore the role and mechanism of circ_0002594 in human airway smooth muscle cells (HASMC) proliferation and metastasis. METHODS Cell proliferative ability, invasion, and migration were detected by 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT), 5-ethynyl-2'-deoxyuridine (EdU), Transwell, and Wound healing assays. The protein levels of E-cadherin, N-cadherin, and tripartite motif 8 (TRIM8) were detected by western blot assay. The levels of interleukin-6 (IL-6) and IL-13 were detected using Enzyme-linked immunosorbent assays (ELISA). Levels of circ_0002594, microRNA-139-5p (miR-139-5p), TRIM8 were determined by real-time quantitative polymerase chain reaction (RT-qPCR). The binding between miR-139-5p and circ_0002594 or TRIM8 was predicted by Circinteractome or Starbase v2.0, and then verified by a dual-luciferase reporter and RNA Immunoprecipitation (RIP) assays. RESULTS Platelet-derived growth factor-BB (PDGF-BB) could trigger HASMC proliferation, metastasis, and inflammation. Circ_0002594 and TRIM8 were elevated in asthma patients and PDGF-BB-treated HASMC, and the miR-139-5p level was decreased. Furthermore, circ_0002594 knockdown could suppress PDGF-BB- stimulated HASMC damage. Mechanism analysis exhibited that circ_0002594 could regulate TRIM8 expression through sponging miR-139-5p. CONCLUSION Our findings revealed that circ_0002594 could act as a regulator in the airway remodelling during asthma development partly by the miR-139-5p/TRIM8 axis, hinting at an underlying therapeutic strategy for asthma.
Collapse
Affiliation(s)
- Liyuan Quan
- Department of Pediatrics, Qinghuangdao First Hospital, Qinhuangdao, China
| | - Gaowei Ren
- Department of Pediatrics, Qinghuangdao First Hospital, Qinhuangdao, China
| | - Ling Liu
- Department of Pediatrics, Qinghuangdao First Hospital, Qinhuangdao, China
| | - Wei Huang
- Department of Pediatrics, Qinghuangdao First Hospital, Qinhuangdao, China
| | - Mingli Li
- Department of Pediatrics, Qinghuangdao First Hospital, Qinhuangdao, China
| |
Collapse
|
7
|
Liu F, Zhang J, Zhang D, Qi Q, Cui W, Pan Y, Liu X, Xu J, Qiao X, Wang Z, Dong L. Follistatin-related protein 1 in asthma: miR-200b-3p interactions affect airway remodeling and inflammation phenotype. Int Immunopharmacol 2022; 109:108793. [PMID: 35483234 DOI: 10.1016/j.intimp.2022.108793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 11/05/2022]
Abstract
Follistatin-related protein 1 (FSTL1) is significantly associated with the asthma severity and outcome in humans and diverse mouse models of asthma. Previous studies have also suggested that FSTL1 could activate autophagy and NLRP3, thus playing as a causative agent in the asthma progression. However, mechanisms that regulate airway epithelial cell-specific FSTL1 expression and function in asthma are unknown. Here, we further evaluated the spatiotemporal relationships between the FSTL1 and asthma development through ovalbumin (OVA) -induced asthma models. Integrative analysis in asthmatics airway epithelium identifies microRNA (miR)-200b-3p as a novel upstream of FSTL1. Next, we collected airway biopsies, induced sputum, and blood samples isolated from asthmatics patients and the OVA-induced mouse model. We revealed that miR-200b-3p expression is downregulated in asthmatics airway epithelium, while its expression was negatively correlated with FSTL1. On this basis, the function and expression pattern analysis of miR-200b-3p were performed using miRNA-target prediction databases and long non-coding RNA (lncRNA) microarray assay. It is illustrated that miR-200b-3p, which is downregulated with pro-fibrotic stimulation of TGF-β1, could also be sponged by lncRNA PCAT19 and regulate FSTL1 expression in asthma progression. In vivo, miR-200b-3p overexpression in mice prevents OVA-induced airway remodeling and inflammation. Lastly, protective roles of miR-200b-3p are partly attributed to the direct and functional repression of FSTL1. Our findings suggest a crucial role for the miR-200b-3p/FSTL1 axis in regulating asthmatic's airway remodeling and inflammation phenotype.
