1
|
Xu C, Sun P, Jiang Q, Meng Y, Dong L, Wang X, Hu X, Li C, Li G, Zheng R, You X, Yang X. Tissue-resident Klebsiella quasipneumoniae contributes to progression of idiopathic pulmonary fibrosis by triggering macrophages mitophagy in mice. Cell Death Discov 2025; 11:168. [PMID: 40221415 PMCID: PMC11993561 DOI: 10.1038/s41420-025-02444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and chronic interstitial lung disease with unclear underlying pathogenic mechanisms. Dysbiosis of the lung microbiota is believed to be associated with the development of fibrosis; however, the roles of the microbiome in the respiratory functions of hosts with IPF remain poorly understood. To investigate the relationship between the lung microbiome and the pathological processes of idiopathic pulmonary fibrosis under laboratory conditions, C57BL/6 J mice were exposed to bleomycin and observed at 7, 14, 21, and 28 days post-exposure. 16S rDNA analysis revealed that the lung microbial community exhibited dysbiosis in the bleomycin-induced pulmonary fibrosis model, characterized by an abnormally high proportion of Klebsiella quasipneumoniae (K. quasipneumoniae), as confirmed by RNA fluorescence in situ hybridization. Throughout the progression of experimental pulmonary fibrosis, Tax4Fun analysis indicated that the abundance of K. quasipneumoniae differed significantly between model mice and control mice, correlating with the sustained activation of reactive oxygen species (ROS) pathways. Importantly, the dysbiosis of K. quasipneumoniae may serve as a critical factor triggering increased ROS levels, accompanied by macrophage mitophagy, ultimately leading to the overexpression of TGF-β1, a key player in the pathogenesis of pulmonary fibrosis. These findings suggest that lung microbiota dysbiosis exacerbates the progression of bleomycin-induced pulmonary fibrosis related to macrophage mitophagy.
Collapse
Affiliation(s)
- Chunjie Xu
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development/ Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
- Division for Medicinal Microorganism-Related Strains, CAMS Collection Center of Pathogenic Microorganisms, 100050, Beijing, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
| | - Peiyi Sun
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development/ Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
- Division for Medicinal Microorganism-Related Strains, CAMS Collection Center of Pathogenic Microorganisms, 100050, Beijing, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
| | - Qiyue Jiang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, 100069, Beijing, China
| | - Yao Meng
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development/ Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
- Division for Medicinal Microorganism-Related Strains, CAMS Collection Center of Pathogenic Microorganisms, 100050, Beijing, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
| | - Luyao Dong
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development/ Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
- Division for Medicinal Microorganism-Related Strains, CAMS Collection Center of Pathogenic Microorganisms, 100050, Beijing, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
| | - Xiukun Wang
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development/ Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
- Division for Medicinal Microorganism-Related Strains, CAMS Collection Center of Pathogenic Microorganisms, 100050, Beijing, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
| | - Xinxin Hu
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development/ Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
- Division for Medicinal Microorganism-Related Strains, CAMS Collection Center of Pathogenic Microorganisms, 100050, Beijing, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
| | - Congran Li
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development/ Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
- Division for Medicinal Microorganism-Related Strains, CAMS Collection Center of Pathogenic Microorganisms, 100050, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China
| | - Guoqing Li
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development/ Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China.
- Division for Medicinal Microorganism-Related Strains, CAMS Collection Center of Pathogenic Microorganisms, 100050, Beijing, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China.
| | - Ruifang Zheng
- Xinjiang Key Laboratory of Uygur Medical Research, Xinjiang Institute of Materia Medica, Urumqi, 841100, China.
| | - Xuefu You
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development/ Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China.
- Division for Medicinal Microorganism-Related Strains, CAMS Collection Center of Pathogenic Microorganisms, 100050, Beijing, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China.
| | - Xinyi Yang
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development/ Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China.
