1
|
Adams TJ, Schuliga M, Pearce N, Bartlett NW, Liang M. Targeting respiratory virus-induced reactive oxygen species in airways diseases. Eur Respir Rev 2025; 34:240169. [PMID: 40240057 PMCID: PMC12000908 DOI: 10.1183/16000617.0169-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 02/02/2025] [Indexed: 04/18/2025] Open
Abstract
The immune response to virus infection in the respiratory tract must be carefully balanced to achieve pathogen clearance without excessive immunopathology. For chronic respiratory diseases where there is ongoing inflammation, such as in asthma and COPD, airway immune balance is perturbed, and viral infection frequently worsens (exacerbates) these conditions. Reactive oxygen species (ROS) are critical to the induction and propagation of inflammation, and when appropriately regulated, ROS are vital cell signalling molecules and contribute to innate immunity. However, extended periods of high ROS concentration can cause excessive cellular damage that dysregulates antiviral immunity and promotes inflammation. Traditional antioxidant therapeutics have had limited success treating inflammatory diseases such as viral exacerbations of asthma or COPD, owing to nonspecific pharmacology and poorly understood pharmacokinetic properties. These drawbacks could be addressed with novel drug delivery technologies and pharmacological agents. This review summarises current research on ROS imbalances during virus infection, discusses the commercially available mitochondrial antioxidant drugs that have progressed to clinical trial and assesses novel drug delivery approaches for antioxidant delivery to the airways. Additionally, it provides a perspective on future research into pharmacological targeting of ROS for the treatment of respiratory virus infection and disease.
Collapse
Affiliation(s)
- Thomas J Adams
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Infection Research Program, Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Michael Schuliga
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Infection Research Program, Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Nyoaki Pearce
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Infection Research Program, Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Nathan W Bartlett
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Infection Research Program, Hunter Medical Research Institute (HMRI), New Lambton Heights, Australia
| | - Mingtao Liang
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
| |
Collapse
|
2
|
Zhang M, Qin Z, Huang C, Liang B, Zhang X, Sun W. The gut microbiota modulates airway inflammation in allergic asthma through the gut-lung axis related immune modulation: A review. BIOMOLECULES & BIOMEDICINE 2025; 25:727-738. [PMID: 39465678 PMCID: PMC11959394 DOI: 10.17305/bb.2024.11280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
The human gut microbiota is a vast and complex microbial community. According to statistics, the number of bacteria residing in the human intestinal tract is approximately ten times that of total human cells, with over 1000 different species. The interaction between the gut microbiota and various organ tissues plays a crucial role in the pathogenesis of local and systemic diseases, exerting a significant influence on disease progression. The relationship between the gut microbiota and intestinal diseases, along with its connection to the pulmonary immune environment and the development of lung diseases, is commonly referred to as the "gut-lung axis." The incidence of bronchial asthma is rising globally. With ongoing research on gut microbiota, it is widely believed that intestinal microorganisms and their metabolic products directly or indirectly participate in the occurrence and development of asthma. Based on the gut-lung axis, this review examines recent research suggesting that the intestinal microbiota can influence the occurrence and progression of allergic asthma through the modulation of cytokine immune balance and mucosal integrity. Though the precise immune pathways or microbial species influencing asthma through the gut-lung axis are still under exploration, summarizing the immune modulation through the gut-lung axis in allergic asthma may provide insights for the clinical management of the condition.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Gastroenterology, People’s Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Ziwen Qin
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Chuanjun Huang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Bin Liang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Xiuqing Zhang
- Department of Radiology, Dongying City Dongying District People’s Hospital, Dongying, Shandong Province, China
| | - Weitao Sun
- Department of Respiratory Medicine, Dongying City Dongying District People’s Hospital, Dongying, Shandong Province, China
| |
Collapse
|
3
|
Liew KY, Chee HY, Abas F, Leong SW, Harith HH, Israf DA, Sulaiman MR, Tham CL. A synthetic curcumin-like diarylpentanoid analog inhibits rhinovirus infection in H1 hela cells via multiple antiviral mechanisms. Daru 2024; 32:729-744. [PMID: 39395148 PMCID: PMC11554966 DOI: 10.1007/s40199-024-00542-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/19/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Rhinovirus (RV) infection is a major cause of common colds and asthma exacerbations, with no antiviral drug available. Curcumin exhibits broad-spectrum antiviral activities, but its therapeutic effect is limited by a poor pharmacokinetics profile. Curcumin-like diarylpentanoid analogs, particularly 2-benzoyl-6-(3,4-dihydroxybenzylidene)cyclohexen-1-ol (BDHBC) and 5-(3,4-dihydroxyphenyl)-3-hydroxy-1-(2-hydroxyphenyl)penta-2,4-dien-1-one (DHHPD), have better solubility and stability compared to curcumin. OBJECTIVES Therefore, this study aims to evaluate and compare the antiviral effects of curcumin, BDHBC, and DHHPD in an in vitro model of RV infection. METHODS The inhibitory effects on RV-16 infection in H1 HeLa cells were assessed using cytopathic effect (CPE) reduction assay, virus yield reduction assay, RT-qPCR, and Western blot. Antiviral effects in different modes of treatment (pre-, co-, and post-treatment) were also compared. Additionally, intercellular adhesion molecule 1 (ICAM-1) expression, RV binding, and infectivity were measured with Western blot, flow cytometry, and virucidal assay, respectively. RESULTS When used as a post-treatment, BDHBC (EC50: 4.19 µM; SI: 8.32) demonstrated stronger antiviral potential on RV-16 compared to DHHPD (EC50: 18.24 µM; SI: 1.82) and curcumin (less than 50% inhibition). BDHBC also showed the strongest inhibitory effect on RV-induced CPE, virus yield, vRNA, and viral proteins (P1, VP0, and VP2). Furthermore, BDHBC pre-treatment has a prophylactic effect against RV infection, which was attributed to reduced basal expression of ICAM-1. However, it did not affect virus binding, but exerted virucidal activity on RV-16, contributing to its antiviral effect during co-treatment. CONCLUSION BDHBC exhibits multiple antiviral mechanisms against RV infection and thus could be a potential antiviral agent for RV.
Collapse
Affiliation(s)
- Kong Yen Liew
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hui-Yee Chee
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Faridah Abas
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Sze Wei Leong
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Hanis Hazeera Harith
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Daud Ahmad Israf
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Mohd Roslan Sulaiman
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| |
Collapse
|
4
|
Yuan Y, Zhu H, Huang S, Zhang Y, Shen Y. Establishment of a diagnostic model based on immune-related genes in children with asthma. Heliyon 2024; 10:e25735. [PMID: 38375253 PMCID: PMC10875436 DOI: 10.1016/j.heliyon.2024.e25735] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/21/2024] Open
Abstract
Objective Allergic asthma is driven by an antigen-specific immune response. This study aimed to identify immune-related differentially expressed genes in childhood asthma and establish a classification diagnostic model based on these genes. Methods GSE65204 and GSE19187 were downloaded and served as training set and validation set. The immune cell composition was evaluated with ssGSEA algorithm based on the immune-related gene set. Modules that significantly related to the asthma were selected by WGCNA algorithm. The immune-related differentially expressed genes (DE-IRGs) were screened, the protein-protein interaction network and diagnostic model of DE-IRGs was constructed. The pathway and immune correlation analysis of hub DE-IRGs was analyzed. Results Eight immune cell types exhibited varying levels of abundance between the asthma and control groups. A total of 112 differentially expressed immune-related genes (DE-IRGs) was identified. Through the application of four ranking methods (MCC, MNC, DEGREE, and EPC), 17 hub DE-IRGs with overlapping significance were further selected. Subsequently, 8 optimized were identified using univariate logistic regression analysis and the LASSO regression algorithm, based on which a robust diagnostic model was constructed. Notably, TNF and CD40LG emerged as direct participants in asthma-related signaling pathways, displaying a positive correlation with the immune cell types of immature B cells, activated B cells, activated CD8 T cells, activated CD4 T cells, and myeloid-derived suppressor cells. Conclusion The diagnostic model constructed using the DE-IRGs (CCL5, CCR5, CD40LG, CD8A, IL2RB, PDCD1, TNF, and ZAP70) exhibited high and specific diagnostic value for childhood asthma. The diagnostic model may contribute to the diagnosis of childhood asthma.
Collapse
Affiliation(s)
- Yuyun Yuan
- Department of Pediatrics, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, 201999, China
| | - Honghua Zhu
- Department of Medical Imaging, Shanghai Seventh People's Hospital, Shanghai, 200137, China
| | - Sihong Huang
- Department of Pediatrics, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, 201999, China
| | - Yantao Zhang
- Department of Pediatrics, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, 201999, China
| | - Yiyun Shen
- Department of Pediatrics, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, 201999, China
| |
Collapse
|
5
|
Khojasteh-Kaffash S, Parhizkar Roudsari P, Ghaffari Jolfayi A, Samieefar N, Rezaei N. Pediatric asthma exacerbation and COVID-19 pandemic: Impacts, challenges, and future considerations. J Asthma 2024; 61:81-91. [PMID: 37610180 DOI: 10.1080/02770903.2023.2251062] [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: 06/29/2023] [Revised: 08/11/2023] [Accepted: 08/19/2023] [Indexed: 08/24/2023]
Abstract
OBJECTIVE Asthma, a common disease among children and adolescents, poses a great health risk when ignored; therefore, a thorough follow-up to prevent exacerbations is emphasized. The aim of the present study is to investigate asthma exacerbation in children during the Coronavirus disease 2019 (COVID-19) era. DATA SOURCES This narrative review has been done by searching the PubMed and Embase databases using Asthma, COVID-19, Pandemic, and Symptom flare up as keywords. STUDY SELECTIONS Studies related to asthma exacerbation in COVID-19 pandemic were included. RESULTS Based on studies, controlled or mild to moderate asthma has not been considered a risk factor for COVID-19 severity and has not affected hospitalization, intensive care unit (ICU) admission, and mortality. Surprisingly, emergent and non-emergent visits and asthmatic attacks decreased during the pandemic. The three main reasons for decreased incidence and exacerbation of asthma episodes in the COVID-19 era included reduced exposure to environmental allergens, increasing the acceptance of treatment by pediatrics and caregivers, and decreased risk of other respiratory viral infections. Based on the available studies, COVID-19 vaccination had no serious side effects, except in cases of uncontrolled severe asthma, and can be injected in these children. Also, there was no conclusive evidence of asthma exacerbation after the injection of COVID-19 vaccines. CONCLUSION Further studies are recommended to follow the pattern of asthma in the post-pandemic situation and to become prepared for similar future conditions.
Collapse
Affiliation(s)
- Soroush Khojasteh-Kaffash
- Student Research Committee, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Peyvand Parhizkar Roudsari
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ghaffari Jolfayi
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Cardiovascular Research Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Noosha Samieefar
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
6
|
Wang H, Tao F, Li CY, Yang GJ, Chen J. Short-term administration of Qipian®, a mixed bacterial lysate, inhibits airway inflammation in ovalbumin-induced mouse asthma by modulating cellular, humoral and neurogenic immune responses. Life Sci 2024; 336:122310. [PMID: 38013140 DOI: 10.1016/j.lfs.2023.122310] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023]
Abstract
AIMS Qipian® is a commercialized agent composed of extracts of three genera of commensal bacteria, and its mechanism of action on asthma is unclear. This study aimed to examine the impact of Qipian® on airway inflammation and investigate the underlying mechanisms. MATERIALS AND METHODS Qipian® or dexamethasone (DEX) was administered before OVA challenge in an ovalbumin-induced asthma mouse model, and then asthmatic symptoms were observed and scored. Samples of lung tissues, blood, and bronchoalveolar lavage fluid (BALF) were collected, and eosinophils (Eos), immunoglobins (Igs), and type 1 T helper (Th1)/Th2 cell cytokines were measured. Mucus production in the lung was assessed by periodic acid-Schiff (PAS) staining. The effects of Qipian® on dendritic and T regulatory (Treg) cells were investigated using flow cytometry. KEY FINDINGS The short-term administration of Qipian® significantly inhibited the cardinal features of allergic asthma, including an elevated asthmatic behaviour score, airway inflammation and immune response. Histological analysis of the lungs showed that Qipian® attenuated airway inflammatory cell infiltration and mucus hyperproduction. Qipian® restored Th1/Th2 imbalance by decreasing interleukin (IL)-4, IL-5, and IL-13 while increasing interferon (IFN)-γ and IL-10. Further investigation revealed that Qipian® treatment induced the upregulation of CD4+CD25+Foxp3+ Treg cells and CD103+ DCs and downregulation of tachykinins neurokinin A (NKA) and NKB in the lung. SIGNIFICANCE Our findings suggested that short-term treatment with Qipian® could alleviate inflammation in allergic asthma through restoring the Th1/Th2 balance by recruiting Treg cells to airways and inducing the proliferation of CD103+ DCs, which actually provides a new treatment option for asthma.