Collapse
Affiliation(s)
- Fen Liu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Jintao Zhang
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dong Zhang
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qian Qi
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Wenjing Cui
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yun Pan
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaofei Liu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Jiawei Xu
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Xinrui Qiao
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zihan Wang
- Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Liang Dong
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Respiratory Diseases, Jinan, China; Department of Respiratory, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
8
|
Jonckheere AC, Seys SF, Steelant B, Decaesteker T, Dekoster K, Cremer J, Dilissen E, Schols D, Iwakura Y, Vande Velde G, Breynaert C, Schrijvers R, Vanoirbeek J, Ceuppens JL, Dupont LJ, Bullens DMA. Innate Lymphoid Cells Are Required to Induce Airway Hyperreactivity in a Murine Neutrophilic Asthma Model. Front Immunol 2022; 13:849155. [PMID: 35371094 PMCID: PMC8965562 DOI: 10.3389/fimmu.2022.849155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/14/2022] [Indexed: 12/04/2022] Open
Abstract
Rationale Non-allergic asthma is driven by multiple endotypes of which neutrophilic and pauci-granulocytic asthma have been best established. However, it is still puzzling what drives inflammation and airway hyperreactivity (AHR) in these patients and how it can be treated effectively. Recently, a potential role of the innate immune system and especially the innate lymphoid cells (ILC) has been proposed. Objective In this study, we investigated the effects of LPS inhalation on airway inflammation and AHR as a potential model for elucidating the pathogenesis of non-allergic asthma. Methods Wild-type (BALB/c), SCID, IL-17A-/-, and Rag2-/- γC-/- mice were endonasally exposed to lipopolysaccharide (LPS, 2 µg) on four consecutive days. Twenty-four hours after the last exposure, AHR to methacholine was assessed. Cytokine levels and ILC subpopulations were determined in lung tissue. Cellular differential analysis was performed in BAL fluid. Main Results In this study, we developed a murine model for non-allergic neutrophilic asthma. We found that repeated endonasal applications of low-dose LPS in BALB/c mice led to AHR, BAL neutrophilia, and a significant increase in lung ILC3 as well as a significant increase in lung chemokines KC and MIP-2 and cytokines IL-1β, IL-17A, IL-22, and TNF. The adoptive transfer of ILC in Rag2-/- γC-/- mice showed that ILC played a causal role in the induction of AHR in this model. Antagonising IL-1β, but not IL-17A or neutrophils, resulted in a partial reduction in LPS-induced AHR. Conclusion In conclusion, we report here a murine model for neutrophilic asthma where ILC are required to induce airway hyperreactivity.
Collapse
Affiliation(s)
- Anne-Charlotte Jonckheere
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Sven F Seys
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Brecht Steelant
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Tatjana Decaesteker
- Department of Chronic Diseases, Metabolism and Ageing, Laboratory of Respiratory Diseases and Thoracic Surgery, KU Leuven, Leuven, Belgium
| | - Kaat Dekoster
- Department of Imaging and Pathology, Biomedical MRI Unit/Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, Leuven, Belgium
| | - Jonathan Cremer
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Ellen Dilissen
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Dominique Schols
- Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| | - Yoichiro Iwakura
- Centre for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Greetje Vande Velde
- Department of Imaging and Pathology, Biomedical MRI Unit/Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, Leuven, Belgium
| | - Christine Breynaert
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Rik Schrijvers
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Jeroen Vanoirbeek
- Department of Public Health and Primary Care, Centre for Environment and Health, KU Leuven, Leuven, Belgium
| | - Jan L Ceuppens
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Lieven J Dupont
- Department of Chronic Diseases, Metabolism and Ageing, Laboratory of Respiratory Diseases and Thoracic Surgery, KU Leuven, Leuven, Belgium.,Clinical Division of Respiratory Medicine, UZ Leuven, Leuven, Belgium
| | - Dominique M A Bullens
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium.,Clinical Division of Paediatrics, UZ Leuven, Leuven, Belgium
| |
Collapse
|
9
|
Li C, Deng C, Zhou T, Hu J, Dai B, Yi F, Tian N, Jiang L, Dong X, Zhu Q, Zhang S, Cui H, Cao L, Shang Y. MicroRNA-370 carried by M2 macrophage-derived exosomes alleviates asthma progression through inhibiting the FGF1/MAPK/STAT1 axis. Int J Biol Sci 2021; 17:1795-1807. [PMID: 33994863 PMCID: PMC8120458 DOI: 10.7150/ijbs.59715] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/12/2021] [Indexed: 12/22/2022] Open
