1
|
Gu BH, Jung HY, Rim CY, Kim TY, Lee SJ, Choi DY, Park HK, Kim M. Comparative Colonisation Ability of Human Faecal Microbiome Transplantation Strategies in Murine Models. Microb Biotechnol 2025; 18:e70173. [PMID: 40448308 DOI: 10.1111/1751-7915.70173] [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: 08/06/2024] [Revised: 05/15/2025] [Accepted: 05/17/2025] [Indexed: 06/02/2025] Open
Abstract
The gut microbiome plays a crucial role in maintaining intestinal homeostasis and influencing immune-mediated diseases. Human faecal microbiota transplantation (FMT) is often employed in murine models to investigate the role of human microbes in disease regulation, but methods for effective colonisation require refinement. This study aimed to assess the colonisation efficiency of human microbiota in a murine model using FMT with human faeces, focusing particularly on the impact of gut microbiota depletion via polyethylene glycol (PEG) and comparing oral-gastric gavage with enema administration routes. Our findings revealed that PEG-induced depletion enhanced human microbiome colonisation in mice. Oral-gastric gavage prolonged colonisation, while enema administration facilitated quicker resolution of dysbiosis, both inducing selective human microbial colonisation in a time-dependent manner. Notably, genera such as Bacteroides, Blautia, Medicaternibacter and Bifidobacteria were successfully colonised, whereas Roseburia, Anaerostipes, Anaerobutyricum and Faecalibacterium failed to establish in the murine gut post-FMT. These findings highlight the challenges of replicating human gut microbiota in murine models and underscore the importance of selecting appropriate FMT methods based on desired outcomes. This study provides valuable insights into the colonisation dynamics of human microbiota in mice, contributing to the development of more effective FMT strategies for disease treatment.
Collapse
Affiliation(s)
- Bon-Hee Gu
- Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Ho Young Jung
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
- Department of Animal Science, College of Natural Resources & Life Science, Pusan National University, Miryang, Korea
| | - Chae-Yun Rim
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Tae-Yong Kim
- Department of Animal Science, College of Natural Resources & Life Science, Pusan National University, Miryang, Korea
| | - Sang-Jin Lee
- Department of Animal Science, College of Natural Resources & Life Science, Pusan National University, Miryang, Korea
| | - Doo Young Choi
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Han-Ki Park
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Myunghoo Kim
- Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
- Department of Animal Science, College of Natural Resources & Life Science, Pusan National University, Miryang, Korea
| |
Collapse
|
2
|
Gurunthalingam MP, Singh MP, Gaikwad NR. Biannual azithromycin mass drug administration for reduction of childhood mortality: a systematic review and meta-analysis. J Antimicrob Chemother 2025; 80:1197-1206. [PMID: 40129224 DOI: 10.1093/jac/dkaf092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/06/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Biannual mass drug administration of azithromycin (MDA-azithromycin) has been proposed as a strategy to reduce childhood mortality in high-mortality regions, particularly sub-Saharan Africa. However, its effectiveness across different age groups and potential risks, including antibiotic resistance, require further evaluation. METHODS We systematically searched PubMed, Cochrane CENTRAL, Web of Science and ClinicalTrials.gov through September 2024 for randomized controlled trials (RCTs) comparing biannual MDA-azithromycin to placebo in children aged 1-59 months. The primary outcomes were mortality in children <1 year and 12-59 months. Secondary outcomes included adverse events and antibiotic resistance. Data were analysed using a random-effects model in Review Manager 5.4, with heterogeneity assessed via I2. Trial sequential analysis (TSA) evaluated cumulative evidence reliability, and the Cochrane RoB2 tool assessed risk of bias. PROSPERO registration: CRD42024589170. RESULTS Five RCTs (691 235 children) were included. Among children <1 year, azithromycin showed a non-significant mortality reduction (RR: 0.90 [0.78, 1.04]; P = 0.14; I2 = 55%), with TSA indicating inconclusive evidence. Among children 12-59 months, MDA-azithromycin significantly reduced mortality (RR: 0.85 [0.79, 0.91]; P < 0.00001; I2 = 26%), with TSA confirming sufficient evidence. Adverse events were rare, but antibiotic resistance data were limited, warranting further monitoring. Evidence quality ranged from moderate to very low, with one trial at high risk of bias. CONCLUSION Biannual MDA-azithromycin significantly reduces mortality in children 12-59 months, supporting its use in high-mortality settings per WHO recommendations. Its impact on infants remains uncertain. Adverse events were minimal, but continued resistance surveillance is essential.
Collapse
Affiliation(s)
| | - Madhusudan Prasad Singh
- Department of Pharmacology, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| | - Nitin Rewaram Gaikwad
- Department of Pharmacology, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| |
Collapse
|
3
|
Wang C, Yu X, Yu X, Xiao H, Song Y, Wang X, Zheng H, Chen K, An Y, Zhou Z, Guo X, Wang F. Gut flora-derived succinate exacerbates Allergic Airway Inflammation by promoting protein succinylation. Redox Biol 2025; 82:103623. [PMID: 40174477 PMCID: PMC11999320 DOI: 10.1016/j.redox.2025.103623] [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: 02/10/2025] [Revised: 03/17/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025] Open
Abstract
Allergic airway inflammation (AAI) is a prevalent respiratory disorder that affects a vast number of individuals globally. There exists a complex interplay among inflammation, immune responses, and metabolic processes, which is of paramount importance in the pathogenesis of AAI. Metabolic dysregulation and protein translational modification (PTM) are well-recognized hallmarks of diseases, playing pivotal roles in the onset and progression of numerous ailments. However, the role of gut microbiota metabolites in the development of AAI, as well as their influence on PTM modifications within this disease context, have not been thoroughly explored and investigated thus far. In AAI patients, succinate was identified as a key metabolite, positively correlated with certain immune parameters and IgE levels, and having good diagnostic value. In AAI mice, gut bacteria were the main source of high succinate levels. Mendelian randomization showed succinate as a risk factor for asthma. Exogenous succinate worsened AAI in mice, increasing airway resistance and inflammatory factor levels. Protein succinylation in AAI mice lungs differed significantly from normal mice, with up-regulated proteins in metabolic pathways. FMT alleviated AAI symptoms by reducing succinate and protein succinylation levels. In vitro, succinate promoted protein succinylation in BEAS-2B cells, and SOD2 was identified as a key succinylated protein, with the K68 site crucial for its modification and enzyme activity regulation. Gut flora-derived succinate exacerbates AAI in mice by increasing lung protein succinylation, and FMT can reverse this. These findings offer new insights into AAI mechanisms and potential therapeutic targets.
Collapse
Affiliation(s)
- Chao Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China; The Medical Basic Research Innovation Center of Airway Disease in North China, Ministry of Education of China, China
| | - Xin Yu
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China; Department of Laboratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Xiao Yu
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Hui Xiao
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Yuemeng Song
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Xinlei Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China; Jilin Provincial International Cooperation Key Laboratory of Pathogen Biology, China
| | - Haoyu Zheng
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Kai Chen
- Department of Oral, Plastic and Aesthetic Surgery, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Yiming An
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Zhengjie Zhou
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China; Jilin Provincial International Cooperation Key Laboratory of Pathogen Biology, China
| | - Xiaoping Guo
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China; Jilin Provincial International Cooperation Key Laboratory of Pathogen Biology, China
| | - Fang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China; The Medical Basic Research Innovation Center of Airway Disease in North China, Ministry of Education of China, China; JLU-USYD Joint Research Center for Respiratory Diseases, China; Jilin Provincial International Cooperation Key Laboratory of Pathogen Biology, China.