Collapse
Affiliation(s)
- Huiying Wang
- Department of Allergy and Clinical Immunology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China.
| | - Fan Tao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang Province 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang Province 315832, China.
| | - Chang-Yun Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang Province 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang Province 315832, China.
| | - Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang Province 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang Province 315832, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang Province 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang Province 315832, China.
| |
Collapse
|
7
|
Lu Q, Hu Y, Nabi F, Li Z, Janyaro H, Zhu W, Liu J. Effect of Penthorum Chinense Pursh Compound on AFB1-Induced Immune Imbalance via JAK/STAT Signaling Pathway in Spleen of Broiler Chicken. Vet Sci 2023; 10:521. [PMID: 37624308 PMCID: PMC10459701 DOI: 10.3390/vetsci10080521] [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: 05/19/2023] [Revised: 07/18/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
Aflatoxin B1(AFB1) is the main secondary metabolite produced by Aspergillus flavus, which is highly toxic, carcinogenic, mutagenic and teratogenic. It can induce immune imbalance in animals or humans. Penthorum chinense Pursh (PCP) is a traditional herbal plant that has been used as a hepatoprotective drug with a long history in China. Based on the theory of traditional Chinese Medicine, we prepared Penthorum chinense Pursh Compound (PCPC) by combining four herbal medicines: 5 g Penthorum chinense Pursh, 5 g Radix bupleuri, 1 g Artemisia capillaris Thunb and 1 g Radix glycyrrhizae. The role of the Penthorum chinense Pursh Compound (PCPC) in preventing AFB1-induced immune imbalance in broiler chickens was studied. A total of 180 broiler chickens were equally distributed in six groups: controls, AFB1, YCHD and high-, medium- and low-dose PCPC treatment groups. After 28 days, broilers were anesthetized, and serum spleen and thymus samples were collected for analysis. Results show that AFB1 significantly increased and decreased the relative organ weight of the spleen and thymus, respectively. Pathological section of hematoxylin/eosin (H&E) stained spleen sections showed that AFB1 resulted in splenic tissue damage. Both the serum levels of Immunoglobulin A (IgA) and Immunoglobulin G (IgG) were suppressed in the AFB1 group. IL-6 was elevated in the AFB1 group. The balance between pro-inflammatory cytokines (IFN-γ and IL-2) and anti-inflammatory cytokine (IL-4) was disturbed by AFB1. The apoptosis-related protein and JAK/STAT pathway-related gene expression indicated that AFB1-induced apoptosis via JAK/STAT pathway. PCPC has proven its immunoprotective effects by preventing AFB1-induced immune imbalance. PCPC can be applied as a novel immune-modulating medicine in broiler chickens. It can be applied as a novel immune modulator in veterinary clinical practice.
Collapse
Affiliation(s)
- Qin Lu
- Immunology Research Center of Medical Research Institute, Southwest University, Chongqing 402460, China;
| | - Yu Hu
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (Y.H.); (F.N.); (Z.L.)
- Wanzhou District Livestock Industry Development Center, Chongqing 404020, China
| | - Fazul Nabi
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (Y.H.); (F.N.); (Z.L.)
| | - Zhenzhen Li
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (Y.H.); (F.N.); (Z.L.)
- College of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing 404155, China
| | - Habibullah Janyaro
- Department of Veterinary Surgery, Shaheed Benazir Bhutto University of Veterinary and Animal Science, Sakrand 67210, Pakistan;
| | - Wenyan Zhu
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Juan Liu
- Immunology Research Center of Medical Research Institute, Southwest University, Chongqing 402460, China;
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (Y.H.); (F.N.); (Z.L.)
| |
Collapse
|
8
|
Losol P, Ji MH, Kim JH, Choi JP, Yun JE, Seo JH, Kim BK, Chang YS, Kim SH. Bronchial epithelial cells release inflammatory markers linked to airway inflammation and remodeling in response to TLR5 ligand flagellin. World Allergy Organ J 2023; 16:100786. [PMID: 37332524 PMCID: PMC10276272 DOI: 10.1016/j.waojou.2023.100786] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/25/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
Background/Aims Flagellin, which is abundant in gram-negative bacteria, including Pseudomonas, is reported to influence on inflammatory responses in various lung diseases. However, its effect on airway epithelial cells in contribution to asthma pathogenesis is not elucidated yet. We aimed to investigate the effect of TLR5 ligand flagellin on the transcriptomic profile of primary human epithelial cells and to determine the markers of airway inflammation. Methods Normal human bronchial epithelial (NHBE) cells were grown and differentiated in air-liquid interface (ALI) culture for 14-16 days. The cells were treated with flagellin in vitro at 10 and 100 ng/ml for 3 and 24 h. The conditioned media and cells were harvested to validate inflammatory markers involved in airway inflammation using ELISA, Western blot, and quantitative PCR methods. RNA-sequencing was performed to investigate the transcriptional response to flagellin in ALI-NHBE cells. Results Altered transcriptional responses to flagellin in differentiated bronchial epithelial cells were determined, including genes encoding chemokines, matrix metalloproteinases, and antimicrobial biomolecules. Pathway analysis of the transcriptionally responsive genes revealed enrichment of signaling pathways. Flagellin induced the mRNA expressions of proinflammatory cytokines and chemokines, and secretion of GM-CSF, CXCL5, CCL5 and CXCL10. Flagellin enhanced the protein expression of MMP-13 in TGF-β1 and TGF-β2 pretreated cell lysates and Wnt/β-catenin signaling. Conclusions These findings suggest that flagellin could be a potent inducer of inflammatory markers that may contribute to airway inflammation and remodeling.
Collapse
Affiliation(s)
- Purevsuren Losol
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Medical Research Center, Seoul National University, Seoul, South Korea
| | - Mi-Hong Ji
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jin Hee Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jun-Pyo Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jeong-Eun Yun
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Jang-Ho Seo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Byung-Keun Kim
- Department of Internal Medicine, Korea University Medical Center Anam Hospital, Seoul, South Korea
| | - Yoon-Seok Chang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Medical Research Center, Seoul National University, Seoul, South Korea
| | - Sae-Hoon Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
9
|
Wang J, Gao Q, Wan S, Hao J, Lian X, Ma J, Zhang X, Zheng Z, Li Q. Antiasthmatic Compounds Targeting β 2-Adrenergic Receptor from Perilla frutescens Improved Lung Inflammation by Inhibiting the NF-κB Signaling Pathway. JOURNAL OF NATURAL PRODUCTS 2022; 85:2656-2666. [PMID: 36322828 DOI: 10.1021/acs.jnatprod.2c00767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Asthma is a highly prevalent and heterogeneous chronic respiratory disease and is often treated with inhaled corticosteroids or in combination with a β2-adrenergic receptor (β2-AR) agonist. However, around 5% of asthma remains uncontrolled, and more effective antiasthmatic drugs with known mechanisms are in high demand. Herein, we immobilized β2-AR on the polystyrene amino microsphere surface in a one-step fashion. The successful immobilization of β2-AR was verified by scanning electron microscopy and chromatographic analysis. We screened rosmarinic acid (RA) as the bioactive compound targeting β2-AR in Perilla frutescens (L.) Britton by mass spectroscopy. The binding constant between RA and β2-AR was determined to be 2.95 × 104 M-1 by adsorption energy distribution and frontal analysis. The antiasthmatic effect and mechanism of RA were examined on a murine model of allergic asthma induced by ovalbumin (OVA) and aluminum hydroxide. The results showed that RA significantly reduced lung inflammatory cell numbers, the production of Th2 cytokines, and the secretion of total IgE, OVA-specific IgE, and eotaxin. The decreased inflammatory cell infiltration and mucus hypersecretion were associated with the inhibition of the NF-κB signaling pathway. Moreover, the mRNA expression levels of AMCase, CCL11, CCR3, Ym2, and E-selectin in the lung tissues were effectively reduced. It is the first time that RA was proven to target β2-AR and be effective in counteracting allergic airway inflammation via the NF-κB signaling pathway. Therefore, the immobilized β2-AR preserves the potential in screening antiasthmatic compounds from herbal medicine, and RA can be developed as an effective agent for the treatment of allergic asthma.
Collapse
Affiliation(s)
- Jing Wang
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Qiuyu Gao
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Shuangru Wan
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Jiaxue Hao
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Xiaojuan Lian
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Jing Ma
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Xinlei Zhang
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Zhe Zheng
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Qian Li
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| |
Collapse
|
10
|
Uwaezuoke SN, Ayuk AC, Eze JN, Odimegwu CL, Ndiokwelu CO, Eze IC. Postnatal probiotic supplementation can prevent and optimize treatment of childhood asthma and atopic disorders: A systematic review of randomized controlled trials. Front Pediatr 2022; 10:956141. [PMID: 36061384 PMCID: PMC9437454 DOI: 10.3389/fped.2022.956141] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Although several randomized controlled trials (RCTs) published over the past 5 years show that prenatal or postnatal probiotics may prevent or optimize the treatment of childhood asthma and atopic disorders, findings from the systematic reviews and meta-analyses of these studies appear inconsistent. More recent RCTs have focused on postnatal probiotics, and linked specific probiotic strains to better disease outcomes. OBJECTIVE This systematic review aimed to determine if postnatal probiotics are as effective as prenatal probiotics in preventing or treating childhood asthma and atopic disorders. METHODS We searched the PubMed, Medline, Google Scholar, and EMBASE databases for RCTs published within the past 5 years (from 2017 to 2022). We included only full-text RCTs on human subjects published in or translated into the English language. We retrieved relevant data items with a preconceived data-extraction form and assessed the methodological quality of the selected RCTs using the Cochrane Collaboration's tool for assessing the risk of bias in randomized trials. We qualitatively synthesized the retrieved data to determine any significant differences in study endpoints of the probiotic and placebo groups. RESULTS A total of 1,320 participants (688 and 632 in the probiotic and placebo groups) from six RCTs were investigated. One RCT showed that early Lactobacillus rhamnosus GG (LGG) led to a reduction in the cumulative incidence rate of asthma. Another study demonstrated that mixed strains of Lactobacillus paracasei and Lactobacillus fermentum could support clinical improvement in children with asthma while one trial reported a significant reduction in the frequency of asthma exacerbations using a mixture of Ligilactobacillus salivarius and Bifidobacterium breve. Three trials showed that a combination of LGG and Bifidobacterium animalis subsp lactis, Lactobacillus rhamnosus alone, and a probiotic mixture of Lactobacillus ŁOCK strains improved clinical outcomes in children with atopic dermatitis and cow-milk protein allergy. CONCLUSIONS Postnatal strain-specific probiotics (in single or mixed forms) are beneficial in preventing and treating atopic dermatitis and other allergies. Similarly, specific strains are more effective in preventing asthma or improving asthma outcomes. We recommend more interventional studies to establish the most useful probiotic strain in these allergic diseases.