- Division for Medicinal Microorganism-Related Strains, CAMS Collection Center of Pathogenic Microorganisms, 100050, Beijing, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, China.
| |
Collapse
|
2
|
Ge Y, Tang G, Fu Y, Deng P, Yao R. The impact of environmental factors on respiratory tract microbiome and respiratory system diseases. Eur J Med Res 2025; 30:236. [PMID: 40186246 PMCID: PMC11970004 DOI: 10.1186/s40001-025-02517-3] [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: 10/16/2024] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
The respiratory tract microbiome, a complex ecosystem of microorganisms colonizing the respiratory mucous layers and epithelial surfaces along with their associated microenvironment, plays a vital role in maintaining respiratory function and promoting the maturation of the respiratory immune system. Current research suggests that environmental changes can disrupt the respiratory microbiota, potentially leading to disease. This review summarizes existing research on the impact of environmental factors on the respiratory microbiome and associated diseases, aiming to offer new insights into the prevention and treatment of respiratory disease.
Collapse
Affiliation(s)
- Yutao Ge
- Emergency Department of West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Guo Tang
- Emergency Department of West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Yawen Fu
- Emergency Department of West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Peng Deng
- Emergency Department of West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Rong Yao
- Emergency Department of West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
3
|
Mikhail SG, O'Dwyer DN. The lung microbiome in interstitial lung disease. Breathe (Sheff) 2025; 21:240167. [PMID: 40255291 PMCID: PMC12004254 DOI: 10.1183/20734735.0167-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/17/2025] [Indexed: 04/22/2025] Open
Abstract
Interstitial lung disease (ILD) is a heterogeneous chronic form of lung disease. The pathogenesis of ILD is poorly understood and a common form of ILD, idiopathic pulmonary fibrosis (IPF) is associated with poor prognosis. There is evidence for substantial dysregulated immune responses in ILD. The microbiome is a key regulator of the immune response, and the lung microbiome correlates with alveolar immunity and clinical outcomes in ILD. Most observational lung microbiome studies have been conducted in patients with IPF. A consistent observation in these studies is that the bacterial burden of the lung is elevated in patients with IPF and predicts mortality. However, our understanding of the mechanism is incomplete and our understanding of the role of the lung microbiome in other forms of ILD is limited. The microbiomes of the oropharynx and gut may have implications for the lung microbiome and pulmonary immunity in ILD but require substantial further research. Here, we discuss the studies supporting a role for the lung microbiome in the pathogenesis of IPF, and briefly describe the putative role of the oral-lung axis and the gut-lung axis in ILD.
Collapse
Affiliation(s)
- Sheridan G. Mikhail
- Division of Pulmonary and Critical Care Medicine, Dept. of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David N. O'Dwyer
- Division of Pulmonary and Critical Care Medicine, Dept. of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Dept. of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
4
|
Blondeau JM, Fitch SD. Effects of Comparative Killing by Pradofloxacin and Seven Other Antimicrobials Against Varying Bacterial Densities of Swine Isolates of Pasteurella multocida. Microorganisms 2025; 13:221. [PMID: 40005588 PMCID: PMC11857592 DOI: 10.3390/microorganisms13020221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/27/2024] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Bacterial killing is important for recovering from infection. Pasteurella multocida is a key bacterial pathogen causing swine respiratory disease and is associated with substantial mortality. Antimicrobial therapy remains an important therapeutic intervention for treating infected animals. Pradofloxacin (fluoroquinolone) is the most recently approved antimicrobial agent for treating pigs with swine respiratory disease. We compared in vitro killing of swine P. multocida strains by pradofloxacin in comparison to ceftiofur, enrofloxacin, florfenicol, marbofloxacin, tildipirosin, tilmicosin, and tulathromycin over a range of bacterial densities and four clinically relevant drug concentrations. Pradofloxacin killed 92-96.9% of cells across 106-108 cfu/mL densities at the mutant prevention drug concentration following 2-24 h of drug exposure, 96.9-98.9% of cells across 106-109 cfu/mL at the maximum serum drug concentration following 30 min of drug exposure, increasing to 99.9-100% kill following 12-24 h of drug exposure. At the maximum tissue drug concentration and against bacterial densities of 106-109 cfu/mL, pradofloxacin killed 91.3-99.8% of cells following 2 h of drug exposure, which increased to 99.9-100% kill following 12-24 h of drug exposure. Pradofloxacin was rapidly bactericidal across a range of bacterial densities and at clinically relevant drug concentrations. Pradofloxacin will be an important antibiotic for treating pigs with swine respiratory disease and where clinically indicated.