| |
Collapse
|
4
|
Luu QQ, Kim T, Cao TBT, Choi I, Yang SY, An BS, Hwang DY, Choi Y, Park HS. Therapeutic Potential of Arginine-Loaded Red Blood Cell Nanovesicles Targeting Obese Asthma. Mediators Inflamm 2025; 2025:8248722. [PMID: 40134943 PMCID: PMC11936518 DOI: 10.1155/mi/8248722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/12/2025] [Indexed: 03/27/2025] Open
Abstract
Purpose: The role of the gut microbiomes has been emphasized in the pathogenesis of obese asthma (OA). However, the molecular mechanism of airway dysfunction underlying OA has not yet been fully elucidated. The effects of microbiomes on arginine metabolism in relation to lung functions and a novel method for delivering arginine to lung tissue based on arginine-loaded red blood cell (RBC)-derived nanovesicles (NVs) (NVArg) will be investigated. Materials and Methods: Inflammatory status, amino acid profiles, and microbial diversity were evaluated in 20 adult patients with OA compared to 30 adult patients with non-OA (NOA) and 10 healthy control (HC) groups. Changes in gut or lung microbial composition that altered arginine metabolism in relation to airway inflammation were investigated in an OA mouse model in vivo. Additionally, this study evaluated the delivery of arginine to lung tissue utilizing NVArg in vivo and in vitro. Results: Significantly increased Bacteroides abundance but decreased serum arginine concentration with lower forced exhaled volume at 1 s (FEV1) (%) was noted in the OA group compared to the NOA and HC groups. In mouse experiments, when OA mice were given living bacteria from normal control (NC) mice, lung arginine concentration and airway resistance were restored. However, the administration of arginine or its metabolite (citrulline) did not increase the arginine levels in the lung tissues. We therefore created NVArg, which successfully delivered arginine into the cytoplasm of the airway epithelial cell line in vitro. Oral administration of NVArg for OA mice significantly induced the AMP-activated protein kinase (AMPK) and endothelial nitric oxide synthase (eNOS) pathways in airway epithelial cells, which reduced airway resistance and inflammation. Conclusion: These findings suggest that microbiomes contribute to airway dysfunction by regulating arginine metabolism, whereas NVArg treatment may be a potential option for managing OA.
Collapse
Affiliation(s)
- Quoc Quang Luu
- Department of Oral and Maxillofacial Surgery, Loma Linda University School of Dentistry, Loma Linda, California, USA
| | - Taejune Kim
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Thi Bich Tra Cao
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Injung Choi
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Seung Yun Yang
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Beum-Soo An
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Dae Youn Hwang
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Youngwoo Choi
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
5
|
Zheng J, Huang Y, Zhang L, Liu T, Zou Y, He L, Guo S. Role of the Gut-Lung Microbiome Axis in Airway Inflammation in OVA-Challenged Mice and the Effect of Azithromycin. J Inflamm Res 2025; 18:2661-2676. [PMID: 40008084 PMCID: PMC11853874 DOI: 10.2147/jir.s506688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Objective This study aimed to investigate the role of the gut-lung microbiome axis in airway inflammation in asthma and to evaluate the effect of azithromycin on this axis, with a focus on the potential mechanism by which azithromycin reduces allergic airway inflammation. Methods Haematoxylin and eosin (H&E) and periodic acid-Schiff (PAS) staining were used to assess pathological changes in the lung tissues of asthmatic mice. Leukocyte cell types in bronchoalveolar lavage fluid (BALF) samples were quantified following Wright-Giemsa staining. Total IgE, OVA-specific IgE, IL-4, IL-6, and IL-17A levels in BALF and total IgE in serum were measured by ELISA. The respiratory and gut microbiota were analysed using 16S rRNA gene sequencing and subsequent taxonomic analysis. Results OVA-challenged asthmatic mice with gut microbiota dysbiosis exhibited alterations in the respiratory microbiota, resulting in further aggravation of airway inflammation. Following faecal microbiota transplantation (FMT) to restore gut microbiota, respiratory microbiota dysbiosis was partially improved, and airway inflammation was significantly alleviated. Furthermore, azithromycin reduced airway inflammation in asthmatic mice, particularly non-eosinophilic inflammation, for which low-dose azithromycin combined with budesonide proved more effective. Azithromycin significantly enhanced the diversity and microbial composition of the gut microbiota and also affected the respiratory microbiota. At the phylum level, azithromycin decreased the abundance of Proteobacteria in the gut microbiota. At the genus level, azithromycin reduced the abundance of Pseudomonas in the respiratory microbiota. Conclusion The gut-lung microbiome axis plays a crucial role in airway inflammation in asthma. Azithromycin may reduce airway inflammation in asthma through modulation of the gut-lung microbiome axis.
Collapse
Affiliation(s)
- Jun Zheng
- Department of Traditional Chinese Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yuying Huang
- Department of Traditional Chinese Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Liang Zhang
- Department of Endocrine, Genetics and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Tiantian Liu
- Department of Traditional Chinese Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Ya Zou
- Department of Traditional Chinese Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Li He
- Department of Traditional Chinese Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Sheng Guo
- Department of Endocrine, Genetics and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
6
|
Perez-Garcia J, Cardenas A, Lorenzo-Diaz F, Pino-Yanes M. Precision medicine for asthma treatment: Unlocking the potential of the epigenome and microbiome. J Allergy Clin Immunol 2025; 155:298-315. [PMID: 38906272 PMCID: PMC12002393 DOI: 10.1016/j.jaci.2024.06.010] [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: 03/06/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Asthma is a leading worldwide biomedical concern. Patients can experience life-threatening worsening episodes (exacerbations) usually controlled by anti-inflammatory and bronchodilator drugs. However, substantial heterogeneity in treatment response exists, and a subset of patients with unresolved asthma carry the major burden of this disease. The study of the epigenome and microbiome might bridge the gap between human genetics and environmental exposure to partially explain the heterogeneity in drug response. This review aims to provide a critical examination of the existing literature on the microbiome and epigenetic studies examining associations with asthma treatments and drug response, highlight convergent pathways, address current challenges, and offer future perspectives. Current epigenetic and microbiome studies have shown the bilateral relationship between asthma pharmacologic interventions and the human epigenome and microbiome. These studies, focusing on corticosteroids and to a lesser extent on bronchodilators, azithromycin, immunotherapy, and mepolizumab, have improved the understanding of the molecular basis of treatment response and identified promising biomarkers for drug response prediction. Immune and inflammatory pathways (eg, IL-2, TNF-α, NF-κB, and C/EBPs) underlie microbiome-epigenetic associations with asthma treatment, representing potential therapeutic pathways to be targeted. A comprehensive evaluation of these omics biomarkers could significantly contribute to precision medicine and new therapeutic target discovery.
Collapse
Affiliation(s)
- Javier Perez-Garcia
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain.
| | - Andres Cardenas
- Department of Epidemiology and Population Health, Stanford University, Stanford, Calif
| | - Fabian Lorenzo-Diaz
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain; Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain
| | - Maria Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
7
|
Wei J, Liu C, Qin D, Ren F, Duan J, Chen T, Wu A. Targeting inflammation and gut microbiota with antibacterial therapy: Implications for central nervous system health. Ageing Res Rev 2024; 102:102544. [PMID: 39419400 DOI: 10.1016/j.arr.2024.102544] [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: 09/02/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
The complex symbiotic relationship between inflammation, the gut microbiota, and the central nervous system (CNS) has become a pivotal focus of contemporary biomedical research. Inflammation, as a physiological defense mechanism, plays a dual role as both a protective and pathological factor, and is intricately associated with gut microbiota homeostasis, often termed the "second brain." The gutbrain axis (GBA) exemplifies this multifaceted interaction, where gut health exerts significantly regulatory effects on CNS functions. Antibacterial therapies represent both promising and challenging strategies for modulating inflammation and gut microbiota composition to confer CNS benefits. However, while such therapies may exert positive modulatory effects on the gut microbiota, they also carry the potential to disrupt microbial equilibrium, potentially exacerbating neurological dysfunction. Recent advances have provided critical insights into the therapeutic implications of antibacterial interventions; nevertheless, the application of these therapies in the context of CNS health warrants a judicious and evidence-based approach. As research progresses, deeper investigation into the microbial-neural interface is essential to fully realize the potential of therapies targeting inflammation and the gut microbiota for CNS health. Future efforts should focus on refining antibacterial interventions to modulate the gut microbiota while minimizing disruption to microbial balance, thereby reducing risks and enhancing efficacy in CNS-related conditions. In conclusion, despite challenges, a more comprehensive understanding of the GBA, along with precise modulation through targeted antibacterial therapies, offers significant promise for advancing CNS disorder treatment. Continued research in this area will lead to innovative interventions and improved patient outcomes.