Collapse
Affiliation(s)
- Samuel N. Uwaezuoke
- Department of Paediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
- Department of Pediatrics, College of Medicine, University of Nigeria, Ituku-Ozalla Enugu Campus, Enugu, Nigeria
| | - Adaeze C. Ayuk
- Department of Paediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
- Department of Pediatrics, College of Medicine, University of Nigeria, Ituku-Ozalla Enugu Campus, Enugu, Nigeria
| | - Joy N. Eze
- Department of Paediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
- Department of Pediatrics, College of Medicine, University of Nigeria, Ituku-Ozalla Enugu Campus, Enugu, Nigeria
| | - Chioma L. Odimegwu
- Department of Paediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
- Department of Pediatrics, College of Medicine, University of Nigeria, Ituku-Ozalla Enugu Campus, Enugu, Nigeria
| | - Chibuzo O. Ndiokwelu
- Department of Paediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu, Nigeria
| | - Ikenna C. Eze
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
11
|
Kaczynska A, Klosinska M, Janeczek K, Zarobkiewicz M, Emeryk A. Promising Immunomodulatory Effects of Bacterial Lysates in Allergic Diseases. Front Immunol 2022; 13:907149. [PMID: 35812388 PMCID: PMC9257936 DOI: 10.3389/fimmu.2022.907149] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/30/2022] [Indexed: 01/04/2023] Open
Abstract
In light of an escalating prevalence of allergic disorders, it is crucial to fully comprehend their pathophysiology and etiology. Such knowledge would play a pivotal role in the search for new therapeutic approaches concerning not only diseases' symptoms, but also their underlying causes. The hygiene hypothesis indicates a high correlation between limited exposure to pathogens in early childhood and the risk of developing allergic disorders. Bearing in mind the significance of respiratory and digestive systems' mucous membrane's first-line exposure to pathogens as well as its implications on the host's immune response, a therapy targeted at aforesaid membranes could guarantee promising and extensive treatment outcomes. Recent years yielded valuable information about bacterial lysates (BLs) known for having immunomodulatory properties. They consist of antigen mixtures obtained through lysis of bacteria which are the most common etiologic agents of respiratory tract infections. They interact with dendritic cells located in the mucous membranes of the upper respiratory tract and the gastrointestinal tract by toll-like receptors. The dendritic cells present acquired antigens resulting in innate immune response development on the release of chemokines, both stimulating monocytes and NK cells maturation and promoting polymorphonuclear neutrophil migration. Moreover, they influence the adaptive immune system by stimulating an increase of specific antibodies against administered bacterial antigens. The significance of BLs includes not only an anti-inflammatory effect on local infections but also restoration of Th1/Th2 balance, as demonstrated mainly in animal models. They decrease Th2-related cytokine levels (IL-4, IL-13) and increase Th1-related cytokine levels (IFN-γ). The reestablishment of the balance of the immune response leads to lowering atopic reactions incidence which, in addition to reduced risk of inflammation, provides the alleviation and improvement of clinical manifestations of allergic disorders. In this review, we hereby describe mechanisms of BLs action, considering their significant immunomodulatory role in innate immunity. The correlation between local, innate, and adaptive immune responses and their impact on the clinical course of allergic disorders are discussed as well. To conclude our review, we present up-to-date literature regarding the outcomes of BLs implemented in atopic dermatitis, allergic rhinitis, and asthma prevention and treatment, especially in children.
Collapse
Affiliation(s)
- Agnieszka Kaczynska
- Department of Pulmonary Diseases and Children Rheumatology, Medical University of Lublin, Lublin, Poland
| | - Martyna Klosinska
- Department of Pulmonary Diseases and Children Rheumatology, Medical University of Lublin, Lublin, Poland
| | - Kamil Janeczek
- Department of Pulmonary Diseases and Children Rheumatology, Medical University of Lublin, Lublin, Poland
| | - Michał Zarobkiewicz
- Department of Clinical Immunology, Medical University of Lublin, Lublin, Poland
| | - Andrzej Emeryk
- Department of Pulmonary Diseases and Children Rheumatology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
12
|
Flores-Torres AS, Samarasinghe AE. Impact of Therapeutics on Unified Immunity During Allergic Asthma and Respiratory Infections. FRONTIERS IN ALLERGY 2022; 3:852067. [PMID: 35386652 PMCID: PMC8974821 DOI: 10.3389/falgy.2022.852067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/21/2022] [Indexed: 11/04/2022] Open
Abstract
Asthma is a common chronic respiratory disease that affects millions of people worldwide. Patients with allergic asthma, the most prevalent asthma endotype, are widely considered to possess a defective immune response against some respiratory infectious agents, including viruses, bacteria and fungi. Furthermore, respiratory pathogens are associated with asthma development and exacerbations. However, growing data suggest that the immune milieu in allergic asthma may be beneficial during certain respiratory infections. Immunomodulatory asthma treatments, although beneficial, should then be carefully prescribed to avoid misuse and overuse as they can also alter the host microbiome. In this review, we summarize and discuss recent evidence of the correlations between allergic asthma and the most significant respiratory infectious agents that have a role in asthma pathogenesis. We also discuss the implications of current asthma therapeutics beyond symptom prevention.
Collapse
Affiliation(s)
- Armando S. Flores-Torres
- Division of Pulmonology, Allergy-Immunology, and Sleep, Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN, United States
| | - Amali E. Samarasinghe
- Division of Pulmonology, Allergy-Immunology, and Sleep, Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN, United States
| |
Collapse
|
13
|
Mikkola H, Honkila M, Tapiainen T, Jartti T. Susceptibility to rhinovirus-induced early wheezing as a risk factor for subsequent asthma development. CURRENT RESPIRATORY MEDICINE REVIEWS 2022. [DOI: 10.2174/1573398x18666220103113813] [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
Abstract:
Rhinovirus is one of the two most common viral agents that cause bronchiolitis in young children. During the first 12 months, it is second to the respiratory syncytial virus, but after 12 months, it begins dominating the statistics. Wheezing and dry cough are typical clinical symptoms indicative of rhinovirus-induced bronchiolitis, although overlap of symptoms with other virus infections is common. Several studies have shown that atopic predisposition and reduced interferon responses increase susceptibility to rhinovirus-induced wheezing. More recent studies have found that certain genetic variations at strong asthma loci also increase susceptibility. Rhinovirus-induced wheezing in the early years of life is known to increase the risk of subsequent asthma development and may be associated with airway remodeling. This risk is increased by aeroallergen sensitization. Currently, there are no clinically approved preventive treatments for asthma. However, studies show promising results indicating that children with rhinovirus-affected first-time wheezing respond to bronchodilators in terms of less short-term symptoms and that controlling airway inflammatory responses with anti-inflammatory medication may markedly decrease asthma development. Also, enhancing resistance to respiratory viruses has been a topic of discussion. Primary and secondary prevention strategies are being developed with the aim of decreasing the incidence of asthma. Here, we review the current knowledge on rhinovirus-induced early wheezing as a risk factor for subsequent asthma development and related asthma-prevention strategies.
Collapse
Affiliation(s)
- Hannele Mikkola
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu, Oulu, Finland
| | - Minna Honkila
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| | - Terhi Tapiainen
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| | - Tuomas Jartti
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
14
|
Tramper‐Stranders G, Ambrożej D, Arcolaci A, Atanaskovic‐Markovic M, Boccabella C, Bonini M, Karavelia A, Mingomataj E, O' Mahony L, Sokolowska M, Untersmayr E, Feleszko W. Dangerous liaisons: Bacteria, antimicrobial therapies, and allergic diseases. Allergy 2021; 76:3276-3291. [PMID: 34390006 DOI: 10.1111/all.15046] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 07/31/2021] [Indexed: 12/15/2022]
Abstract
Microbiota composition and associated metabolic activities are essential for the education and development of a healthy immune system. Microbial dysbiosis, caused by risk factors such as diet, birth mode, or early infant antimicrobial therapy, is associated with the inception of allergic diseases. In turn, allergic diseases increase the risk for irrational use of antimicrobial therapy. Microbial therapies, such as probiotics, have been studied in the prevention and treatment of allergic diseases, but evidence remains limited due to studies with high heterogeneity, strain-dependent effectiveness, and variable outcome measures. In this review, we sketch the relation of microbiota with allergic diseases, the overuse and rationale for the use of antimicrobial agents in allergic diseases, and current knowledge concerning the use of bacterial products in allergic diseases. We urgently recommend 1) limiting antibiotic therapy in pregnancy and early childhood as a method contributing to the reduction of the allergy epidemic in children and 2) restricting antibiotic therapy in exacerbations and chronic treatment of allergic diseases, mainly concerning asthma and atopic dermatitis. Future research should be aimed at antibiotic stewardship implementation strategies and biomarker-guided therapy, discerning those patients that might benefit from antibiotic therapy.
Collapse
Affiliation(s)
- Gerdien Tramper‐Stranders
- Department of Pediatrics Franciscus Gasthuis & Vlietland Rotterdam the Netherlands
- Department of Neonatology Erasmus Medical CenterSophia Children's Hospital Rotterdam the Netherlands
| | - Dominika Ambrożej
- Department of Pediatric Pneumonology and Allergy Medical University of Warsaw Warsaw Poland
- Doctoral School Medical University of Warsaw Warsaw Poland
| | - Alessandra Arcolaci
- Immunology Unit University of Verona and General Hospital Borgo Roma Hospital Verona Italy
| | | | - Cristina Boccabella
- Department of Cardiovascular and Thoracic Sciences Università Cattolica del Sacro CuoreFondazione Policlinico Universitario A. Gemelli – IRCCS Rome Italy
| | - Matteo Bonini
- Department of Cardiovascular and Thoracic Sciences Università Cattolica del Sacro CuoreFondazione Policlinico Universitario A. Gemelli – IRCCS Rome Italy
- National Heart and Lung Institute (NHLI) Imperial College London London UK
| | - Aspasia Karavelia
- Department of Ear‐Nose‐Throat surgery General Hospital of Kozani Kozani Greece
| | - Ervin Mingomataj
- Department of Allergology & Clinical Immunology ‘Mother Theresa’ School of Medicine Tirana Albania
| | - Liam O' Mahony
- Departments of Medicine and Microbiology APC Microbiome IrelandNational University of Ireland Cork Ireland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Zurich Switzerland
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Wojciech Feleszko
- Department of Pediatric Pneumonology and Allergy Medical University of Warsaw Warsaw Poland
| | | |
Collapse
|
15
|
Teijeiro A, Gómez RM. Wheezing-Related Relevant Factors and the Role of Viral Bronchiolitis. FRONTIERS IN ALLERGY 2021; 2:726972. [PMID: 35387057 PMCID: PMC8974738 DOI: 10.3389/falgy.2021.726972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/01/2021] [Indexed: 12/17/2022] Open
Abstract
Bronchiolitis is a virus-associated infection of the lower respiratory tract exhibiting signs and symptoms of airway obstruction. Respiratory Syncytial Virus (RSV) is responsible in most cases; however, different rhinoviruses have also been implicated. Specific viruses and time until the first infection, severity of the respiratory condition, and atopic status have a determinant role in the recurrence of wheezing and asthma development. Genetics, lung function, atopic condition, the role of microbiota and environment, pollution, and obesity are considered in the present review. Emergency room visits and hospitalizations because of severe wheezing and smoking during pregnancy among others were identified as risk factors for significant morbidity in our population. Approaching determinant conditions like genetics, allergy, antiviral immunity, and environmental exposures such as farm vs. urban and viral virulence provides an opportunity to minimize morbidity of viral illness and asthma in children.