Abstract
Emerging evidence has suggested the functions of exosomes in allergic diseases including asthma. By using a mouse model with asthma induced by ovalbumin (OVA), we explored the roles of M2 macrophage-derived exosomes (M2Φ-Exos) in asthma progression. M2Φ-Exos significantly alleviated OVA-induced fibrosis and inflammatory responses in mouse lung tissues, as well as inhibited abnormal proliferation, invasion, and fibrosis-related protein production in platelet derived growth factor (PDGF-BB) treated primary mouse airway smooth muscle cells (ASMCs). The OVA administration in mice or the PDGF-BB treatment in ASMCs reduced the expression of miR-370, which was detected in M2Φ-Exos by miRNA sequencing. However, treating the mice or ASMCs with M2Φ-Exos reversed the inhibitory effect of OVA or PDGF-BB on miR-370 expression. We identified that the target of miR-370 was fibroblast growth factor 1 (FGF1). Downregulation of miR-370 by Lv-miR-370 inhibitor or overexpression of FGF1 by Lv-FGF1 blocked the protective roles of M2Φ-Exos in asthma-like mouse and cell models. M2Φ-Exos were found to inactivate the MAPK signaling pathway, which was recovered by miR-370 inhibition or FGF1 overexpression. Collectively, we conclude that M2Φ-Exos carry miR-370 to alleviate asthma progression through downregulating FGF1 expression and the MAPK/STAT1 signaling pathway. Our study may offer a novel insight into asthma treatment.
Collapse
Affiliation(s)
- Chunlu Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Chengsi Deng
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Tingting Zhou
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Jiapeng Hu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Bing Dai
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Fei Yi
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Na Tian
- Jilin Tuohua Biotechnology Co., Ltd. Changchun, Jilin 13000, China
| | - Lijun Jiang
- Jilin Tuohua Biotechnology Co., Ltd. Changchun, Jilin 13000, China
| | - Xiang Dong
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Qingfeng Zhu
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Siyi Zhang
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Hongyan Cui
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Liu Cao
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
10
|
Wang WL, Luo XM, Zhang Q, Zhu HQ, Chen GQ, Zhou Q. The lncRNA PVT1/miR-590-5p/FSTL1 axis modulates the proliferation and migration of airway smooth muscle cells in asthma. Autoimmunity 2021; 54:138-147. [PMID: 33825599 DOI: 10.1080/08916934.2021.1897977] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Asthma is a prevalent chronic inflammatory airway disease that is characterised by airway remodelling and airway hyperresponsiveness. Abnormal proliferation and migration of airway smooth muscle cells (ASMCs) contribute to airway remodelling in asthma. However, the molecular mechanism underlying an increased ASMC mass in asthma remains elusive. Herein, we aimed at investigating the regulation of lncRNA PVT1 on ASMCs and focussing on the mechanism in the proliferation and migration. METHODS Expression levels of lncRNA PVT1 and miR-590-5p in the serum collected from 24 children with asthma and 10 control children were determined by qRT-PCR. ASMCs proliferation and migration prior to and post platelet-derived growth factor subunit B (PDGF-BB) stimulation were examined by CCK-8 test and transwell assay. Dual-luciferase reporter assay was performed to determine miR-590-5p interaction with lncRNA PVT1 and follistatin-like 1 (FSTL1). Expression of lncRNA PVT1, miR-590-5p, FSTL1, C-Myc, cyclin D1, and cyclin-dependent kinase 1 (CDK1) was tested by quantitative real-time PCR (qRT-PCR) and immunoblotting analysis. RESULTS The expression level of lncRNA PVT1 was higher but the expression level of miR-590-5p was lower in the serum of children with asthma than in control children. The expression level of lncRNA PVT1 was negatively correlated with the expression level of miR-590-5p in asthma. LncRNA PVT1 was upregulated upon PDGF-BB stimulation. LncRNA PVT1 knockdown by its specific shRNA repressed PDGF-BB-induced promotion of proliferation and migration in ASMCs and triggered an elevated miR-590-5p along with declined C-Myc, cyclin D1, and CDK1. The effects of lncRNA PVT1 knockdown on PDGF-BB-induced ASMCs were lost upon miR-590-5p inhibition. MiR-590-5p targeted FSTL1 gene and declined its expression, thus suppressing ASMC proliferation and migration following PDGF-BB stimulation and downregulating C-Myc, cyclin D1, and CDK1 expressions. The effects of miR-590-5p on PDGF-BB-induced ASMCs were lost upon FSTL1 overexpression. CONCLUSION These results support the notion that the lncRNA PVT1/miR-590-5p/FSTL1 axis modulates ASMCs proliferation and migration following PDGF-BB stimulation, providing a potential therapeutic target to attenuate airway remodelling in asthma.