Collapse
Affiliation(s)
- Joseph M. Blondeau
- Department of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, SK S7N 0W8, Canada;
- Pathology and Laboratory Medicine and Ophthalmology, Departments of Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Shantelle D. Fitch
- Department of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, SK S7N 0W8, Canada;
| |
Collapse
|
5
|
Matics Z, Bardóczi A, Galkó C, Szabó B, Gede N, Molnár Z, Duray G, Turan C, Hegyi P, Horváth G, Müller V. Treatable traits in idiopathic pulmonary fibrosis: focus on respiratory tract infections-a systematic review and a meta-analysis. EClinicalMedicine 2025; 79:102966. [PMID: 39720602 PMCID: PMC11665700 DOI: 10.1016/j.eclinm.2024.102966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 12/26/2024] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a progressive, deadly lung disease with several factors, including respiratory tract infections (RTI), for disease worsening. There's no comprehensive data on RTI incidence in IPF patients across different therapies, including antifibrotic (nintedanib or pirfenidone), investigative or placebo treatments. Methods A systematic search of databases Medline, EMBASE, Cochrane Central, Web of Science and Scopus was conducted on September 30th 2024 (PROSPERO registration number: CRD42023484213). Only randomized controlled trials of drugs intended for IPF treatment in adults and reporting RTI incidence were included. Pooled risk ratio with 95% confidence interval (CI), risk of bias, GRADE and CINEMA assessments were conducted along with subgroup analyses for upper and lower RTI and for different antifibrotic doses. Findings A total of 27 trials of different drugs aimed for IPF therapy were pooled in a pairwise meta-analysis, 11,542 patients were analyzed with an overall number of 4156 RTI events, representing an average incidence of 38.4 ± 23.5%. Most therapies did not affect RTI risk in IPF, although single trials with everolimus and trimethoprim/sulfamethoxazole showed a significant decrease compared to placebo. For antifibrotics, RTI incidence was similar with pirfenidone treatment compared to nintedanib (RR: 0.98 CI: [0.71; 1.36]) and compared to placebo (RR: 0.88 CI: [0.69; 1.10]) and nintedanib compared to placebo (RR: 0.89 CI: [0.71; 1.12]). Interpretation RTIs are frequently reported adverse events in IPF patients over a one-year period, with different investigated treatments showing no profound impact compared to placebo. Future clinical trials should focus on targeting treatable traits like RTIs. Funding None.
Collapse
Affiliation(s)
- Zsombor Matics
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Bardóczi
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Csongor Galkó
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Bence Szabó
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Noémi Gede
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Zsolt Molnár
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Poznan University of Medical Sciences, Poznan, Poland
| | - Gábor Duray
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Cardiology, Central Hospital of Northern Pest - Military Hospital, Budapest, Hungary
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Caner Turan
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Gábor Horváth
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Veronika Müller
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
6
|
Guo X, Xu K, Wang Q, Han Z, Yu G. Assessing the impact of triiodothyronine treatment on the lung microbiome of mice with pulmonary fibrosis. BMC Pulm Med 2024; 24:405. [PMID: 39180004 PMCID: PMC11344337 DOI: 10.1186/s12890-024-03214-3] [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: 11/22/2023] [Accepted: 08/12/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF), an interstitial lung disease, is characterized by the exacerbation of progressive pulmonary fibrosis (PF). IPF primarily affects older individuals and can lead to respiratory failure. This study aimed to assess the effects of triiodothyronine (T3) treatment on the lung microbiome of mice with PF. METHODS Mice were perfused with bleomycin (BLM) to establish a PF model. Using a randomized design, 40 female specific pathogen-free (SPF) C57BL6/N mice were divided into four groups: saline, saline + T3, BLM, and BLM + T3. Histological morphology was assessed through Hematoxylin and Eosin staining as well as Masson's Trichrome staining. For the identification of lung bacteria, 16S rRNA gene sequencing was employed. An Enzyme-Linked Immunosorbent Assay was used to measure total T3 (TT3), free T3 (FT3, and reverse T3 (rT3) levels in the peripheral serum. RESULTS T3 treatment ameliorated BLM-induced lung fibrosis and structural damage. The microbiome experienced a decrease in the abundance of Proteobacteria, Bacteroides, and Actinomycetes and an increase in the abundance of Firmicutes when exposed to BLM; however, T3 treatment reversed this effect. The four groups showed no significant difference in alpha microbiome diversity (P > 0.05). Serum concentrations of TT3 and FT3 were positively correlated with microbiome abundance (P < 0.05). Administration of T3 enhanced the microbiota in PF without affecting the diversity and biological functions of the microbiome (P > 0.05). CONCLUSION The administration of T3 demonstrated a favorable impact on the lung microbiota of mice afflicted with PF, thereby partially substantiating the potential role of T3 as a therapeutic agent in the management of PF.