Collapse
Affiliation(s)
- Alvaro Teijeiro
- Respiratory Department, Children's Hospital, Córdoba, Argentina
| | | |
Collapse
|
16
|
Prevention and Treatment of Asthma Exacerbations in Adults. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:2578-2586. [PMID: 34246434 DOI: 10.1016/j.jaip.2021.05.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 02/08/2023]
Abstract
Asthma exacerbations are major contributors to disease morbidity in patients of all ages. To develop strategies that reduce the disease burden from exacerbations, it is helpful to review current concepts about the risk factors for asthma attacks and current approaches for prevention and treatment. Multiple factors contribute as risks and to the development of asthma exacerbations, including allergic and infectious processes. Viral respiratory infections, primarily from rhinoviruses, are the dominant exacerbating cause for most asthma patients. Allergic sensitization and allergen exposure contribute directly and enhance susceptibility for respiratory viral infections. Respiratory viruses infect airway epithelium to promote underlying type 2 inflammation with eosinophils, the predominant cellular component of increased inflammation. Deficiencies of antiviral interferon responses and generation have been identified that increase susceptibility to viral infections in asthma. Exacerbation treatment focuses on reducing airflow obstruction and suppressing inflammation, followed by improving long-term asthma control. Increasing concern exists regarding the side effects associated with frequent systemic corticosteroid use. A major advance has been the selective use of biologics to prevent exacerbations, primarily in patients with existing type 2 inflammation. Future research to prevent exacerbations is being directed toward antiviral activity and a more encompassing regulation of underlying airway inflammation.
Collapse
|
17
|
de Boer GM, Braunstahl G, van der Ploeg EK, van Zelst CM, van Bruggen A, Epping G, van Nimwegen M, Verhoeven G, Birnie E, Boxma‐de Klerk BM, de Bruijn MJW, Stadhouders R, Hendriks RW, Tramper‐Stranders GA. Bacterial lysate add-on therapy to reduce exacerbations in severe asthma: A double-blind placebo-controlled trial. Clin Exp Allergy 2021; 51:1172-1184. [PMID: 34289183 PMCID: PMC9292626 DOI: 10.1111/cea.13990] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 04/28/2021] [Accepted: 07/19/2021] [Indexed: 11/26/2022]
Abstract
Background Asthma exacerbations are frequently induced by respiratory tract infections (RTIs). Bacterial lysates have been described to possess immune‐modulatory effects and reduce RTIs as well as asthma symptoms in children. However, whether bacterial lysates have similar effects in adult asthma patients is unknown. Aims To reduce asthma exacerbations by add‐on bacterial lysate therapy in adults with severe asthma and to characterize the clinical and immune‐modulatory effects of this treatment. Methods Asthma patients (GINA 4) with ≥2 annual exacerbations in the previous year were included. The intervention regimen consisted of OM‐85/placebo for 10 consecutive days per month for 6 months during two winter seasons. Primary end‐point was the number of severe asthma exacerbations within 18 months. The study was approved by the national and local ethical review board and registered in the Dutch Trial Registry (NL5752). All participants provided written informed consent. Results Seventy‐five participants were included (38 OM‐85; 37 placebo). Exacerbation frequencies were not different between the groups after 18 months (incidence rate ratio 1.07, 95%CI [0.68–1.69], p = 0.77). With the use of OM‐85, FEV1% increased by 3.81% (p = 0.04) compared with placebo. Nasopharyngeal swabs taken during RTIs detected a virus less frequently in patients using OM‐85 compared to placebo (30.5% vs. 48.0%, p = 0.02). In subjects with type 2 inflammation adherent to the protocol (22 OM‐85; 20 placebo), a non‐statistically significant decrease in exacerbations in the OM‐85 group was observed (IRR = 0.71, 95%CI [0.39–1.26], p = 0.25). Immune‐modulatory effects included an increase in several plasma cytokines in the OM‐85 group, especially IL‐10 and interferons. Peripheral blood T‐ and B cell subtyping, including regulatory T cells, did not show differences between the groups. Conclusion Although OM‐85 may have immune‐modulatory effects, it did not reduce asthma exacerbations in this heterogeneous severe adult asthma group. Post hoc analysis showed a potential clinical benefit in patients with type 2 inflammation.
Collapse
Affiliation(s)
- Geertje M. de Boer
- Department of Pulmonary MedicineFranciscus Gasthuis & VlietlandRotterdamThe Netherlands
- Department of Pulmonary MedicineErasmus University Medical CenterRotterdamThe Netherlands
| | - Gert‐Jan Braunstahl
- Department of Pulmonary MedicineFranciscus Gasthuis & VlietlandRotterdamThe Netherlands
- Department of Pulmonary MedicineErasmus University Medical CenterRotterdamThe Netherlands
| | - Esmee K. van der Ploeg
- Department of Pulmonary MedicineErasmus University Medical CenterRotterdamThe Netherlands
- Department of Cell BiologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Cathelijne M. van Zelst
- Department of Pulmonary MedicineFranciscus Gasthuis & VlietlandRotterdamThe Netherlands
- Department of Pulmonary MedicineErasmus University Medical CenterRotterdamThe Netherlands
| | - Alie van Bruggen
- Department of Pulmonary MedicineFranciscus Gasthuis & VlietlandRotterdamThe Netherlands
| | - Guido Epping
- Department of Pulmonary MedicineFranciscus Gasthuis & VlietlandRotterdamThe Netherlands
| | - Menno van Nimwegen
- Department of Pulmonary MedicineErasmus University Medical CenterRotterdamThe Netherlands
| | - Gert Verhoeven
- Department of Pulmonary MedicineMaasstad hospitalRotterdamThe Netherlands
| | - Erwin Birnie
- Department of Scientific EducationFranciscus Gasthuis & VlietlandRotterdamThe Netherlands
| | | | | | - Ralph Stadhouders
- Department of Pulmonary MedicineErasmus University Medical CenterRotterdamThe Netherlands
- Department of Cell BiologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Rudi W. Hendriks
- Department of Pulmonary MedicineErasmus University Medical CenterRotterdamThe Netherlands
| | - Gerdien A. Tramper‐Stranders
- Department of Pulmonary MedicineErasmus University Medical CenterRotterdamThe Netherlands
- Department of PediatricsFranciscus Gasthuis & VlietlandRotterdamThe Netherlands
| |
Collapse
|
18
|
Hong H, Tan KS, Yan Y, Chen F, Ong HH, Oo Y, Liu J, Ong YK, Thong M, Sugrue R, Chow VT, Wang DY. Induction of IL-25 Expression in Human Nasal Polyp Epithelium by Influenza Virus Infection is Abated by Interferon-Alpha Pretreatment. J Inflamm Res 2021; 14:2769-2780. [PMID: 34234504 PMCID: PMC8254189 DOI: 10.2147/jir.s304320] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 06/08/2021] [Indexed: 01/23/2023] Open
Abstract
Background Epithelial cytokines including IL-25, IL-33 and thymic stromal lymphopoietin (TLSP) are recently established as drivers of type 2 chronic inflammatory diseases such as chronic rhinosinusitis with nasal polyps (CRSwNP). Here, we further confirmed the increased expression of IL-25 in CRSwNP and investigated potential contributors of IL-25 in CRSwNP epithelium. Methods Sixty CRSwNP, 25 CRSsNP and 15 healthy control tissues were examined for IL-25 expression and for the accompanying type 2 inflammatory cytokines. We then tested different respiratory virus infections on human nasal epithelial cells (hNECs) for their ability to trigger IL-25 expression. In addition, we subjected hNECs generated from CRSwNP tissues to pretreatment with recombinant interferon-alpha (IFN-α) prior to viral infection to evaluate IFN effects on IL-25 induction. Results We confirmed that significantly enhanced levels of IL-25 were observed in CRSwNP tissues, and that IL-25 expression correlated with type 2 inflammatory cytokine expression. In vitro, we observed significantly elevated IL-25 in hNECs infected with influenza A virus as early as 24 hours post-infection (hpi), regardless of tissue origin, and IL-25 correlated positively with viral load. While other respiratory viruses exhibited increasing trends of IL-25, these were not significant at the time-points tested. IFN-α treatment of CRSwNP epithelium was found to exert bimodal effects, ie IFN-α treatment alone induced moderate IL-25 expression, whereas IFN-α pretreatment of hNECs before influenza infection significantly diminished IL-25 induction by active influenza virus infection. Conclusion We have authenticated the observation of elevated IL-25 in CRSwNP, which is correlated with type 2 inflammatory cytokines. Notably, we identified influenza virus infection as a potential contributor of IL-25 in both control and CRSwNP epithelium during active infection. This IL-25 induction can be abated by IFN-α pretreatment which ameliorated active influenza infection. Trial Registration Chictr.org.cn ChiCTR-BON-16010179, Registered 18 December 2016, http://www.chictr.org.cn/showproj.aspx?proj=17331. The authors agree on the sharing of deidentified participant data where it pertains to request directly related to the data in this article when contacted (Haiyu Hong; honghy@mail.sysu.edu.cn).
Collapse
Affiliation(s)
- Haiyu Hong
- Allergy Center, Department of Otolaryngology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People's Republic of China.,Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China.,Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | - Kai Sen Tan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,NUHS Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Biosafety level 3 Core Facility, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore
| | - Yan Yan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,Guangdong Provincial Key Laboratory of Biomedical and Guangdong Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People's Republic of China.,Central Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People's Republic of China
| | - Fenghong Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hsiao Hui Ong
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,NUHS Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yukei Oo
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | - Jing Liu
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,NUHS Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yew Kwang Ong
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,Department of Otolaryngology Head & Neck Surgery, National University Hospital, National University Health System, Singapore
| | - Mark Thong
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,Department of Otolaryngology Head & Neck Surgery, National University Hospital, National University Health System, Singapore
| | - Richard Sugrue
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Vincent T Chow
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,NUHS Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - De Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,NUHS Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
19
|
Shilovskiy IP, Yumashev KV, Nikolsky AA, Vishnyakova LI, Khaitov MR. Molecular and Cellular Mechanisms of Respiratory Syncytial Viral Infection: Using Murine Models to Understand Human Pathology. BIOCHEMISTRY. BIOKHIMIIA 2021; 86:290-306. [PMID: 33838630 PMCID: PMC7957450 DOI: 10.1134/s0006297921030068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/18/2020] [Accepted: 10/18/2020] [Indexed: 12/28/2022]
Abstract
Respiratory syncytial virus (RSV) causes severe pathology of the lower respiratory tract in infants, immunocompromised people, and elderly. Despite decades of research, there is no licensed vaccine against RSV, and many therapeutic drugs are still under development. Detailed understanding of molecular and cellular mechanisms of the RSV infection pathology can accelerate the development of efficacious treatment. Current studies on the RSV pathogenesis are based on the analysis of biopsies from the infected patients; however deeper understanding of molecular and cellular mechanisms of the RSV pathology could be achieved using animal models. Mice are the most often used model for RSV infection because they exhibit manifestations similar to those observed in humans (bronchial obstruction, mucous hypersecretion, and pulmonary inflammation mediated by lymphocytes, macrophages, and neutrophils). Additionally, the use of mice is economically feasible, and many molecular tools are available for studying RSV infection pathogenesis at the molecular and cellular levels. This review summarizes new data on the pathogenesis of RSV infection obtained in mouse models, which demonstrated the role of T cells in both the antiviral defense and the development of lung immunopathology. T cells not only eliminate the infected cells, but also produce significant amounts of the proinflammatory cytokines TNFα and IFNγ. Recently, a new subset of tissue-resident memory T cells (TRM) was identified that provide a strong antiviral defense without induction of lung immunopathology. These cells accumulate in the lungs after local rather than systemic administration of RSV antigens, which suggests new approaches to vaccination. The studies in mouse models have revealed a minor role of interferons in the anti-RSV protection, as RSV possesses mechanisms to escape the antiviral action of type I and III interferons, which may explain the low efficacy of interferon-containing drugs. Using knockout mice, a significant breakthrough has been achieved in understanding the role of many pro-inflammatory cytokines in lung immunopathology. It was found that in addition to TNFα and IFNγ, the cytokines IL-4, IL-5, IL-13, IL-17A, IL-33, and TSLP mediate the major manifestations of the RSV pathogenesis, such as bronchial obstruction, mucus hyperproduction, and lung infiltration by pro-inflammatory cells, while IL-6, IL-10, and IL-27 exhibit the anti-inflammatory effect. Despite significant differences between the mouse and human immune systems, mouse models have made a significant contribution to the understanding of molecular and cellular mechanisms of the pathology of human RSV infection.