Collapse
Affiliation(s)
- Jing Wei
- Eye School of Chengdu University of TCM, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, China; School of Pharmaceutical Sciences, China-Pakistan International Science and Technology Innovation Cooperation Base for Ethnic Medicine Development in Hunan Province, Hunan University of Medicine, Huaihua 418000, China.
| | - Chunmeng Liu
- Eye School of Chengdu University of TCM, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, China.
| | - Dalian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Department of Cardiology, the Affiliated Hospital of Southwest Medical University and Key Laboratory of Medical Electrophysiology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| | - Fang Ren
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, China.
| | - Junguo Duan
- Eye School of Chengdu University of TCM, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, China.
| | - Ting Chen
- School of Pharmaceutical Sciences, China-Pakistan International Science and Technology Innovation Cooperation Base for Ethnic Medicine Development in Hunan Province, Hunan University of Medicine, Huaihua 418000, China.
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Department of Cardiology, the Affiliated Hospital of Southwest Medical University and Key Laboratory of Medical Electrophysiology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
8
|
Kim YC, Sohn KH, Kang HR. Gut microbiota dysbiosis and its impact on asthma and other lung diseases: potential therapeutic approaches. Korean J Intern Med 2024; 39:746-758. [PMID: 39252487 PMCID: PMC11384250 DOI: 10.3904/kjim.2023.451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 03/08/2024] [Accepted: 04/07/2024] [Indexed: 09/11/2024] Open
Abstract
The emerging field of gut-lung axis research has revealed a complex interplay between the gut microbiota and respiratory health, particularly in asthma. This review comprehensively explored the intricate relationship between these two systems, focusing on their influence on immune responses, inflammation, and the pathogenesis of respiratory diseases. Recent studies have demonstrated that gut microbiota dysbiosis can contribute to asthma onset and exacerbation, prompting investigations into therapeutic strategies to correct this imbalance. Probiotics and prebiotics, known for their ability to modulate gut microbial compositions, were discussed as potential interventions to restore immune homeostasis. The impact of antibiotics and metabolites, including short-chain fatty acids produced by the gut microbiota, on immune regulation was examined. Fecal microbiota transplantation has shown promise in various diseases, but its role in respiratory disorders is not established. Innovative approaches, including mucus transplants, inhaled probiotics, and microencapsulation strategies, have been proposed as novel therapeutic avenues. Despite challenges, including the sophisticated adaptability of microbial communities and the need for mechanistic clarity, the potential for microbiota-based interventions is considerable. Collaboration between researchers, clinicians, and other experts is essential to unravel the complexities of the gut-lung axis, paving a way for innovative strategies that could transform the management of respiratory diseases.
Collapse
Affiliation(s)
- Young-Chan Kim
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kyoung-Hee Sohn
- Division of Respiratory, Allergy and Critical Care Medicine, Department of Internal Medicine, Kyung Hee University Hospital, Seoul, Korea
| | - Hye-Ryun Kang
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
9
|
Dong L, Tang Y, Wen S, He Y, Li F, Deng Y, Tao Z. Fecal Microbiota Transplantation Alleviates Allergic Rhinitis via CD4 + T Cell Modulation Through Gut Microbiota Restoration. Inflammation 2024; 47:1278-1297. [PMID: 38294580 DOI: 10.1007/s10753-024-01975-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: 11/26/2023] [Revised: 01/07/2024] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
Allergic rhinitis (AR) is an allergic condition of the upper respiratory tract with a complex pathogenesis, including epithelial barrier disruption, immune regulation, and gut microbiota, which is not yet fully understood. Gut microbiota is closely linked to allergic diseases, including AR. Fecal microbiota transplantation (FMT) has recently been recognized as a potentially effective therapy for allergic diseases. However, the efficacy and mechanism of action of FMT in AR remain unknown. Herein, we aimed to observe the implications of gut microbiota on epithelial barrier function and T cell homeostasis, as well as the effect of FMT in AR, using the ovalbumin (OVA)-induced AR mice. The intestinal microbiota of recipient mice was cleared using an antibiotic cocktail; thereafter, FMT was performed. Subsequently, the nasal symptom scores and histopathological features of colon and nasal mucosa tissues of mice were monitored, and serum OVA-sIgE and cytokines of IL-4, IFNγ, IL-17A, and IL-10 cytokine concentrations were examined. Thereafter, tight junction protein and CD4+ T cell-related transcription factor and cytokine expressions were observed in the colon and nasal mucosa, and changes in the expression of PI3K/AKT/mTOR and NFκB signaling pathway were detected by WB assay in each group. Fecal DNA was extracted from the four mice groups for high-throughput 16S rRNA sequencing. FMT ameliorated nasal symptoms and reduced nasal mucosal inflammation in AR mice. Moreover, according to 16S rRNA sequencing, FMT restored the disordered gut microbiota in AR mice. Following FMT, ZO-1 and claudin-1 and Th1/Th2/Th17-related transcription factor and cytokine expressions were upregulated, whereas Treg cell-related Foxp3 and IL-10 expressions were downregulated. Mechanistic studies have revealed that FMT also inhibited PI3K/AKT/mTOR and NF-κB pathway protein phosphorylation in AR mouse tissues. FMT alleviates allergic inflammation in AR by repairing the epithelial barrier and modulating CD4+ T cell balance and exerts anti-inflammatory effects through the PI3K/AKT/mTOR and NF-κB signaling pathways. Moreover, gut microbiota disorders are involved in AR pathogenesis. Disturbed gut microbiota causes an altered immune-inflammatory state in mice and increases susceptibility to AR. This study suggested the regulatory role of the gut-nose axis in the pathogenesis of AR is an emerging field, which provides novel directions and ideas for the treatment of AR.
Collapse
Affiliation(s)
- Lin Dong
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Yulei Tang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Silu Wen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Yan He
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Fen Li
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Yuqin Deng
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China.
| | - Zezhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China.
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China.
| |
Collapse
|
10
|
Lin S, Zhu N, Zhu Y, Mao H, Zhang S. Exploratory analysis on the association of dietary live microbe and non-dietary prebiotic/probiotic intake with serum cotinine levels in the general adult population. Front Nutr 2024; 11:1405539. [PMID: 38863585 PMCID: PMC11165358 DOI: 10.3389/fnut.2024.1405539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/06/2024] [Indexed: 06/13/2024] Open
Abstract
Background Previous research has indicated the potential involvement of the microbiota in smoking-related processes. The present study seeks to examine the relationship between dietary live microbes, as well as probiotic or prebiotic consumption, and serum cotinine levels. Methods This study used data from the National Health and Nutrition Examination Survey 1999-2018. Dietary intake information and probiotic/prebiotic intake data was collected through self-reported questionnaires. Participants were stratified into low, medium, and high intake groups according to their consumption of foods with varying microbial content. Multiple linear models were applied to explore the relationships of dietary live microbes, probiotic or prebiotic use with the serum cotinine level. Results A total of 42,000 eligible participants were included in the final analysis. The weighted median serum cotinine level was 0.05 (0.01, 10.90) ng/ml. Participants with low, medium, and high dietary microbe intake represented 35.4, 43.6, and 21.0% of the cohort, respectively. Furthermore, participants were stratified into three groups based on their overall consumption of foods with variable microbe contents. The association between dietary live microbe intake and serum cotinine levels remained robust across all models, with medium intake as the reference (Model 2: β = -0.14, 95% CI: -0.20, -0.07; High: β = -0.31, 95% CI: -0.39, -0.22). Moreover, both prebiotic and probiotic use exhibited an inverse relationship with serum cotinine levels (Prebiotic: β = -0.19, 95% CI: -0.37, -0.01; Probiotic: β = -0.47, 95% CI: -0.64, -0.30). Subgroup analyses revealed no discernible interactions between dietary live microbe, prebiotic, probiotic use, and serum cotinine levels. Conclusion Our findings suggest a negative correlation between dietary live microbe intake, as well as non-dietary prebiotic/probiotic consumption, and serum cotinine levels.