Collapse
Affiliation(s)
- Xiaoshu Guo
- State Key Laboratory of Cell Differentiation and Regulation; Henan International Joint Laboratory of Pulmonary Fibrosis; Henan Center for Outstanding Overseas Scientists of Organ Fibrosis; Pingyuan Laboratory; College of Life Science , Henan Normal University, No.46 Jianshe Road, Xinxiang City, 453007, Henan, China.
- Department of Physiology, Department of Fundamental Medicine, Changzhi Medical College, Changzhi, 046000, Shanxi, China.
| | - Kai Xu
- State Key Laboratory of Cell Differentiation and Regulation; Henan International Joint Laboratory of Pulmonary Fibrosis; Henan Center for Outstanding Overseas Scientists of Organ Fibrosis; Pingyuan Laboratory; College of Life Science , Henan Normal University, No.46 Jianshe Road, Xinxiang City, 453007, Henan, China
| | - Qiwen Wang
- State Key Laboratory of Cell Differentiation and Regulation; Henan International Joint Laboratory of Pulmonary Fibrosis; Henan Center for Outstanding Overseas Scientists of Organ Fibrosis; Pingyuan Laboratory; College of Life Science , Henan Normal University, No.46 Jianshe Road, Xinxiang City, 453007, Henan, China
| | - Zongyuan Han
- State Key Laboratory of Cell Differentiation and Regulation; Henan International Joint Laboratory of Pulmonary Fibrosis; Henan Center for Outstanding Overseas Scientists of Organ Fibrosis; Pingyuan Laboratory; College of Life Science , Henan Normal University, No.46 Jianshe Road, Xinxiang City, 453007, Henan, China
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation; Henan International Joint Laboratory of Pulmonary Fibrosis; Henan Center for Outstanding Overseas Scientists of Organ Fibrosis; Pingyuan Laboratory; College of Life Science , Henan Normal University, No.46 Jianshe Road, Xinxiang City, 453007, Henan, China.
| |
Collapse
|
7
|
O’Dwyer DN, Noth I, Oldham JM. Reply to Fujimoto et al.: Leveraging Microbiome Composition Variability for Precision Medicine in Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2024; 210:528-530. [PMID: 38271703 PMCID: PMC11351812 DOI: 10.1164/rccm.202312-2262le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/25/2024] [Indexed: 01/27/2024] Open
Affiliation(s)
- David N. O’Dwyer
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; and
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Justin M. Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; and
| |
Collapse
|
8
|
Bongers KS, Massett A, O'Dwyer DN. The Oral-Lung Microbiome Axis in Connective Tissue Disease-Related Interstitial Lung Disease. Semin Respir Crit Care Med 2024; 45:449-458. [PMID: 38626906 DOI: 10.1055/s-0044-1785673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
Connective tissue disease-related interstitial lung disease (CTD-ILD) is a frequent and serious complication of CTD, leading to high morbidity and mortality. Unfortunately, its pathogenesis remains poorly understood; however, one intriguing contributing factor may be the microbiome of the mouth and lungs. The oral microbiome, which is a major source of the lung microbiome through recurrent microaspiration, is altered in ILD patients. Moreover, in recent years, several lines of evidence suggest that changes in the oral and lung microbiota modulate the pulmonary immune response and thus may play a role in the pathogenesis of ILDs, including CTD-ILD. Here, we review the existing data demonstrating oral and lung microbiota dysbiosis and possible contributions to the development of CTD-ILD in rheumatoid arthritis, Sjögren's syndrome, systemic sclerosis, and systemic lupus erythematosus. We identify several areas of opportunity for future investigations into the role of the oral and lung microbiota in CTD-ILD.