Collapse
Affiliation(s)
- Wen-Lan Wang
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Xiao-Ming Luo
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Qin Zhang
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Hai-Qiao Zhu
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Guo-Qing Chen
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Qin Zhou
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| |
Collapse
|
11
|
Li C, Dai B, Hu J, Shang Y. WITHDRAWN: M2 macrophage-derived exosomes carry microRNA-370 to alleviate asthma progression through inhibiting the FGF1/MAPK/STAT1 axis. Exp Cell Res 2020:112285. [PMID: 32941809 DOI: 10.1016/j.yexcr.2020.112285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 01/16/2023]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Chunlu Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, P.R. China
| | - Bing Dai
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, P.R. China
| | - Jiapeng Hu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, P.R. China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, P.R. China
| |
Collapse
|
12
|
Rosser F, Han YY, Forno E, Acosta-Pérez E, Canino G, Celedón JC. Indoor endotoxin, proximity to a major roadway, and severe asthma exacerbations among children in Puerto Rico. Ann Allergy Asthma Immunol 2020; 125:658-664.e2. [PMID: 32911057 DOI: 10.1016/j.anai.2020.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/31/2020] [Accepted: 09/02/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Few studies have examined concurrent exposure to household endotoxin and traffic-related air pollution in relation to childhood asthma, yet both factors are associated with asthma outcomes. OBJECTIVE To examine whether proximity to a major roadway (a traffic-related air pollution proxy) modifies the estimated effects of indoor endotoxin on asthma outcomes in children. METHODS Cross-sectional study of 200 children with asthma (ages, 6-14 years) living in Puerto Rico. Residential distance to a major roadway was calculated as the distance from the participant's residential US census block centroid to the nearest major road. The outcomes of interest were severe asthma exacerbations, missed school days for asthma, atopy, lung function, and bronchodilator response (BDR). Logistic, linear, or negative binomial regression was used for the multivariable analysis. RESULTS In the multivariable analysis, there was an interaction between indoor endotoxin and residential distance to a roadway on severe asthma exacerbations (P = .02) and BDR (P = .07). In an analysis stratified by distance to a roadway, each log10-unit increase in endotoxin was associated with 4.21 times increased odds of severe asthma exacerbations among children living within 499 m (the lower 3 quartiles of residential distance) to a road (95% confidence interval, 1.5-12.0). Among subjects living further than 499 m away from a roadway, each log10-unit increase in endotoxin was associated with reduced odds of severe asthma exacerbations (odds ratio, 0.03; 95% confidence interval, 0.001-0.67). Similar but less substantial findings were observed for BDR. CONCLUSION Our findings suggest that residential proximity to a major road modifies the estimated effect of endotoxin on severe asthma exacerbations in children.
Collapse
Affiliation(s)
- Franziska Rosser
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yueh-Ying Han
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Erick Forno
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Edna Acosta-Pérez
- Department of Pediatrics, Behavioral Sciences Research Institute, University of Puerto Rico, San Juan, Puerto Rico
| | - Glorisa Canino
- Department of Pediatrics, Behavioral Sciences Research Institute, University of Puerto Rico, San Juan, Puerto Rico
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
13
|
Tiotiu AI, Novakova P, Nedeva D, Chong-Neto HJ, Novakova S, Steiropoulos P, Kowal K. Impact of Air Pollution on Asthma Outcomes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17176212. [PMID: 32867076 PMCID: PMC7503605 DOI: 10.3390/ijerph17176212] [Citation(s) in RCA: 220] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 12/15/2022]
Abstract
Asthma is a chronic respiratory disease characterized by variable airflow obstruction, bronchial hyperresponsiveness, and airway inflammation. Evidence suggests that air pollution has a negative impact on asthma outcomes in both adult and pediatric populations. The aim of this review is to summarize the current knowledge on the effect of various outdoor and indoor pollutants on asthma outcomes, their burden on its management, as well as to highlight the measures that could result in improved asthma outcomes. Traffic-related air pollution, nitrogen dioxide and second-hand smoking (SHS) exposures represent significant risk factors for asthma development in children. Nevertheless, a causal relation between air pollution and development of adult asthma is not clearly established. Exposure to outdoor pollutants can induce asthma symptoms, exacerbations and decreases in lung function. Active tobacco smoking is associated with poorer asthma control, while exposure to SHS increases the risk of asthma exacerbations, respiratory symptoms and healthcare utilization. Other indoor pollutants such as heating sources and molds can also negatively impact the course of asthma. Global measures, that aim to reduce exposure to air pollutants, are highly needed in order to improve the outcomes and management of adult and pediatric asthma in addition to the existing guidelines.