Collapse
Affiliation(s)
- Shanhong Lin
- Department of Ultrasound, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Ning Zhu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yujing Zhu
- Department of Stomatology, The Affiliated Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Haiping Mao
- Department of Ultrasound, Ninghai Third Hospital, Ningbo, China
| | - Shengmin Zhang
- Department of Ultrasound, The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
11
|
Sim S, Park HJ, Kim YK, Choi Y, Park HS. Lactobacillus paracasei-derived extracellular vesicles alleviate neutrophilic asthma by inhibiting the JNK pathway in airway epithelium. Allergol Int 2024; 73:302-312. [PMID: 37953104 DOI: 10.1016/j.alit.2023.10.008] [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: 05/03/2023] [Revised: 09/28/2023] [Accepted: 09/30/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Lactobacillus paracasei has been known to reduce airway resistance and inflammation in asthma. However, the therapeutic effect of its extracellular vesicles (EVs) in patients with asthma remains unclear. METHODS To validate the clinical relevance of L. paracasei-derived EVs (LpEV) in asthma, the composition of gut microbial EVs was verified by metagenomics in LPS-induced C57BL/6 mice. The components of proteins and metabolites in LpEV were identified by peptide mass fingerprinting and metabolomic analysis. The serum levels of specific IgG1 or IgG4 antibodies to LpEV were compared by ELISA between patients with eosinophilic asthma (EA, n = 10) and those with neutrophilic asthma (NA, n = 10) as well as with healthy controls (HCs, n = 10). Finally, therapeutic effects of LpEV and their metabolites in asthma were validated in vivo/in vitro. RESULTS Significantly lower proportions of EVs derived from Lactobacillus at the genus level were noted in mice with NA than in control mice. Moreover, the serum levels of LpEV-specific IgG4, but not IgG1, were lower in patients with NA than in those with EA or in HCs and positively correlated with FEV1 (%) values. In addition, oral administration of LpEV reduced airway resistance and inflammation in mice with NA. Finally, LpEV and their 3 metabolites (dodecanoic acid, palmitoleic acid, and D-(-)-tagatose) significantly inhibited JNK phosphorylation/IL-8 production in airway epithelium in vitro. CONCLUSIONS These findings suggest that LpEV may have a therapeutic potential targeting NA by suppressing the JNK pathway and proinflammatory cytokine production in airway epithelium.
Collapse
Affiliation(s)
- Soyoon Sim
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Science, Graduate School of Ajou University, Suwon, South Korea
| | | | | | - Youngwoo Choi
- Department of Biomaterials Science, College of Natural Resources and Life Science, Pusan National University, Miryang, South Korea.
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Science, Graduate School of Ajou University, Suwon, South Korea.
| |
Collapse
|
12
|
Kong J, Yang F, Zong Y, Wang M, Jiang S, Ma Z, Li Z, Li W, Cai Y, Zhang H, Zhao X, Wang J. Early-life antibiotic exposure promotes house dust mite-induced allergic airway inflammation by impacting gut microbiota and lung lipid metabolism. Int Immunopharmacol 2024; 128:111449. [PMID: 38199196 DOI: 10.1016/j.intimp.2023.111449] [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: 09/27/2023] [Revised: 12/04/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024]
Abstract
Asthma is a chronic inflammatory respiratory disease. Early-life antibiotic exposure is a unique risk factor for the incidence and severity of asthma later in life. Perturbations in microbial-metabolite-immune interaction caused by antibiotics are closely associated with the pathogenesis of allergy and asthma. We investigated the effect of early intervention with common oral antibiotics on later asthma exacerbations and found that different antibiotic exposures can amplify different types of immune responses induced by HDM. Cefixime (CFX) promoted a biased type 2 inflammation, azithromycin (AZM) enhanced Th17 immune response, and cefuroxime axetil (CFA) induced eosinophils recruitment. Moreover, early-life antibiotic exposure can have short- and long-term effects on the abundance, composition, and diversity of the gut microbiota. In the model of CFX-promoted type 2 airway inflammation, fecal metabolomics indicated abnormal lipid metabolism and T cell response. Lipidomic also suggested allergic airway inflammation amplified by CFX is closely associated with abnormal lipid metabolism in lung tissues. Moreover, abnormalities in lipid metabolism-related genes (LMRGs) were found to have cellular heterogeneity be associated with asthma severity by bioinformatics analysis.
Collapse
Affiliation(s)
- Jingwei Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fan Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuhan Zong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Manting Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shiyuan Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhaotian Ma
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhuqing Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wenle Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuyang Cai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huixian Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoshan Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China; School of Chinese Medicine, Southern Medical University, Guangzhou, China.
| | - Ji Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
13
|
Wang H, Wang Y. What Makes the Gut-Lung Axis Working? From the Perspective of Microbiota and Traditional Chinese Medicine. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:8640014. [PMID: 38274122 PMCID: PMC10810697 DOI: 10.1155/2024/8640014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Abstract
Background An increasing number of studies have proved that gut microbiota is involved in the occurrence and development of various lung diseases and can interact with the diseased lung. The concept of the gut-lung axis (GLA) provides a new idea for the subsequent clinical treatment of lung diseases through human microbiota. This review aims to summarize the microbiota in the lung and gut and the interaction between them from the perspectives of traditional Chinese medicine and modern medicine. Method We conducted a literature search by using the search terms "GLA," "gut microbiota," "spleen," and "Chinese medicine" in the databases PubMed, Web of Science, and CNKI. We then explored the mechanism of action of the gut-lung axis from traditional Chinese medicine and modern medicine. Results The lung and gut microbiota enable the GLA to function through immune regulation, while metabolites of the gut microbiota also play an important role. The spleen can improve the gut microbiota to achieve the regulation of the GLA. Conclusion Improving the gut microbiota through qi supplementation and spleen fortification provides a new approach to the clinical treatment of lung diseases by regulating the GLA. Currently, our understanding of the GLA is limited, and more research is needed to explain its working principle.
Collapse
Affiliation(s)
- Hui Wang
- Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Ying Wang
- Zhejiang Chinese Medical University, Hangzhou 310000, China
| |
Collapse
|
14
|
Vallet N, Salmona M, Malet-Villemagne J, Bredel M, Bondeelle L, Tournier S, Mercier-Delarue S, Cassonnet S, Ingram B, Peffault de Latour R, Bergeron A, Socié G, Le Goff J, Lepage P, Michonneau D. Circulating T cell profiles associate with enterotype signatures underlying hematological malignancy relapses. Cell Host Microbe 2023; 31:1386-1403.e6. [PMID: 37463582 DOI: 10.1016/j.chom.2023.06.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 05/23/2023] [Accepted: 06/19/2023] [Indexed: 07/20/2023]
Abstract
Early administration of azithromycin after allogeneic hematopoietic stem cell transplantation was shown to increase the relapse of hematological malignancies. To determine the impact of azithromycin on the post-transplant gut ecosystem and its influence on relapse, we characterized overtime gut bacteriome, virome, and metabolome of 55 patients treated with azithromycin or a placebo. We describe four enterotypes and the network of associated bacteriophage species and metabolic pathways. One enterotype associates with sustained remission. One taxon from Bacteroides specifically associates with relapse, while two from Bacteroides and Prevotella correlate with complete remission. These taxa are associated with lipid, pentose, and branched-chain amino acid metabolic pathways and several bacteriophage species. Enterotypes and taxa associate with exhausted T cells and the functional status of circulating immune cells. These results illustrate how an antibiotic influences a complex network of gut bacteria, viruses, and metabolites and may promote cancer relapse through modifications of immune cells.