Collapse
Affiliation(s)
- Kale S Bongers
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Angeline Massett
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - David N O'Dwyer
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
9
|
Combs MP, Luth JE, Falkowski NR, Wheeler DS, Walker NM, Erb-Downward JR, Wakeam E, Sjoding MW, Dunlap DG, Admon AJ, Dickson RP, Lama VN. The Lung Microbiome Predicts Mortality and Response to Azithromycin in Lung Transplant Recipients with Chronic Rejection. Am J Respir Crit Care Med 2024; 209:1360-1375. [PMID: 38271553 PMCID: PMC11146567 DOI: 10.1164/rccm.202308-1326oc] [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: 08/29/2023] [Accepted: 01/24/2024] [Indexed: 01/27/2024] Open
Abstract
Rationale: Chronic lung allograft dysfunction (CLAD) is the leading cause of death after lung transplant, and azithromycin has variable efficacy in CLAD. The lung microbiome is a risk factor for developing CLAD, but the relationship between lung dysbiosis, pulmonary inflammation, and allograft dysfunction remains poorly understood. Whether lung microbiota predict outcomes or modify treatment response after CLAD is unknown. Objectives: To determine whether lung microbiota predict post-CLAD outcomes and clinical response to azithromycin. Methods: Retrospective cohort study using acellular BAL fluid prospectively collected from recipients of lung transplant within 90 days of CLAD onset. Lung microbiota were characterized using 16S rRNA gene sequencing and droplet digital PCR. In two additional cohorts, causal relationships of dysbiosis and inflammation were evaluated by comparing lung microbiota with CLAD-associated cytokines and measuring ex vivo P. aeruginosa growth in sterilized BAL fluid. Measurements and Main Results: Patients with higher bacterial burden had shorter post-CLAD survival, independent of CLAD phenotype, azithromycin treatment, and relevant covariates. Azithromycin treatment improved survival in patients with high bacterial burden but had negligible impact on patients with low or moderate burden. Lung bacterial burden was positively associated with CLAD-associated cytokines, and ex vivo growth of P. aeruginosa was augmented in BAL fluid from transplant recipients with CLAD. Conclusions: In recipients of lung transplants with chronic rejection, increased lung bacterial burden is an independent risk factor for mortality and predicts clinical response to azithromycin. Lung bacterial dysbiosis is associated with alveolar inflammation and may be promoted by underlying lung allograft dysfunction.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Elliot Wakeam
- Division of Thoracic Surgery, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Michael W. Sjoding
- Division of Pulmonary and Critical Care and
- Weil Institute for Critical Care Research and Innovation, Ann Arbor, Michigan
| | - Daniel G. Dunlap
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Andrew J. Admon
- Division of Pulmonary and Critical Care and
- Weil Institute for Critical Care Research and Innovation, Ann Arbor, Michigan
| | - Robert P. Dickson
- Division of Pulmonary and Critical Care and
- Weil Institute for Critical Care Research and Innovation, Ann Arbor, Michigan
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan; and
| | - Vibha N. Lama
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
10
|
Darawshy F, Molyneaux PL, Segal LN. Looking Beyond the Lower Airways for Microbes Affecting Pulmonary Fibrosis. Am J Respir Crit Care Med 2024; 209:1054-1056. [PMID: 38227734 PMCID: PMC11092958 DOI: 10.1164/rccm.202312-2255ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/16/2024] [Indexed: 01/18/2024] Open
Affiliation(s)
- Fares Darawshy
- New York University Grossman School of Medicine NYU Langone Health New York, New York
- Pulmonology Institute Hadassah Medical Center Jerusalem, Israel
- Faculty of Medicine Hebrew University of Jerusalem Jerusalem, Israel
| | - Philip L Molyneaux
- Interstitial Lung Disease Unit Royal Brompton Hospital London, United Kingdom
- National Heart and Lung Institute Imperial College London, United Kingdom
| | - Leopoldo N Segal
- New York University Grossman School of Medicine NYU Langone Health New York, New York