Collapse
Affiliation(s)
- Igor P Shilovskiy
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia.
| | - Kirill V Yumashev
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia
| | - Alexandr A Nikolsky
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia
| | - Liudmila I Vishnyakova
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia
| | - Musa R Khaitov
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia
| |
Collapse
|
20
|
Zhang C, Kong Y, Shen K. The Age, Sex, and Geographical Distribution of Self-Reported Asthma Triggers on Children With Asthma in China. Front Pediatr 2021; 9:689024. [PMID: 34540763 PMCID: PMC8448385 DOI: 10.3389/fped.2021.689024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/27/2021] [Indexed: 11/20/2022] Open
Abstract
Background: Asthma can be exacerbated by many triggers, and the heterogeneity of asthma triggers is clear among children with asthma. This study describes asthma triggers using a large-scale electronic dataset from the smartphone-based Chinese Children's Asthma Action Plan (CCAAP) app and aims to examine the difference in asthma triggers among different subgroups of children with asthma. Methods: Data from the smartphone-based CCAAP app between February 22, 2017, and November 23, 2020, were reviewed, and children with asthma who reported their asthma triggers were enrolled. Eight common asthma triggers were listed in the software: upper respiratory infection (URI), allergen sensitization, exercise, emotional disturbances, pungent odors, air pollution/smog, weather change, and tobacco smoke. We compared the incidence of asthma triggers among different subgroups (<6 years vs. 6-17 years; boy vs. girl; eastern region vs. central region vs. western region). Results: We enrolled 6,835 patients with self-reported asthma triggers. When compared by sex, boys had a higher proportion of exercise-triggered asthma than girls (boys vs. girls, 22.5 vs. 19.7%, p < 0.05). The proportion of patients <6 years of age with URI-triggered asthma was higher than that of patients 6-17 years of age (<6 vs. 6-17 years, 80.9 vs. 74.9%, p < 0.001). Patients 6-17 years of age were more likely than patients <6 years of age to report five of the asthma triggers: allergen sensitization (<6 vs. 6-17 years, 26.6 vs. 35.8%, p < 0.001), exercise (<6 vs. 6-17 years, 19.3 vs. 23.7%, p < 0.001), pungent odors (<6 vs. 6-17 years, 8.8 vs. 12.7%, p < 0.001), air pollution/smog (<6 vs. 6-17 years, 9.4 vs. 16.2%, p < 0.001), and tobacco smoke (<6 vs. 6-17 years, 3.5 vs. 5.3%, p < 0.001). In subgroups based on geographical distribution, asthma triggering of allergen sensitization was reported to be the most common in patients from the eastern region (eastern region vs. central region vs. western region, 35.0 vs. 24.6 vs. 28.0%, p < 0.001). Exercise-triggered asthma was found to be the most prevalent among patients from the central region (eastern region vs. central region vs. western region, 21.6 vs. 24.8 vs. 20.4%, p < 0.05). However, the proportion of patients with air pollution/smog as an asthma trigger was the lowest among those from the western region (eastern region vs. central region vs. western region, 14.1 vs. 14.1 vs. 10.8%, p < 0.05). Conclusion: Children with asthma present different types of asthma triggers, both allergenic and nonallergenic. Age, sex, and geographical distribution affect specific asthma triggers. Preventive measures can be implemented based on a patient's specific asthma trigger.
Collapse
Affiliation(s)
- Changhao Zhang
- Department of Respiratory Diseases, Beijing Children's Hospital, Capital Medical University, National Clinical Research Center of Respiratory Diseases, National Center for Children's Health, Beijing, China
| | - Yan Kong
- Department of Respiratory Diseases, Beijing Children's Hospital, Capital Medical University, National Clinical Research Center of Respiratory Diseases, National Center for Children's Health, Beijing, China
| | - Kunling Shen
- Department of Respiratory Diseases, Beijing Children's Hospital, Capital Medical University, National Clinical Research Center of Respiratory Diseases, National Center for Children's Health, Beijing, China
| |
Collapse
|
21
|
de Boer GM, Żółkiewicz J, Strzelec KP, Ruszczyński M, Hendriks RW, Braunstahl GJ, Feleszko W, Tramper-Stranders GA. Bacterial lysate therapy for the prevention of wheezing episodes and asthma exacerbations: a systematic review and meta-analysis. Eur Respir Rev 2020; 29:190175. [PMID: 33246991 PMCID: PMC9488706 DOI: 10.1183/16000617.0175-2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/26/2020] [Indexed: 11/05/2022] Open
Abstract
Wheezing and asthma are a growing cause of morbidity in children and adults. Treatment is aimed at prevention of disease exacerbations and preservation of lung function. Respiratory viruses are involved in ∼40-60% of exacerbations. Bacterial lysates prevent recurrent respiratory tract infections and might reduce exacerbations. Moreover, immunomodulatory effects have been observed in human and animal studies. Here we aimed to assess the effects of bacterial lysate therapy on preschool wheezing episodes and asthma exacerbation frequency. We performed a systematic literature review based on the Preferred Reporting Items for Systematic reviews and Meta-Analysis (PRISMA) statement and a meta-analysis using Cochrane Review Manager. Out of 2016 retrieved articles, 22 studies were included, of which five provided sufficient data for a meta-analysis.The use of bacterial lysates showed a decrease of both wheezing episodes (mean difference -2.35 (-3.03- -1.67), p<0.001) and asthma exacerbations in children (mean difference -0.90 (-1.23- -0.57), p<0.001). Additionally, antibiotic use was reduced, and the duration of wheezing episodes was also decreased. No data for adults with asthma are currently available. The immunomodulatory effect seems to be dependent on increased T-helper (Th)1-cell activation and Th2-cell suppression.These favourable effects of bacterial lysates indicate that they show promise as add-on therapy in preschool wheezing and childhood asthma.
Collapse
Affiliation(s)
- Geertje Maria de Boer
- Dept of Pulmonary Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
- Dept of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Jakub Żółkiewicz
- Dept of Paediatric Allergy and Pulmonology, The Medical University of Warsaw, Warsaw, Poland
- Joint first authors
| | - Konrad Piotr Strzelec
- Dept of Paediatric Allergy and Pulmonology, The Medical University of Warsaw, Warsaw, Poland
| | - Marek Ruszczyński
- Dept of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Rudi W Hendriks
- Dept of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Gert-Jan Braunstahl
- Dept of Pulmonary Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
- Dept of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Wojciech Feleszko
- Dept of Paediatric Allergy and Pulmonology, The Medical University of Warsaw, Warsaw, Poland
| | - Gerdien A Tramper-Stranders
- Dept of Paediatric Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
- Dept of Neonatology, Erasmus MC-Sophia, Rotterdam, The Netherlands
| |
Collapse
|
22
|
Early life microbial exposures and allergy risks: opportunities for prevention. Nat Rev Immunol 2020; 21:177-191. [PMID: 32918062 DOI: 10.1038/s41577-020-00420-y] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2020] [Indexed: 02/07/2023]
Abstract
Allergies, including asthma, food allergy and atopic dermatitis, are increasing in prevalence, particularly in westernized countries. Although a detailed mechanistic explanation for this increase is lacking, recent evidence indicates that, in addition to genetic predisposition, lifestyle changes owing to modernization have an important role. Such changes include increased rates of birth by caesarean delivery, increased early use of antibiotics, a westernized diet and the associated development of obesity, and changes in indoor and outdoor lifestyle and activity patterns. Most of these factors directly and indirectly impact the formation of a diverse microbiota, which includes bacterial, viral and fungal components; the microbiota has a leading role in shaping (early) immune responses. This default programme is markedly disturbed under the influence of environmental and lifestyle risk factors. Here, we review the most important allergy risk factors associated with changes in our exposure to the microbial world and the application of this knowledge to allergy prevention strategies.
Collapse
|
23
|
Janeczek K, Emeryk A, Rachel M, Duma D, Zimmer Ł, Poleszak E. Polyvalent Mechanical Bacterial Lysate Administration Improves the Clinical Course of Grass Pollen-Induced Allergic Rhinitis in Children: A Randomized Controlled Trial. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2020; 9:453-462. [PMID: 32858239 DOI: 10.1016/j.jaip.2020.08.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 07/23/2020] [Accepted: 08/11/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Recent studies highlight the immunoregulatory potential of bacterial lysates, indicating their potential use in the prevention and treatment of allergic diseases. OBJECTIVE To investigate the clinical efficacy of polyvalent mechanical bacterial lysates (PMBLs) in children with grass pollen-induced allergic rhinitis. METHODS Seventy children with seasonal allergic rhinitis were enrolled to this study and were randomly assigned to the PMBL and placebo groups. Severity of seasonal allergic rhinitis symptoms was assessed by the total nasal symptom score, total ocular symptom score, and visual analogue scale. During 3 visits, peak nasal inspiratory flow was measured, and nasal smears for the presence of eosinophils and nasal lavage fluids for the presence of allergen-specific IgE against timothy grass pollen allergens were sampled. RESULTS A statistically significant decrease in total nasal symptom score (P = .001), total ocular symptom score (P = .04), and visual analogue scale score for nasal and eye symptoms (P < .001 and P < .001, respectively) and an increase in peak nasal inspiratory flow (P = .04) were observed in the PMBL group versus the placebo group. During the grass pollen season, an increase and then a decrease in the number of eosinophils in nasal smears was observed in both groups; however, the number of eosinophils was significantly lower in the PMBL group versus the placebo group. No significant changes in allergen-specific IgE concentrations were observed in the PMBL group, whereas in the placebo group a statistically significant increase in allergen-specific IgE concentration was observed. CONCLUSIONS Sublingual administration of PMBLs during the grass pollen season offers significant efficacy in alleviating seasonal allergic rhinitis symptoms in children sensitized to grass pollen allergens. PMBLs probably affect mucosal immunity, weakening the response of TH2 cells.
Collapse
Affiliation(s)
- Kamil Janeczek
- Department of Pulmonary Diseases and Children Rheumatology, Medical University of Lublin, Lublin, Poland.
| | - Andrzej Emeryk
- Department of Pulmonary Diseases and Children Rheumatology, Medical University of Lublin, Lublin, Poland
| | - Marta Rachel
- Chair and Department of Human Physiology and Pathophysiology, University of Rzeszów, Rzeszów, Poland
| | - Dariusz Duma
- Department of Laboratory Diagnostics, Medical University of Lublin, Lublin, Poland
| | - Łukasz Zimmer
- Department of Applied and Social Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Ewa Poleszak
- Department of Applied and Social Pharmacy, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
24
|
Feleszko W, Rossi GA, Krenke R, Canonica GW, Van Gerven L, Kalyuzhin O. Immunoactive preparations and regulatory responses in the respiratory tract: potential for clinical application in chronic inflammatory airway diseases. Expert Rev Respir Med 2020; 14:603-619. [PMID: 32250709 DOI: 10.1080/17476348.2020.1744436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Introduction: The prevalence of chronic inflammatory airway diseases is rising. Their treatment with corticosteroids increases infection risk, while overuse of antimicrobial agents may increase morbidity and antimicrobial resistance. Nonspecific immunomodulatory compounds alter immune responses to both infectious and atopic challenges. These compounds may offer an alternative approach for symptom reduction and prophylaxis against both infections and exacerbations in chronic inflammatory airway disease.Areas covered: We assessed the available data on the efficacy of nonspecific immunomodulators including bacterial lysates, synthetic compounds, and vaccines in chronic rhinosinusitis (CRS); allergic and non-allergic rhinitis; chronic obstructive pulmonary disease (COPD), and asthma. A search of PubMed was carried out using the 'Clinical Trials' filter for each condition and immunomodulatory product detailed below, where available, data from meta-analyses were reported.Expert opinion: Pre-clinical data has revealed a coherent mechanistic path of action for oral immunomodulators on the respiratory immune system, principally via the gut-lung immune axis. In patients with asthma, allergic rhinitis, CRS, and COPD immunomodulatory therapy reduces symptoms, exacerbations, hospitalizations, and drug consumption. However, data are heterogeneous, and study quality remains limited. A lack of high-quality recent trials remains the major unmet research need in the field.