Collapse
Affiliation(s)
- Nicolas Vallet
- Université de Paris Cité, INSERM U976, 75010 Paris, France
| | - Maud Salmona
- Université de Paris Cité, INSERM U976, 75010 Paris, France; Virology Department, AP-HP, Saint-Louis Hospital, 75010 Paris, France
| | - Jeanne Malet-Villemagne
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Maxime Bredel
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - Louise Bondeelle
- Pneumology Unit, AP-HP, Saint-Louis Hospital, 75010 Paris, France
| | - Simon Tournier
- Core Facilities, Saint-Louis Research Institute, Université de Paris Cité, UAR 2030/US 53, 75010 Paris, France
| | | | - Stéphane Cassonnet
- Service de Biostatistique et Information Médicale, AP-HP, Saint-Louis Hospital, 75010 Paris, France
| | | | - Régis Peffault de Latour
- Hematology Transplantation, AP-HP, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75010 Paris, France; Cryostem Consortium, 13382 Marseille, France
| | - Anne Bergeron
- Pneumology Department, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Gérard Socié
- Université de Paris Cité, INSERM U976, 75010 Paris, France; Hematology Transplantation, AP-HP, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75010 Paris, France
| | - Jérome Le Goff
- Université de Paris Cité, INSERM U976, 75010 Paris, France; Virology Department, AP-HP, Saint-Louis Hospital, 75010 Paris, France
| | - Patricia Lepage
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | - David Michonneau
- Université de Paris Cité, INSERM U976, 75010 Paris, France; Hematology Transplantation, AP-HP, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75010 Paris, France.
| |
Collapse
|
15
|
Ramos-Tapia I, Reynaldos-Grandón KL, Pérez-Losada M, Castro-Nallar E. Characterization of the upper respiratory tract microbiota in Chilean asthmatic children reveals compositional, functional, and structural differences. FRONTIERS IN ALLERGY 2023; 4:1223306. [PMID: 37577334 PMCID: PMC10419220 DOI: 10.3389/falgy.2023.1223306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/20/2023] [Indexed: 08/15/2023] Open
Abstract
Around 155 million people worldwide suffer from asthma. In Chile, the prevalence of this disease in children is around 15% and has a high impact in the health system. Studies suggest that asthma is caused by multiple factors, including host genetics, antibiotic use, and the development of the airway microbiota. Here, we used 16S rRNA high-throughput sequencing to characterize the nasal and oral mucosae of 63 asthmatic and 89 healthy children (152 samples) from Santiago, Chile. We found that the nasal mucosa was dominated by a high abundance of Moraxella, Dolosigranulum, Haemophilus, Corynebacterium, Streptococcus, and Staphylococcus. In turn, the oral mucosa was characterized by a high abundance of Streptococcus, Haemophilus, Gemella, Veillonella, Neisseria, and Porphyromonas. Our results showed significantly (P < 0.001) lower alpha diversity and an over-abundance of Streptococcus (P < 0.01) in nasal samples from asthmatics compared to samples from healthy subjects. Community structure, as revealed by co-occurrence networks, showed different microbial interactions in asthmatic and healthy subjects, particularly in the nasal microbiota. The networks revealed keystone genera in each body site, including Prevotella, Leptotrichia, and Porphyromonas in the nasal microbiota, and Streptococcus, Granulicatella, and Veillonella in the oral microbiota. We also detected 51 functional pathways differentially abundant on the nasal mucosa of asthmatic subjects, although only 13 pathways were overrepresented in the asthmatic subjects (P < 0.05). We did not find any significant differences in microbial taxonomic (composition and structure) and functional diversity between the oral mucosa of asthmatic and healthy subjects. This study explores for the first time the relationships between the upper respiratory airways bacteriome and asthma in Chile. It demonstrates that the nasal cavity of children from Santiago harbors unique bacterial communities and identifies potential taxonomic and functional biomarkers of pediatric asthma.
Collapse
Affiliation(s)
- Ignacio Ramos-Tapia
- Centro de Bioinformática y Biología Integrativa, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | | | - Marcos Pérez-Losada
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, The George Washington University, Washington, DC, United States
| | - Eduardo Castro-Nallar
- Departamento de Microbiología, Facultad de Ciencias de la Salud, Universidad de Talca, Talca, Chile
- Centro de Ecología Integrativa, Universidad de Talca, Talca, Chile
| |
Collapse
|
16
|
He R, Wang S, Yang S, Liu R, Nan N, Lu X, Gong M, Li J. Shaoyao-Gancao-Tang regulates the T-helper-type 1/T-helper-type 2 ratio in the lung and gut and alters gut microbiota in rats with ovalbumin-induced asthma. JOURNAL OF ETHNOPHARMACOLOGY 2023; 309:116300. [PMID: 36868437 DOI: 10.1016/j.jep.2023.116300] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shaoyao-Gancao Tang (SGT) is a traditional Chinese medicine formulation. It has been used to treat kinds of pain and to alleviate asthma in clinic. However, the mechanism of action is not known. AIM OF THE STUDY To investigate the anti-asthma effect of SGT involving modulation of the T-helper type 1 (Th1) Th1/Th2 ratio in the gut-lung axis and alteration of the gut microbiota (GM) in rats with ovalbumin (OVA)-induced asthma. MATERIALS AND METHODS The main constituents of SGT were analyzed by high-performance liquid chromatography (HPLC). A model of asthma was established in rats by OVA-induced allergen challenge. Rats suffering from asthma (RSAs) were treated with SGT (2.5, 5.0 and 10.0 g/kg), dexamethasone (1 mg/kg) or physiologic saline for 4 weeks. The level of immunoglobulin (Ig)E in bronchoalveolar lavage fluid (BALF) and serum was determined by enzyme-linked immunosorbent assay. Histology of lung and colon tissues was investigated using staining (hematoxylin and eosin and periodic acid-Schiff). The Th1/Th2 ratio and levels of cytokines (interferon (IFN)-γ and interleukin (IL)-4) in the lung and colon were detected by immunohistochemistry. The GM in fresh feces was analyzed by 16 S rRNA gene sequencing. RESULTS Twelve main constituents (gallic acid, albiflorin, paeoniflorin, liquiritin apioside, liquiritin, benzoic acid, isoliquiritin apioside, isoliquiritin, liquiritigenin, glycyrrhizic acid, isoliquiritigenin and glycyrrhetinic acid) of SGT were simultaneously determined by HPLC. SGT treatment (5.0 and 10.0 g/kg) was found to reduce the IgE level (a vital marker of hyper-responsiveness) in BALF and serum, improve typical morphological changes (inflammatory-cell infiltration and goblet cell metaplasia) in the lung and colon, alleviate airway remodeling (including bronchiostenosis and basement membrane-thickening) in the lung, significantly decrease the IL-4 level and increase the IFN-γ level in the lung and colon, which led to restoration of the IFN-γ/IL-4 ratio. The dysbiosis and dysfunction of GM in RSAs were modulated by SGT. The abundance of bacteria of the genera Ethanoligenens and Harryflintia was increased in RSAs and was decreased upon SGT treatment. The abundance of Family_XIII_AD3011_group was decreased in RSAs and increased upon SGT treatment. Moreover, SGT therapy increased the abundance of bacteria of the genera Ruminococcaceae_UCG-005 and Candidatus_Sacchrimonas, and decreased that of Ruminococcus_2 and Alistipes. CONCLUSIONS SGT ameliorated rats with OVA-induced asthma via regulation of the Th1/Th2 ratio in the lung and gut, and modulated the GM.