Collapse
Affiliation(s)
- Angelica I. Tiotiu
- Department of Pulmonology, University Hospital of Nancy, 54395 Nancy, France
- Development of Adaptation and Disadvantage, Cardiorespiratory Regulations and Motor Control (EA 3450 DevAH), University of Lorraine, 54395 Nancy, France
- Correspondence: ; Tel.: +33-383-154-299
| | - Plamena Novakova
- Clinic of Clinical Allergy, Medical University, 1000 Sofia, Bulgaria;
| | | | - Herberto Jose Chong-Neto
- Division of Allergy and Immunology, Department of Pediatrics, Federal University of Paraná, Curitiba 80000-000, Brazil;
| | - Silviya Novakova
- Allergy Unit, Internal Consulting Department, University Hospital “St. George”, 4000 Plovdiv, Bulgaria;
| | - Paschalis Steiropoulos
- Department of Respiratory Medicine, Medical School, Democritus University of Thrace, University General Hospital Dragana, 68100 Alexandroupolis, Greece;
| | - Krzysztof Kowal
- Department of Allergology and Internal Medicine, Medical University of Bialystok, 15-037 Bialystok, Poland;
| |
Collapse
|
14
|
Gehring U, Wijga AH, Koppelman GH, Vonk JM, Smit HA, Brunekreef B. House dust endotoxin, asthma and allergic sensitization through childhood into adolescence. Clin Exp Allergy 2020; 50:1055-1064. [PMID: 32640057 PMCID: PMC7540017 DOI: 10.1111/cea.13705] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/28/2020] [Accepted: 07/02/2020] [Indexed: 12/17/2022]
Abstract
Background House dust endotoxin may have beneficial effects on allergic sensitization and asthma in children. Evidence is scarce for adolescents and most studies so far have been cross‐sectional and limited to a single exposure measurement. Objective We assessed associations of house dust endotoxin with asthma and allergic sensitization from birth to age 17 years longitudinally taking into account exposure early in life and at primary school age. Methods We used data of 854 participants of the prospective Dutch PIAMA birth cohort study with house dust endotoxin measurements at 3 months and/or 5‐6 years and data on asthma and/or allergic sensitization from at least one of 11 follow‐ups until age 17. We assessed overall and age‐specific associations of the prevalence of asthma and sensitization with mattress and living room floor dust concentrations (per gram of dust) and loads (per m2 of sampling surface). Results Higher living room floor dust endotoxin concentrations at 3 months were associated with lower odds of asthma until age 4 [odds ratio (95% confidence interval) ranging from 0.70 (0.51‐0.97) at age 1 to 0.76 (0.57‐1.00) at age 3 per interquartile range increase], but not thereafter in children of allergic mothers. Higher living room floor dust endotoxin at 5‐6 years was associated with higher odds of sensitization at 8‐16 years [eg odds ratio (95% confidence interval) 1.70 (1.17‐2.47) per interquartile range increase in endotoxin load]. Conclusions and clinical relevance House dust endotoxin may have beneficial effects on asthma in preschool children of allergic mothers, which do not persist into adolescence. Beneficial associations with allergic sensitization could not be confirmed.
Collapse
Affiliation(s)
- Ulrike Gehring
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Alet H Wijga
- Center for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands
| | - Judith M Vonk
- Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands.,Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Henriëtte A Smit
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Bert Brunekreef
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands.,Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|