Collapse
Affiliation(s)
- Wojciech Feleszko
- Department of Pediatric Respiratory Diseases and Allergy, The Medical University of Warsaw, Warsaw, Poland
| | - Giovanni A Rossi
- Chief Emeritus, Pediatric Pulmonology and Allergy Units, Cystic Fibrosis Regional Centre, IRCCS G. Gaslini, Genoa, Italy
| | - Rafal Krenke
- Department of Internal Medicine, Pulmonary Diseases & Allergy, Medical University of Warsaw, Warsaw, Poland
| | - G Walter Canonica
- Personalized Medicine Asthma & Allergy, Clinic-Humanitas University & Research Hospital, Milan, Italy
| | - Laura Van Gerven
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Oleg Kalyuzhin
- Professor of Department of Clinical Immunology and Allergy, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| |
Collapse
|
25
|
Hufnagl K, Pali-Schöll I, Roth-Walter F, Jensen-Jarolim E. Dysbiosis of the gut and lung microbiome has a role in asthma. Semin Immunopathol 2020; 42:75-93. [PMID: 32072252 PMCID: PMC7066092 DOI: 10.1007/s00281-019-00775-y] [Citation(s) in RCA: 235] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/15/2019] [Indexed: 02/07/2023]
Abstract
Worldwide 300 million children and adults are affected by asthma. The development of asthma is influenced by environmental and other exogenous factors synergizing with genetic predisposition, and shaping the lung microbiome especially during birth and in very early life. The healthy lung microbial composition is characterized by a prevalence of bacteria belonging to the phyla Bacteroidetes, Actinobacteria, and Firmicutes. However, viral respiratory infections are associated with an abundance of Proteobacteria with genera Haemophilus and Moraxella in young children and adult asthmatics. This dysbiosis supports the activation of inflammatory pathways and contributes to bronchoconstriction and bronchial hyperresponsiveness. Exogenous factors can affect the natural lung microbiota composition positively (farming environment) or negatively (allergens, air pollutants). It is evident that also gut microbiota dysbiosis has a high influence on asthma pathogenesis. Antibiotics, antiulcer medications, and other drugs severely impair gut as well as lung microbiota. Resulting dysbiosis and reduced microbial diversity dysregulate the bidirectional crosstalk across the gut-lung axis, resulting in hypersensitivity and hyperreactivity to respiratory and food allergens. Efforts are undertaken to reconstitute the microbiota and immune balance by probiotics and engineered bacteria, but results from human studies do not yet support their efficacy in asthma prevention or treatment. Overall, dysbiosis of gut and lung seem to be critical causes of the increased emergence of asthma.
Collapse
Affiliation(s)
- Karin Hufnagl
- The Interuniversity Messerli Research Institute, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna, Austria
| | - Isabella Pali-Schöll
- The Interuniversity Messerli Research Institute, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna, Austria
| | - Franziska Roth-Walter
- The Interuniversity Messerli Research Institute, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna, Austria
| | - Erika Jensen-Jarolim
- The Interuniversity Messerli Research Institute, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna, Austria.
- Center for Pathophysiology, Infectiology and Immunology, Institute of Pathophysiology and Allergy Research, Medical University Vienna, Währinger G. 18-20, 1090, Vienna, Austria.
| |
Collapse
|
26
|
Jartti T, Bønnelykke K, Elenius V, Feleszko W. Role of viruses in asthma. Semin Immunopathol 2020; 42:61-74. [PMID: 31989228 PMCID: PMC7066101 DOI: 10.1007/s00281-020-00781-5] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/08/2020] [Indexed: 12/24/2022]
Abstract
Respiratory viral infections are the most important triggers of asthma exacerbations. Rhinovirus (RV), the common cold virus, is clearly the most prevalent pathogen constantly circulating in the community. This virus also stands out from other viral factors due to its large diversity (about 170 genotypes), very effective replication, a tendency to create Th2-biased inflammatory environment and association with specific risk genes in people predisposed to asthma development (CDHR3). Decreased interferon responses, disrupted airway epithelial barrier, environmental exposures (including biased airway microbiome), and nutritional deficiencies (low in vitamin D and fish oil) increase risk to RV and other virus infections. It is intensively debated whether viral illnesses actually cause asthma. Respiratory syncytial virus (RSV) is the leading causative agent of bronchiolitis, whereas RV starts to dominate after 1 year of age. Breathing difficulty induced by either of these viruses is associated with later asthma, but the risk is higher for those who suffer from severe RV-induced wheezing. The asthma development associated with these viruses has unique mechanisms, but in general, RV is a risk factor for later atopic asthma, whereas RSV is more likely associated with later non-atopic asthma. Treatments that inhibit inflammation (corticosteroids, omalizumab) effectively decrease RV-induced wheezing and asthma exacerbations. The anti-RSV monoclonal antibody, palivizumab, decreases the risk of severe RSV illness and subsequent recurrent wheeze. A better understanding of personal and environmental risk factors and inflammatory mechanisms leading to asthma is crucial in developing new strategies for the prevention and treatment of asthma.
Collapse
Affiliation(s)
- Tuomas Jartti
- Department of Paediatrics, Turku University Hospital and University of Turku, Turku, Finland
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Varpu Elenius
- Department of Paediatrics, Turku University Hospital and University of Turku, Turku, Finland
| | - Wojciech Feleszko
- Department of Pediatric Pneumonology and Allergy, The Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
27
|
Role of early life immune regulation in asthma development. Semin Immunopathol 2019; 42:29-42. [PMID: 31873782 PMCID: PMC7079989 DOI: 10.1007/s00281-019-00774-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023]
Abstract
Development of childhood asthma is complex with a strong interaction of genetic, epigenetic, and environmental factors. Ultimately, it is critical how the immune system of a child responds to these influences and whether effective strategies for a balanced and healthy immune maturation can be assured. Pregnancy and early childhood are particularly susceptible for exogenous influences due to the developing nature of a child’s immune system. While endogenous influences such as family history and the genetic background are immutable, epigenetic regulations can be modulated by both heredity and environmental exposures. Prenatal influences such as a mother’s nutrition, smoking, or infections influence the complex interplay of innate and adaptive immune regulation as well as peri- and postnatal influences including mode of delivery. Early in life, induction and continuous training of healthy maturation include balanced innate immunity (e.g., via innate lymphoid cells) and an equilibrium of T-cell subpopulations (e.g., via regulatory T cells) to counter-regulate potential pro-inflammatory or exuberant immune reactions. Later in childhood, rather compensatory immune mechanisms are required to modulate deviant regulation of a child’s already primed immune trajectory. The specific effects of exogenous and endogenous influences on a child’s maturing immune system are summarized in this review, and its importance and potential intervention for early prevention and treatment strategies are delineated.
Collapse
|
28
|
Effect of polyvalent bacterial lysate on the clinical course of pollen allergic rhinitis in children. Postepy Dermatol Alergol 2019; 36:504-505. [PMID: 31616231 PMCID: PMC6791147 DOI: 10.5114/ada.2019.87457] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 08/22/2018] [Indexed: 11/22/2022] Open
|
29
|
Doward L, Svedsater H, Whalley D, Crawford R, Leather D, Lay-Flurrie J, Bosanquet N. A descriptive follow-up interview study assessing patient-centred outcomes: Salford Lung Study in Asthma (SLS Asthma). NPJ Prim Care Respir Med 2019; 29:31. [PMID: 31417102 PMCID: PMC6695403 DOI: 10.1038/s41533-019-0142-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 07/08/2019] [Indexed: 11/24/2022] Open
Abstract
The Salford Lung Study in Asthma (SLS Asthma) was a multicentre, randomised, controlled, open-label trial that assessed initiating once-daily, single-inhaler fluticasone furoate/vilanterol (FF/VI) 100 μg/25 μg or 200 μg/25 μg versus continuing usual care. A subgroup (n = 400) from SLS Asthma was enrolled in this exploratory, interview-based follow-up study. Quantitative and qualitative data were collected via questionnaires. The primary objective was to capture patient-centred outcomes (symptom experience, quality of life [QoL], disease management behaviours) and patient experience. Secondary objectives were to assess the correlation of patient-reported outcomes with pre-defined variables from SLS Asthma (Asthma Control Test [ACT] score). The follow-up sample was representative of the SLS Asthma population; half reported asthma improvement during the study. Breathlessness was the most likely symptom to improve (47.8% of patients reported improvement). Most patients reported ‘no change’ in overall QoL (57.5%) and daily life domains (functioning 66.3%, activities 68.3%, relationships 86.8%, psychological 68.5%). Functioning was reported as the most frequently improved domain (29.8% of patients). Perceived improvement in asthma control (42.5%) and confidence (37.3%) was frequent. ACT responders (defined as patients achieving an ACT score ≥20 and/or an increase of ≥3 in ACT score from baseline at Week 52) were more likely to report asthma improvement (88.7% of patients reporting ‘a lot’ of improvement) than non-responders. Patients’ asthma experiences generally improved during SLS Asthma. Clinical improvements were often associated with perceived improvement by patients, particularly among ACT responders.
Collapse
|
30
|
Hasegawa K, Mansbach JM, Bochkov YA, Gern JE, Piedra PA, Bauer CS, Teach SJ, Wu S, Sullivan AF, Camargo CA. Association of Rhinovirus C Bronchiolitis and Immunoglobulin E Sensitization During Infancy With Development of Recurrent Wheeze. JAMA Pediatr 2019; 173:544-552. [PMID: 30933255 PMCID: PMC6547078 DOI: 10.1001/jamapediatrics.2019.0384] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
IMPORTANCE Rhinovirus infection in early life, particularly with allergic sensitization, is associated with higher risks of developing recurrent wheeze and asthma. While emerging evidence links different rhinovirus species (eg, rhinovirus C) to a higher severity of infection and asthma exacerbation, to our knowledge, little is known about longitudinal associations of rhinovirus C infection during infancy with subsequent morbidities. OBJECTIVE To examine the association of different viruses (respiratory syncytial virus [RSV], rhinovirus species) in bronchiolitis with risks of developing recurrent wheeze. DESIGN, SETTING, AND PARTICIPANTS This multicenter prospective cohort study of infants younger than 1 year who were hospitalized for bronchiolitis was conducted at 17 hospitals across 14 US states during 3 consecutive fall to winter seasons (2011-2014). EXPOSURES Major causative viruses of bronchiolitis, including RSV (reference group) and 3 rhinovirus species (rhinovirus A, B, and C). MAIN OUTCOMES AND MEASURES Development of recurrent wheeze (as defined in national asthma guidelines) by age 3 years. RESULTS This analytic cohort comprised 716 infants who were hospitalized for RSV-only or rhinovirus bronchiolitis. The median age was 2.9 months (interquartile range, 1.6-3.8 months), 541 (76%) had bronchiolitis with RSV only, 85 (12%) had rhinovirus A, 12 (2%) had rhinovirus B, and 78 (11%) had rhinovirus C infection. Overall, 231 (32%) developed recurrent wheeze by age 3 years. In the multivariable Cox model, compared with infants with RSV-only infection, the risk of recurrent wheeze was not significantly different in those with rhinovirus A or B (rhinovirus A: hazard ratio [HR], 1.27; 95% CI, 0.86-1.88; rhinovirus B: HR, 1.39; 95% CI, 0.51-3.77; both P > .10). By contrast, infants with rhinovirus C had a significantly higher risk (HR, 1.58; 95% CI, 1.08-2.32). There was a significant interaction between virus groups and IgE sensitization on the risk of recurrent wheeze (P for interaction < .01). Only infants with both rhinovirus C infection and IgE sensitization (to food or aeroallergens) during infancy had significantly higher risks of recurrent wheeze (HR, 3.03; 95% CI, 1.20-7.61). Furthermore, compared with RSV-only, rhinovirus C infection with IgE sensitization was associated with significantly higher risks of recurrent wheeze with subsequent development of asthma at age 4 years (HR, 4.06; 95% CI, 1.17-14.1). CONCLUSIONS AND RELEVANCE This multicenter cohort study of infants hospitalized for bronchiolitis demonstrated between-virus differences in the risk of developing recurrent wheeze. Infants with rhinovirus C infection, along with IgE sensitization, had the highest risk. This finding was driven by the association with a subtype of recurrent wheeze: children with subsequent development of asthma.