Collapse
Affiliation(s)
- Rui He
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Sihui Wang
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
| | - Shengxi Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Renhui Liu
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Nan Nan
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Xuran Lu
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Muxin Gong
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Jing Li
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
17
|
Choi Y, Park HS, Kim YK. Bacterial Extracellular Vesicles: A Candidate Molecule for the Diagnosis and Treatment of Allergic Diseases. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2023; 15:279-289. [PMID: 37188485 DOI: 10.4168/aair.2023.15.3.279] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023]
Abstract
Extracellular vesicles (EVs) are an end product released from almost all living cells such as eukaryotic cells and bacteria. These membrane vesicles containing proteins, lipids, and nucleic acids are mainly involved in intracellular communications through the transfer of their components from donor to acceptor cells. Moreover, EVs have been implicated in many functions in response to environmental changes, contributing to health and disease; bacterial EVs depending on their specific parental bacterium have diverse effects on immune responses to play a beneficial or pathogenic role in patients with various allergic and immunologic diseases. As bacterial EVs are a completely new area of investigation in this field, we highlight our current understanding of bacterial EVs and discuss their diagnostic and therapeutic potentials (as immunomodulators) for targeting asthma and atopic dermatitis.
Collapse
Affiliation(s)
- Youngwoo Choi
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea.
| | | |
Collapse
|
18
|
Tashiro H, Takahashi K, Uchida M, Kurihara Y, Sadamatsu H, Takamori A, Kimura S, Sueoka-Aragane N. Effect of Azithromycin on Exacerbations in Asthma Patients with Obesity: Protocol for a Multi-Center, Prospective, Single-Arm Intervention Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1861. [PMID: 36767227 PMCID: PMC9915079 DOI: 10.3390/ijerph20031861] [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: 12/06/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 06/18/2023]
Abstract
INTRODUCTION Obesity is associated with severe asthma, but no specific treatment has been established. The gut microbiome is increasingly recognized as a crucial factor, but specific treatments focused on the gut microbiome have not been established. Recently, azithromycin has been found to have the capacity to attenuate exacerbations, a characteristic of severe asthma. The effect of azithromycin on obesity-induced severe asthma is not understood. METHODS The purpose of the present study is to clarify the effect of azithromycin on exacerbations in asthmatic patients with obesity. To explore the mechanism, the gut microbiome, metabolites of microbes such as short-chain fatty acids, and blood inflammatory cytokines will be analyzed to evaluate the correlation with the effect of azithromycin on exacerbations in obesity-induced severe asthma. A multi-center, prospective, single-arm intervention study is planned. DISCUSSION The present study will allow us to evaluate the effect of azithromycin on exacerbations, particularly in asthma patients with obesity, and explore biomarkers, targeting molecules including the gut microbiome, which are correlated with decreased exacerbations. The present results could contribute to identifying new therapeutic prospects and targeted microbes or molecules associated with severe clinical characteristics in asthmatic patients with obesity. TRIAL REGISTRATION This study has been registered as a prospective study with the University Hospital Medical Information Network (UMIN0000484389) and the Japan Registry of Clinical Trials (jRCTs071220023).
Collapse
Affiliation(s)
- Hiroki Tashiro
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 8498501, Japan
- TARGET Investigator Group, Saga 8498501, Japan
| | - Koichiro Takahashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 8498501, Japan
- TARGET Investigator Group, Saga 8498501, Japan
| | - Masaru Uchida
- TARGET Investigator Group, Saga 8498501, Japan
- Division of Internal Medicine, Japan Community Health Care Organization Saga Central Hospital, Saga 8498522, Japan
| | - Yuki Kurihara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 8498501, Japan
- TARGET Investigator Group, Saga 8498501, Japan
| | - Hironori Sadamatsu
- TARGET Investigator Group, Saga 8498501, Japan
- Division of Respiratory Medicine, Saga Prefectural Medical Center Koseikan, Saga 8408571, Japan
| | - Ayako Takamori
- Clinical Research Center, Saga University Hospital, Saga 8498501, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 8498501, Japan
| | - Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 8498501, Japan
| |
Collapse
|
19
|
Franz T, Negele J, Bruno P, Böttcher M, Mitchell-Flack M, Reemts L, Krone A, Mougiakakos D, Müller AJ, Zautner AE, Kahlfuss S. Pleiotropic effects of antibiotics on T cell metabolism and T cell-mediated immunity. Front Microbiol 2022; 13:975436. [DOI: 10.3389/fmicb.2022.975436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
T cells orchestrate adaptive and innate immune responses against pathogens and transformed cells. However, T cells are also the main adaptive effector cells that mediate allergic and autoimmune reactions. Within the last few years, it has become abundantly clear that activation, differentiation, effector function, and environmental adaptation of T cells is closely linked to their energy metabolism. Beyond the provision of energy equivalents, metabolic pathways in T cells generate building blocks required for clonal expansion. Furthermore, metabolic intermediates directly serve as a source for epigenetic gene regulation by histone and DNA modification mechanisms. To date, several antibiotics were demonstrated to modulate the metabolism of T cells especially by altering mitochondrial function. Here, we set out to systematically review current evidence about how beta-lactam antibiotics, macrolides, fluoroquinolones, tetracyclines, oxazolidinones, nitroimidazoles, and amphenicols alter the metabolism and effector functions of CD4+ T helper cell populations and CD8+ T cells in vitro and in vivo. Based on this evidence, we have developed an overview on how the use of these antibiotics may be beneficial or detrimental in T cell-mediated physiological and pathogenic immune responses, such as allergic and autoimmune diseases, by altering the metabolism of different T cell populations.
Collapse
|
20
|
Lee SH, Park HK, Kang CD, Choi DH, Park SC, Park JM, Nam SJ, Chae GB, Lee KY, Cho H, Lee SJ. High Dose Intramuscular Vitamin D3 Supplementation Impacts the Gut Microbiota of Patients With Clostridioides Difficile Infection. Front Cell Infect Microbiol 2022; 12:904987. [PMID: 35774395 PMCID: PMC9239168 DOI: 10.3389/fcimb.2022.904987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Background and Aim Current therapeutic strategies for Clostridioides difficile infections (CDI), including oral vancomycin, metronidazole and fecal microbial transplantation, have limited efficacy and treatment failure may occur in as many as one- third of cases. Recent studies have reported that lower concentrations of 25-hydroxyvitamin D are associated with CDI severity and recurrence. However, there have been no studies on microbiota composition after the administration of vitamin D in patients with CDI. Therefore, our study aimed to compare the microbiota composition between the two groups, including eight CDI-positive patients with vitamin D supplementation and ten CDI-positive patients without vitamin D supplementation by using 16S rRNA microbial profiling. Methods Twenty subjects were enrolled in this prospective randomized controlled study. One subject dropped out due to lack of contact with the guardian after discharge and one subject dropped out due to withdrawal of consent. Thus, 18 patients with CDI and vitamin D insufficiency (vitamin D level < 17 ng/mL) were divided into two groups: CDI with vitamin D supplementation (n = 8) and CDI without vitamin D supplementation (control: n = 10). Subjects with vitamin D insufficiency were randomized to receive 200,000 IU intramuscular cholecalciferol whereas patients in the control group received only oral vancomycin. Stool samples were obtained twice before vancomycin was administered and eight weeks after treatment; the V3-V4 16S rRNA metagenomic sequencing was performed using EzBioCloud. Results The alpha diversity of the gut microbiota in the recovery state was significantly higher than that in the CDI state. Analysis of bacterial relative abundance showed significantly lower Proteobacteria and higher Lachnospiraceae, Ruminococcaceae, Akkermansiaceae, and Bifidobacteriaceae in the recovery state. When comparing the control and vitamin D treatment groups after eight weeks, increase in alpha diversity and, abundance of Lachnospiraceae, and Ruminococcaceae exhibited the same trend in both groups. A significant increase in Bifidobacteriaceae and Christensenellaceae was observed in the vitamin D group; Proteobacteria abundance was significantly lower in the vitamin D treatment group after eight weeks than that in the control group. Conclusion Our study confirmed that the increase in the abundance of beneficial bacteria such as Bifidobacteriaceae, and Christensenellaceae were prominently evident during recovery after administration of a high dose of cholecalciferol. These findings indicate that vitamin D administration may be useful in patients with CDI, and further studies with larger sample sizes are required.
Collapse
Affiliation(s)
- Sang Hoon Lee
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Han-Ki Park
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, School of Medicine, Kyungpook National University Chilgok Hospital, Kyungpook National University, Daegu, South Korea
| | - Chang Don Kang
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Dae Hee Choi
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Sung Chul Park
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Jin Myung Park
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Seung-Joo Nam
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Gi Bong Chae
- Department of Surgery, Kangwon National University Hospital, Kangwon NationalUniversity School of Medicine, Chuncheon, South Korea
| | - Kyoung yul Lee
- Department of Pathology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Hyunseok Cho
- Department of Hospital Medicine, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Sung Joon Lee
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, South Korea
- *Correspondence: Sung Joon Lee, ; orcid.org/0000-0002-6451-0400
| |
Collapse
|
21
|
Lee DH, Park HK, Lee HR, Sohn H, Sim S, Park HJ, Shin YS, Kim YK, Choi Y, Park HS. Immunoregulatory effects of Lactococcus lactis-derived extracellular vesicles in allergic asthma. Clin Transl Allergy 2022; 12:e12138. [PMID: 35344296 PMCID: PMC8967260 DOI: 10.1002/clt2.12138] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/23/2022] [Accepted: 03/11/2022] [Indexed: 11/12/2022] Open
Abstract
Background Probiotics have been shown to prevent various allergic diseases by producing extracellular vesicles (EVs). However, the role of EVs in allergic asthma has not yet been completely determined. Methods Gut microbial composition, mainly genera related to probiotics, was investigated in allergic asthmatic mice. Moreover, EVs were isolated from Lactococcus lactis (L. lactis, a selected bacterium) and EV proteins were identified by peptide mass fingerprinting. EV functions in immune responses were evaluated in vivo or ex vivo. Furthermore, the levels of specific IgG antibodies (an alternative marker for EV quantification) to L. lactis‐EVs were measured by ELISA in the sera of 27 asthmatic patients and 26 healthy controls. Results Allergic asthmatic mice showed a lower proportion of Lactococcus compared to healthy mice. L. lactis was cultured and its EVs abundantly contained pyruvate kinase. When allergic asthmatic mice were intranasally treated with EVs, airway hyperresponsiveness, eosinophil number, cytokine secretion, and mucus production were significantly decreased. Moreover, L. lactis‐EV treatment shifted immune responses from Th2 to Th1 by stimulating dendritic cells to produce IL‐12. In addition, significantly lower levels of serum specific IgG4 (but not IgG1) to L. lactis‐EVs were noted in asthmatic patients than in healthy controls. A positive correlation between the levels of EV‐specific IgG4 and FEV1 (%), but a negative correlation between the levels of EV‐specific IgG4 and IL‐13 were observed. Conclusion These findings suggest that L. lactis‐EVs may have immune‐regulating effects on airway inflammation mediated by dendritic cell activation, providing a potential benefit for allergic asthma.
Collapse
Affiliation(s)
- Dong-Hyun Lee
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Han-Ki Park
- Department of Allergy and Clinical Immunology, School of Medicine, Kyungpook National University, Daegu, Korea
| | | | - Hyeukjun Sohn
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Soyoon Sim
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | | | - Yoo Seob Shin
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | | | - Youngwoo Choi
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
22
|
Kullberg RFJ, Haak BW, Abdel-Aziz MI, Davids M, Hugenholtz F, Nieuwdorp M, Galenkamp H, Prins M, Maitland-van der Zee AH, Wiersinga WJ. Gut microbiota of adults with asthma is broadly similar to non-asthmatics in a large population with varied ethnic origins. Gut Microbes 2022; 13:1995279. [PMID: 34743654 PMCID: PMC8583066 DOI: 10.1080/19490976.2021.1995279] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Bacterial gut communities might predispose children to develop asthma. Yet, little is known about the role of these micro-organisms in adult asthmatics. We aimed to profile the relationship between fecal microbiota and asthma in a large-scale, ethnically diverse, observational cohort of adults. Fecal microbiota composition of 1632 adults (172 asthmatics and 1460 non-asthmatics) was analyzed using 16S ribosomal RNA gene sequencing. Using extremely randomized trees machine learning models, we assessed the discriminatory ability of gut bacterial features to identify asthmatics from non-asthmatics. Asthma contributed 0.019% to interindividual dissimilarities in intestinal microbiota composition, which was not significant (P = .97). Asthmatics could not be distinguished from non-asthmatics based on individual microbiota composition by an extremely randomized trees classifier model (area under the receiver operating characteristic curve = 0.54). In conclusion, there were no prominent differences in fecal microbiota composition in adult asthmatics when compared to non-asthmatics in an urban, large-sized and ethnically diverse cohort.
Collapse
Affiliation(s)
- Robert F. J. Kullberg
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands,Microbiota Center Amsterdam, Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands,CONTACT Robert F. J. Kullberg Amsterdam UMC, University of Amsterdam, Center for Experimental and Molecular Medicine, Meibergdreef 9, Room G2-130, Amsterdam1105 AZ, The Netherlands
| | - Bastiaan W. Haak
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands,Microbiota Center Amsterdam, Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands
| | - Mahmoud I. Abdel-Aziz
- Department of Respiratory Medicine, Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands
| | - Mark Davids
- Microbiota Center Amsterdam, Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands
| | - Floor Hugenholtz
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands,Microbiota Center Amsterdam, Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Microbiota Center Amsterdam, Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands,Amsterdam Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Academic Medical Center, Vu University Medical Center, Amsterdam, The Netherlands,Wallenberg Laboratory, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden,Department of Vascular Medicine, Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands
| | - Henrike Galenkamp
- Department of Public Health, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Maria Prins
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, The Netherlands,Department of Internal Medicine, Division of Infectious Diseases, Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands
| | - Anke H. Maitland-van der Zee
- Department of Respiratory Medicine, Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands
| | - W. Joost Wiersinga
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands,Microbiota Center Amsterdam, Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands,Department of Internal Medicine, Division of Infectious Diseases, Amsterdam University Medical Centers - Location Amc, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Altered diversity and composition of gut microbiota in patients with allergic rhinitis. Microb Pathog 2021; 161:105272. [PMID: 34740809 DOI: 10.1016/j.micpath.2021.105272] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Recently, multiple studies have suggested an association between gut dysbiosis and allergic rhinitis (AR) development. However, the role of gut microbiota in AR development remains obscure. METHODS The goal of this study was to compare the gut microbiota composition and short-chain fatty acid (SCFAs) differences associated with AR (N = 18) and HCs (healthy controls, N = 17). Gut microbiota 16SrRNA gene sequences were analyzed based on next-generation sequencing. SCFAs in stool samples were analyzed by gas chromatography-mass spectrometry (GC-MS). RESULTS Compared with HCs, the gut microbiota composition of AR was significantly different in diversity and richness. At the phylum level, the abundance of Firmicutes in the AR group were significantly lower than those in the HCs group. At the genus level, the abundance of Blautia, Eubacterium_hallii_group, Romboutsia, Collinsella, Dorea, Subdoligranulum and Fusicatenibacter in the AR group were significantly lower than that in the HCs group. The concentrations of SCFAs were significantly lower in the AR group compared with the HCs group. Correlation analysis showed that the Eubacterium-hallii-group and Blautia correlated positively with SCFAs. CONCLUSION Our results demonstrate compositional and functional alterations of the gut microbiome in AR.