Collapse
Affiliation(s)
- Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jonathan M. Mansbach
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yury A. Bochkov
- Departments of Pediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Madison
| | - James E. Gern
- Departments of Pediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Madison
| | - Pedro A. Piedra
- Department of Molecular Virology and Microbiology and Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Cindy S. Bauer
- Division of Allergy and Immunology, Phoenix Children's Hospital, Phoenix, Arizona
| | - Stephen J. Teach
- Division of Emergency Medicine and Department of Pediatrics, Children’s National Health System, Washington, DC
| | - Susan Wu
- Division of Hospital Medicine, Children's Hospital of Los Angeles, Los Angeles, California
| | - Ashley F. Sullivan
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Carlos A. Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
31
|
Murrison LB, Brandt EB, Myers JB, Hershey GKK. Environmental exposures and mechanisms in allergy and asthma development. J Clin Invest 2019; 129:1504-1515. [PMID: 30741719 DOI: 10.1172/jci124612] [Citation(s) in RCA: 221] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Environmental exposures interplay with human host factors to promote the development and progression of allergic diseases. The worldwide prevalence of allergic disease is rising as a result of complex gene-environment interactions that shape the immune system and host response. Research shows an association between the rise of allergic diseases and increasingly modern Westernized lifestyles, which are characterized by increased urbanization, time spent indoors, and antibiotic usage. These environmental changes result in increased exposure to air and traffic pollution, fungi, infectious agents, tobacco smoke, and other early-life and lifelong risk factors for the development and exacerbation of asthma and allergic diseases. It is increasingly recognized that the timing, load, and route of allergen exposure affect allergic disease phenotypes and development. Still, our ability to prevent allergic diseases is hindered by gaps in understanding of the underlying mechanisms and interaction of environmental, viral, and allergen exposures with immune pathways that impact disease development. This Review highlights epidemiologic and mechanistic evidence linking environmental exposures to the development and exacerbation of allergic airway responses.
Collapse
Affiliation(s)
- Liza Bronner Murrison
- Division of Asthma Research, Cincinnati Children's Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Eric B Brandt
- Division of Asthma Research, Cincinnati Children's Medical Center, Cincinnati, Ohio, USA
| | - Jocelyn Biagini Myers
- Division of Asthma Research, Cincinnati Children's Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Cincinnati Children's Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
32
|
Wang HT, Anvari S, Anagnostou K. The Role of Probiotics in Preventing Allergic Disease. CHILDREN (BASEL, SWITZERLAND) 2019; 6:E24. [PMID: 30764558 PMCID: PMC6406271 DOI: 10.3390/children6020024] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 01/16/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022]
Abstract
The prevalence of allergic disorders has been increasing worldwide and significantly impacts the quality of life of the atopic individual. There has been an increased interest in the role of probiotics for the prevention and treatment of allergic disorders, given the recent evidence that atopy risk may be associated with a dysbiosis of the gut microbiome. Research in this area is ongoing with some studies showing possible benefits of probiotics, with seemingly little to no risk. While these studies suggest that there may be a promise in probiotic use for the prevention or treatment of allergy, further evidence is needed to determine its efficacy, optimal dosing, and strains needed for treatment. In this review, we discuss recently published studies examining the benefits, risks, and role of probiotics in preventing atopic dermatitis, asthma, allergic rhinitis, and food allergy.
Collapse
Affiliation(s)
- Helen T Wang
- Department of Pediatrics, Section of Immunology, Allergy, Rheumatology, and Retrovirology, Texas Children's Hospital, Houston, TX 77030, USA.
- Baylor College of Medicine, Department of Pediatrics, Section of Immunology, Allergy, Rheumatology, and Retrovirology, Houston, TX 77030, USA.
| | - Sara Anvari
- Department of Pediatrics, Section of Immunology, Allergy, Rheumatology, and Retrovirology, Texas Children's Hospital, Houston, TX 77030, USA.
- Baylor College of Medicine, Department of Pediatrics, Section of Immunology, Allergy, Rheumatology, and Retrovirology, Houston, TX 77030, USA.
| | - Katherine Anagnostou
- Department of Pediatrics, Section of Immunology, Allergy, Rheumatology, and Retrovirology, Texas Children's Hospital, Houston, TX 77030, USA.
- Baylor College of Medicine, Department of Pediatrics, Section of Immunology, Allergy, Rheumatology, and Retrovirology, Houston, TX 77030, USA.
| |
Collapse
|
33
|
Jartti T, Smits HH, Bønnelykke K, Bircan O, Elenius V, Konradsen JR, Maggina P, Makrinioti H, Stokholm J, Hedlin G, Papadopoulos N, Ruszczynski M, Ryczaj K, Schaub B, Schwarze J, Skevaki C, Stenberg‐Hammar K, Feleszko W. Bronchiolitis needs a revisit: Distinguishing between virus entities and their treatments. Allergy 2019; 74:40-52. [PMID: 30276826 PMCID: PMC6587559 DOI: 10.1111/all.13624] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 09/07/2018] [Accepted: 09/15/2018] [Indexed: 12/13/2022]
Abstract
Current data indicate that the “bronchiolitis” diagnosis comprises more than one condition. Clinically, pathophysiologically, and even genetically three main clusters of patients can be identified among children suffering from severe bronchiolitis (or first wheezing episode): (a) respiratory syncytial virus (RSV)‐induced bronchiolitis, characterized by young age of the patient, mechanical obstruction of the airways due to mucus and cell debris, and increased risk of recurrent wheezing. For this illness, an effective prophylactic RSV‐specific monoclonal antibody is available; (b) rhinovirus‐induced wheezing, associated with atopic predisposition of the patient and high risk of subsequent asthma development, which may, however, be reversed with systemic corticosteroids in those with severe illness; and (c) wheeze due to other viruses, characteristically likely to be less frequent and severe. Clinically, it is important to distinguish between these partially overlapping patient groups as they are likely to respond to different treatments. It appears that the first episode of severe bronchiolitis in under 2‐year‐old children is a critical event and an important opportunity for designing secondary prevention strategies for asthma. As data have shown bronchiolitis cannot simply be diagnosed using a certain cutoff age, but instead, as we suggest, using the viral etiology as the differentiating factor.
Collapse
Affiliation(s)
- Tuomas Jartti
- Department of Pediatrics Turku University Hospital and University of Turku Turku Finland
| | - Hermelijn H. Smits
- Department of Parasitology Leiden University Medical Center Leiden The Netherlands
| | - Klaus Bønnelykke
- COPSAC Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital University of Copenhagen Copenhagen Denmark
| | - Ozlem Bircan
- Department of Pediatric Allergy Istanbul Medeniyet University Göztepe Training and Research Hospital Istanbul Turkey
| | - Varpu Elenius
- Department of Pediatrics Turku University Hospital and University of Turku Turku Finland
| | - Jon R. Konradsen
- Astrid Lindgren Children's Hospital Karolinska University Hospital Stockholm Sweden
- Department of Women's and Children's Health Karolinska Institutet Stockholm Sweden
- Department of Medicine Solna Immunology and Allergy Unit Karolinska Institutet Karolinska University Hospital Stockholm Sweden
| | - Paraskevi Maggina
- Allergy Department 2nd Pediatric Clinic University of Athens Athens Greece
| | | | - Jakob Stokholm
- COPSAC Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital University of Copenhagen Copenhagen Denmark
| | - Gunilla Hedlin
- Astrid Lindgren Children's Hospital Karolinska University Hospital Stockholm Sweden
- Department of Women's and Children's Health Karolinska Institutet Stockholm Sweden
| | - Nikolaos Papadopoulos
- Allergy Department 2nd Pediatric Clinic University of Athens Athens Greece
- Division of Infection Immunity & Respiratory Medicine University of Manchester Manchester UK
| | | | - Klaudia Ryczaj
- Department of Pediatric Pneumonology and Allergy Medical University of Warsaw Warsaw Poland
| | - Bianca Schaub
- Pediatric Allergology Department of Pediatrics Dr. von Hauner Children′s Hospital University Hospital German Center for Lung Research (DZL) LMU Munich Munich Germany
| | - Jürgen Schwarze
- Centre for Inflammation Research Queen's Medical Research Institute and Child Life and Health University of Edinburgh Edinburgh UK
| | - Chrysanthi Skevaki
- Institute of Laboratory Medicine Philipps Universität Marburg Marburg Germany
- Universities of Giessen and Marburg Lung Center (UGMLC) Philipps Universität, Marburg German Center for Lung Research (DZL) Marburg Germany
| | - Katarina Stenberg‐Hammar
- Astrid Lindgren Children's Hospital Karolinska University Hospital Stockholm Sweden
- Department of Women's and Children's Health Karolinska Institutet Stockholm Sweden
| | - Wojciech Feleszko
- Department of Pediatric Pneumonology and Allergy Medical University of Warsaw Warsaw Poland
| | | |
Collapse
|
34
|
Abstract
Respiratory viral infections including human rhinovirus (RV) infection have been identified as the most important environmental trigger of exacerbations of chronic lung diseases. While well established as the most common viral infections associated with exacerbations of asthma and chronic obstructive pulmonary disease, RVs and other respiratory viruses are also now thought to be important in triggering exacerbations of cystic fibrosis and the interstitial lung diseases. Here, we summarize the epidemiological evidence the supports respiratory viruses including RV as triggers of exacerbations of chronic lung diseases. We propose that certain characteristics of RVs may explain why they are the most common trigger of exacerbations of chronic lung diseases. We further highlight the latest mechanistic evidence supporting how and why common respiratory viral infections may enhance and promote disease triggering exacerbation events, through their interactions with the host immune system, and may be affected by ongoing treatments. We also provide a commentary on how new treatments may better manage the disease burden associated with respiratory viral infections and the exacerbation events that they trigger.
Collapse
|
35
|
Katsoulis K, Ismailos G, Kipourou M, Kostikas K. Microbiota and asthma: Clinical implications. Respir Med 2018; 146:28-35. [PMID: 30665515 DOI: 10.1016/j.rmed.2018.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 10/01/2018] [Accepted: 11/20/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Konstantinos Katsoulis
- Pulmonary Department, 424 Army General Hospital, Periferiaki Odos, 56429, Efkarpia, Thessaloniki, Greece
| | - Georgios Ismailos
- Experimental-Research Center ELPEN, ELPEN Pharmaceuticals, Leoforos Marathonos 95, 19009, Pikermi, Attika, Greece
| | - Maria Kipourou
- Pulmonary Department, 424 Army General Hospital, Periferiaki Odos, 56429, Efkarpia, Thessaloniki, Greece.
| | - Konstantinos Kostikas
- 2nd Respiratory Medicine Department, University of Athens Medical School, Attikon Hospital, Athens, Greece
| |
Collapse
|
36
|
Tan KS, Yan Y, Koh WLH, Li L, Choi H, Tran T, Sugrue R, Wang DY, Chow VT. Comparative Transcriptomic and Metagenomic Analyses of Influenza Virus-Infected Nasal Epithelial Cells From Multiple Individuals Reveal Specific Nasal-Initiated Signatures. Front Microbiol 2018; 9:2685. [PMID: 30487780 PMCID: PMC6246735 DOI: 10.3389/fmicb.2018.02685] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/22/2018] [Indexed: 12/25/2022] Open
Abstract
In vitro and in vivo research based on cell lines and animals are likely to be insufficient in elucidating authentic biological and physiological phenomena mimicking human systems, especially for generating pre-clinical data on targets and biomarkers. There is an obvious need for a model that can further bridge the gap in translating pre-clinical findings into clinical applications. We have previously generated a model of in vitro differentiated human nasal epithelial cells (hNECs) which elucidated the nasal-initiated repertoire of immune responses against respiratory viruses such as influenza A virus and rhinovirus. To assess their clinical utility, we performed a microarray analysis of influenza virus-infected hNECs to elucidate nasal epithelial-initiated responses. This was followed by a metagenomic analysis which revealed transcriptomic changes comparable with clinical influenza datasets. The primary target of influenza infection was observed to be the initiator of innate and adaptive immune genes, leaning toward type-1 inflammatory activation. In addition, the model also elucidated a down-regulation of metabolic processes specific to the nasal epithelium, and not present in other models. Furthermore, the hNEC model detected all 11 gene signatures unique to influenza infection identified from a previous study, thus supporting the utility of nasal-based diagnosis in clinical settings. In conclusion, this study highlights that hNECs can serve as a model for nasal-based clinical translational studies and diagnosis to unravel nasal epithelial responses to influenza in the population, and as a means to identify novel molecular diagnostic markers of severity.