Collapse
|
24
|
Li N, Dai Z, Wang Z, Deng Z, Zhang J, Pu J, Cao W, Pan T, Zhou Y, Yang Z, Li J, Li B, Ran P. Gut microbiota dysbiosis contributes to the development of chronic obstructive pulmonary disease. Respir Res 2021; 22:274. [PMID: 34696775 PMCID: PMC8543848 DOI: 10.1186/s12931-021-01872-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 10/18/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Dysbiosis of the gut microbiome is involved in the pathogenesis of various diseases, but the contribution of gut microbes to the progression of chronic obstructive pulmonary disease (COPD) is still poorly understood. METHODS We carried out 16S rRNA gene sequencing and short-chain fatty acid analyses in stool samples from a cohort of 73 healthy controls, 67 patients with COPD of GOLD stages I and II severity, and 32 patients with COPD of GOLD stages III and IV severity. Fecal microbiota from the three groups were then inoculated into recipient mice for a total of 14 times in 28 days to induce pulmonary changes. Furthermore, fecal microbiota from the three groups were inoculated into mice exposed to smoke from biomass fuel to induce COPD-like changes. RESULTS We observed that the gut microbiome of COPD patients varied from that of healthy controls and was characterized by a distinct overall microbial diversity and composition, a Prevotella-dominated gut enterotype and lower levels of short-chain fatty acids. After 28 days of fecal transplantation from COPD patients, recipient mice exhibited elevated lung inflammation. Moreover, when mice were under both fecal transplantation and biomass fuel smoke exposure for a total of 20 weeks, accelerated declines in lung function, severe emphysematous changes, airway remodeling and mucus hypersecretion were observed. CONCLUSION These data demonstrate that altered gut microbiota in COPD patients is associated with disease progression in mice model.
Collapse
Affiliation(s)
- Naijian Li
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, Guangdong, People's Republic of China
| | - Zhouli Dai
- College of Medicine, Lishui University, Lishui, Zhejiang, People's Republic of China
| | - Zhang Wang
- Institute of Ecological Science, School of Life Science, South China Normal University, Guangzhou, Guangdong, People's Republic of China
| | - Zhishan Deng
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, Guangdong, People's Republic of China
| | - Jiahuan Zhang
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, Guangdong, People's Republic of China
| | - Jinding Pu
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, Guangdong, People's Republic of China
| | - Weitao Cao
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, Guangdong, People's Republic of China
| | - Tianhui Pan
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, Guangdong, People's Republic of China
| | - Yumin Zhou
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, Guangdong, People's Republic of China
| | - Zhaowei Yang
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, Guangdong, People's Republic of China
| | - Jing Li
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, Guangdong, People's Republic of China
| | - Bing Li
- The GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Pixin Ran
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
25
|
Wang G, McDonald VM. Contemporary Concise Review 2020: Asthma. Respirology 2021; 26:804-811. [PMID: 34164877 DOI: 10.1111/resp.14099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/19/2021] [Accepted: 05/25/2021] [Indexed: 02/05/2023]
Abstract
Bushfires and coronavirus 2019 (COVID-19) were dominate features of 2020. Patients with asthma were significantly affected by the 2019/2020 bushfire season with an increased burden compared to the general population. Patients with controlled asthma do not appear to be at higher risk of severe COVID-19 infection or death than the general population. Personalized medicine is proposed as the next era for asthma management, with treatable traits as a strategy to implement personalized medicine into practice. Patient engagement in personalized medicine strategies is important and needs to be further explored. Oral corticosteroid (OCS) use in asthma is common and contributes a major burden. OCS stewardship is recommended. Biologic therapies reduce exacerbations of severe asthma and biomarkers can be used to predict treatment responders. Epithelia at mucosal and cutaneous surfaces are components in asthma pathogenesis, through airway immunity and inflammation. Dysregulation of resident microbial communities in the lung, gut and skin microbiome is relevant to asthma pathogenesis, but there are still many unknowns in this field.
Collapse
Affiliation(s)
- Gang Wang
- Department of Respiratory and Critical Care Medicine, Clinical Research Centre for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Pulmonary Immunology and Inflammation, Frontiers Science Centre for Disease-Related Molecular Network, Sichuan University, Chengdu, China
| | - Vanessa M McDonald
- Priority Research Centre of Healthy Lungs, School of Nursing and Midwifery, The University of Newcastle, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
26
|
Sadeghdoust M, Mirsadraee M, Aligolighasemabadi F, Khakzad MR, Hashemi Attar A, Naghibi S. Effect of azithromycin on bronchial wall thickness in severe persistent asthma: A double-blind placebo-controlled randomized clinical trial. Respir Med 2021; 185:106494. [PMID: 34116330 DOI: 10.1016/j.rmed.2021.106494] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/17/2021] [Accepted: 05/30/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Azithromycin reduced airway remodeling in animal models of asthma. However, its effect on human subjects has not been studied yet. This study aimed to investigate the effect of long-term treatment with azithromycin on airways wall thickness in patients with severe persistent asthma. METHODS In this randomized, double-blind, placebo-controlled clinical trial, patients with severe persistent asthma received azithromycin (250 mg, BID, three days a week), prednisolone (5 mg, BID), or placebo for eight months in three separate groups in addition to the standard therapy. The improvement in right upper lobe apical segmental bronchus (RB1) wall thickness obtained by high resolution computed tomography was set as the primary outcome. Secondary outcomes included: cough severity, dyspnea severity, asthma control test (ACT) score, asthma exacerbation rate, pulmonary function tests, and fractional exhaled nitric oxide (FENO). RESULTS Seventy-eight out of ninety randomized subjects completed eight months of treatment with azithromycin (n = 25), prednisolone (n = 27), or placebo (n = 26). Bronchial wall thickness percentage did not change significantly in any of the groups. However, the inner radius and lumen area of azithromycin and prednisolone-treated subjects increased significantly (p < 0.05 for both). Azithromycin also significantly improved the dyspnea severity, ACT score, FENO, and FEV1, FEF25-75, and FEV1/FVC (p < 0.05 for all). Cough severity or asthma exacerbation rate did not change significantly after eight months of treatment with azithromycin. CONCLUSION Long-term treatment with azithromycin increased lumen radius and lumen area in patients with severe persistent asthma. However, there was no significant change in wall thickness in any of the treatment groups. TRIAL REGISTRATION IRCT.com (IRCT20091111002695N8).
Collapse
Affiliation(s)
- Mohammadamin Sadeghdoust
- Department of Internal Medicine, Mashhad Medical Sciences Branch, Islamic Azad University, Mashhad, Iran; Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Majid Mirsadraee
- Internist and Pulmonologist, Department of Internal Medicine, Medical School, Islamic Azad University- Mashhad Branch, Mashhad, Iran.
| | - Farnaz Aligolighasemabadi
- Department of Internal Medicine, Mashhad Medical Sciences Branch, Islamic Azad University, Mashhad, Iran; Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad Reza Khakzad
- Innovative Medical Research Center and Department of Immunology, Mashhad Branch Islamic Azad University, Mashhad, Iran.
| | | | - Saeed Naghibi
- Department of Radiology, Mashhad Medical Sciences Branch, Islamic Azad University, Mashhad, Iran.
| |
Collapse
|
27
|
Abstract
The rapid spread of severe acute respiratory coronavirus syndrome 2 (SARS-CoV-2) in the population and throughout the cells within our body has been developing. Another major cycle of coronavirus disease 2019 (COVID-19), which is expected in the coming fall, could be even more severe than the current one. Therefore, effective countermeasures should be developed based on the already obtained clinical and research information about SARS-CoV-2. The aim of this review was to summarize the data on the empirical treatment of COVID-19 acquired during this SARS-CoV-2 infection cycle; this would aid the establishment of an appropriate healthcare policy to meet the challenges in the future. The infectious disease caused by SARS-CoV-2 is characterized by common cold along with hypersensitivity reaction. Thus, in addition to treating common cold, it is essential to minimize the exposure of cells to the virus and to mitigate the uncontrolled immune response. A proper combination of antiviral agents, immune modulators such as prednisolone, and anticoagulants such as heparin and anti-C5a antagonists could be employed to minimize lung damage and prevent systemic involvements. Finally, strategies to achieve population immunity against SARS-CoV-2 should be developed through understanding of the interaction between the immune system and the virus.
Collapse
Affiliation(s)
- Hyoung Shik Shin
- Infectious Diseases Specialist, Korean Society of Zoonoses, Seoul, Korea.
| |
Collapse
|