Collapse
Affiliation(s)
- Kai Sen Tan
- Department of Otolaryngology, National University of Singapore, Singapore, Singapore
| | - Yan Yan
- Department of Otolaryngology, National University of Singapore, Singapore, Singapore.,Center for Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Wai Ling Hiromi Koh
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Liang Li
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hyungwon Choi
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore.,Institute of Molecular and Cell Biology, A∗STAR, Singapore, Singapore
| | - Thai Tran
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Richard Sugrue
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - De Yun Wang
- Department of Otolaryngology, National University of Singapore, Singapore, Singapore
| | - Vincent T Chow
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
37
|
Hahn A, Warnken S, Pérez-Losada M, Freishtat RJ, Crandall KA. Microbial diversity within the airway microbiome in chronic pediatric lung diseases. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2018; 63:316-325. [PMID: 29225146 PMCID: PMC5992000 DOI: 10.1016/j.meegid.2017.12.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/22/2017] [Accepted: 12/07/2017] [Indexed: 12/31/2022]
Abstract
The study of the airway microbiome in children is an area of emerging research, especially in relation to the role microbial diversity may play in acute and chronic inflammation. Three such pediatric airway diseases include cystic fibrosis, asthma, and chronic lung disease of prematurity. In cystic fibrosis, the presence of Pseudomonas spp. is associated with decreased microbial diversity. Decreasing microbial diversity is also associated with poor lung function. In asthma, early viral infections appear to drive changes in bacterial diversity which may be associated with asthma risk. Premature infants with Ureaplasma spp. are at higher risk for chronic lung disease due to inflammation. Microbiome changes due to prematurity also appear to affect the inflammatory response to viral infections post-natally. Importantly, microbial diversity can be measured using metataxonomic (e.g., 16S rRNA sequencing) and metagenomic (e.g., shotgun sequencing) approaches. A metagenomics approach may be preferable as it can provide further granularity of the sample composition, identifying the bacterial species or strain, information on additional microbial components, including fungal and viral components, information about functional genomics of the microbiome, and information about antimicrobial resistance mutations. Future studies of pediatric airway diseases incorporating these techniques may provide evidence for new treatment approaches for these vulnerable patient populations.
Collapse
Affiliation(s)
- Andrea Hahn
- Division of Infectious Diseases, Children's National Health System (CNHS), Washington, D.C. 20010, USA; Department of Pediatrics, George Washington University (GWU) School of Medicine and Health Sciences (SMHS), Washington, D.C. 20052, USA.
| | - Stephanie Warnken
- Computational Biology Institute, Milken Institute School of Public Health, GWU, Washington, D.C. 20052, USA
| | - Marcos Pérez-Losada
- Computational Biology Institute, Milken Institute School of Public Health, GWU, Washington, D.C. 20052, USA; CIBIO-InBIO, Universidade do Porto, Campus Agrário de Vairão, Vairão 4485-661, Portugal
| | - Robert J Freishtat
- Department of Pediatrics, George Washington University (GWU) School of Medicine and Health Sciences (SMHS), Washington, D.C. 20052, USA; Division of Emergency Medicine, CNHS, Washington, D.C. 20010, USA
| | - Keith A Crandall
- Computational Biology Institute, Milken Institute School of Public Health, GWU, Washington, D.C. 20052, USA
| |
Collapse
|
38
|
Edwards MR, Walton RP, Jackson DJ, Feleszko W, Skevaki C, Jartti T, Makrinoti H, Nikonova A, Shilovskiy IP, Schwarze J, Johnston SL, Khaitov MR. The potential of anti-infectives and immunomodulators as therapies for asthma and asthma exacerbations. Allergy 2018; 73:50-63. [PMID: 28722755 PMCID: PMC7159495 DOI: 10.1111/all.13257] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2017] [Indexed: 12/30/2022]
Abstract
Asthma is responsible for approximately 25,000 deaths annually in Europe despite available medicines that maintain asthma control and reduce asthma exacerbations. Better treatments are urgently needed for the control of chronic asthma and reduction in asthma exacerbations, the major cause of asthma mortality. Much research spanning >20 years shows a strong association between microorganisms including pathogens in asthma onset, severity and exacerbation, yet with the exception of antibiotics, few treatments are available that specifically target the offending pathogens. Recent insights into the microbiome suggest that modulating commensal organisms within the gut or lung may also be a possible way to treat/prevent asthma. The European Academy of Allergy & Clinical Immunology Task Force on Anti-infectives in Asthma was initiated to investigate the potential of anti-infectives and immunomodulators in asthma. This review provides a concise summary of the current literature and aimed to identify and address key questions that concern the use of anti-infectives and both microbe- and host-based immunomodulators and their feasibility for use in asthma.
Collapse
Affiliation(s)
- M. R. Edwards
- Airway Disease Infection Section National Heart Lung Institute Imperial College London London UK
- MRC and Asthma UK Centre for Allergic Mechanisms of Asthma London UK
| | - R. P. Walton
- Airway Disease Infection Section National Heart Lung Institute Imperial College London London UK
- MRC and Asthma UK Centre for Allergic Mechanisms of Asthma London UK
| | - D. J. Jackson
- Airway Disease Infection Section National Heart Lung Institute Imperial College London London UK
- MRC and Asthma UK Centre for Allergic Mechanisms of Asthma London UK
- Division of Asthma, Allergy & Lung Biology King's College London & Guy's and St Thomas' NHS Trust London UK
| | - W. Feleszko
- Department of Pediatric Respiratory Diseases and Allergy The Medical University of Warsaw Warsaw Poland
| | - C. Skevaki
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics Philipps University Marburg & University Hospital Giessen Marburg Germany
| | - T. Jartti
- The Department of Pediatrics Turku University Hospital Turku Finland
| | - H. Makrinoti
- Airway Disease Infection Section National Heart Lung Institute Imperial College London London UK
- MRC and Asthma UK Centre for Allergic Mechanisms of Asthma London UK
| | - A. Nikonova
- National Research Center Institute of Immunology of Federal Medicobiological Agency Moscow Russia
- Mechnikov Research Institute of Vaccines and Sera Moscow Russia
| | - I. P. Shilovskiy
- National Research Center Institute of Immunology of Federal Medicobiological Agency Moscow Russia
| | - J. Schwarze
- Centre for Inflammation Research University of Edinburgh The Queens Medical Research Institute Edinburgh Edinburgh UK
| | - S. L. Johnston
- Airway Disease Infection Section National Heart Lung Institute Imperial College London London UK
- MRC and Asthma UK Centre for Allergic Mechanisms of Asthma London UK
| | - M. R. Khaitov
- National Research Center Institute of Immunology of Federal Medicobiological Agency Moscow Russia
| | | |
Collapse
|
39
|
Lu J, Xiong L, Zhang X, Liu Z, Wang S, Zhang C, Zheng J, Wang G, Zheng R, Simpson JL, Wang F. The Role of Lower Airway Dysbiosis in Asthma: Dysbiosis and Asthma. Mediators Inflamm 2017; 2017:3890601. [PMID: 29386750 PMCID: PMC5745728 DOI: 10.1155/2017/3890601] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/13/2017] [Accepted: 10/04/2017] [Indexed: 12/12/2022] Open
Abstract
With the development of culture-independent techniques, numerous studies have demonstrated that the lower airway is not sterile in health and harbors diverse microbial communities. Furthermore, new evidence suggests that there is a distinct lower airway microbiome in those with chronic respiratory disease. To understand the role of lower airway dysbiosis in the pathogenesis of asthma, in this article, we review the published reports about the lung microbiome of healthy controls, provide an outlook on the contribution of lower airway dysbiosis to asthma, especially steroid-resistant asthma, and discuss the potential therapies targeted for lower airway dysbiosis.
Collapse
Affiliation(s)
- Junying Lu
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- Department of Intensive Care Unit, First Hospital of Jilin University, Changchun 130021, China
| | - Lingxin Xiong
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Xiaohao Zhang
- Department of Cardiology, Second Hospital of Jilin University, Changchun 130041, China
| | - Zhongmin Liu
- Department of Intensive Care Unit, First Hospital of Jilin University, Changchun 130021, China
| | - Shiji Wang
- Department of Intensive Care Unit, First Hospital of Jilin University, Changchun 130021, China
| | - Chao Zhang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Jingtong Zheng
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Guoqiang Wang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Ruipeng Zheng
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- Department of Interventional Therapy, First Hospital of Jilin University, Changchun 130021, China
| | - Jodie L. Simpson
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- Department of Respiratory and Sleep Medicine, University of Newcastle, New Lambton, NSW, Australia
| | - Fang Wang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
40
|
Jartti T, Gern JE. Role of viral infections in the development and exacerbation of asthma in children. J Allergy Clin Immunol 2017; 140:895-906. [PMID: 28987219 PMCID: PMC7172811 DOI: 10.1016/j.jaci.2017.08.003] [Citation(s) in RCA: 313] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/03/2017] [Accepted: 08/22/2017] [Indexed: 12/31/2022]
Abstract
Viral infections are closely linked to wheezing illnesses in children of all ages. Respiratory syncytial virus (RSV) is the main causative agent of bronchiolitis, whereas rhinovirus (RV) is most commonly detected in wheezing children thereafter. Severe respiratory illness induced by either of these viruses is associated with subsequent development of asthma, and the risk is greatest for young children who wheeze with RV infections. Whether viral illnesses actually cause asthma is the subject of intense debate. RSV-induced wheezing illnesses during infancy influence respiratory health for years. There is definitive evidence that RSV-induced bronchiolitis can damage the airways to promote airway obstruction and recurrent wheezing. RV likely causes less structural damage and yet is a significant contributor to wheezing illnesses in young children and in the context of asthma. For both viruses, interactions between viral virulence factors, personal risk factors (eg, genetics), and environmental exposures (eg, airway microbiome) promote more severe wheezing illnesses and the risk for progression to asthma. In addition, allergy and asthma are major risk factors for more frequent and severe RV-related illnesses. Treatments that inhibit inflammation have efficacy for RV-induced wheezing, whereas the anti-RSV mAb palivizumab decreases the risk of severe RSV-induced illness and subsequent recurrent wheeze. Developing a greater understanding of personal and environmental factors that promote more severe viral illnesses might lead to new strategies for the prevention of viral wheezing illnesses and perhaps reduce the subsequent risk for asthma.
Collapse
Affiliation(s)
- Tuomas Jartti
- Department of Paediatrics, Turku University Hospital and University of Turku, Turku, Finland.
| | - James E Gern
- Departments of Pediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| |
Collapse
|
41
|
Cho YS. Effective Strategies for Managing Asthma Exacerbations for Precision Medicine. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2017; 9:463-465. [PMID: 28913984 PMCID: PMC5603473 DOI: 10.4168/aair.2017.9.6.463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 08/20/2017] [Indexed: 01/27/2023]
Affiliation(s)
- You Sook Cho
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|