| |
Collapse
|
11
|
Sakamachi Y, Wiley E, Solis A, Johnson CG, Meng X, Hussain S, Lipinski JH, O'Dwyer DN, Randall T, Malphurs J, Papas B, Wu BG, Li Y, Kugler M, Mehta S, Trempus CS, Thomas SY, Li JL, Zhou L, Karmaus PW, Fessler MB, McGrath JA, Gibson K, Kass DJ, Gleiberman A, Walts A, Invernizzi R, Molyneaux PL, Yang IV, Zhang Y, Kaminski N, Segal LN, Schwartz DA, Gudkov AV, Garantziotis S. Toll-Like-Receptor 5 protects against pulmonary fibrosis by reducing lung dysbiosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591719. [PMID: 39605370 PMCID: PMC11601505 DOI: 10.1101/2024.04.30.591719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating pulmonary disease with no curative treatment other than lung transplantation. IPF results from maladaptive responses to lung epithelial injury, but the underlying mechanisms remain unclear. Here, we show that deficiency in the innate immune receptor, toll-like receptor 5 (TLR5), is associated with IPF in humans and with increased susceptibility to epithelial injury and experimental fibrosis in mice, while activation of lung epithelial TLR5 through a synthetic flagellin analogue protects from experimental fibrosis. Mechanistically, epithelial TLR5 activation induces antimicrobial gene expression and ameliorates dysbiosis after lung injury. In contrast, TLR5 deficiency in mice and IPF patients is associated with lung dysbiosis. Elimination of the microbiome in mice through antibiotics abolishes the protective effect of TLR5 and reconstitution of the microbiome rescues the observed phenotype. In aggregate, TLR5 deficiency is associated with IPF and dysbiosis in humans and in the murine model of pulmonary fibrosis. Furthermore, TLR5 protects against pulmonary fibrosis in mice and this protection is mediated by effects on the microbiome. One-sentence summary Deficiency in the innate immune receptor TLR5 is a risk factor for pulmonary fibrosis, because TLR5 prevents microbial dysbiosis after lung injury.
Collapse
|
12
|
Lipinksi JH, Ranjan P, Dickson RP, O’Dwyer DN. The Lung Microbiome. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1269-1275. [PMID: 38560811 PMCID: PMC11073614 DOI: 10.4049/jimmunol.2300716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/01/2024] [Indexed: 04/04/2024]
Abstract
Although the lungs were once considered a sterile environment, advances in sequencing technology have revealed dynamic, low-biomass communities in the respiratory tract, even in health. Key features of these communities-composition, diversity, and burden-are consistently altered in lung disease, associate with host physiology and immunity, and can predict clinical outcomes. Although initial studies of the lung microbiome were descriptive, recent studies have leveraged advances in technology to identify metabolically active microbes and potential associations with their immunomodulatory by-products and lung disease. In this brief review, we discuss novel insights in airway disease and parenchymal lung disease, exploring host-microbiome interactions in disease pathogenesis. We also discuss complex interactions between gut and oropharyngeal microbiota and lung immunobiology. Our advancing knowledge of the lung microbiome will provide disease targets in acute and chronic lung disease and may facilitate the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Jay H. Lipinksi
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Piyush Ranjan
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Dept. of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Robert P. Dickson
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Dept. of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
- Weil Institute for Critical Care Research and Innovation, Ann Arbor, MI, USA
| | - David N. O’Dwyer
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
13
|
Zheng L, Liu C, Wang H, Zhang J, Mao L, Dong X, Hu S, Li N, Pi D, Qiu J, Xu F, Chen C, Zou Z. Intact lung tissue and bronchoalveolar lavage fluid are both suitable for the evaluation of murine lung microbiome in acute lung injury. MICROBIOME 2024; 12:56. [PMID: 38494479 PMCID: PMC10946114 DOI: 10.1186/s40168-024-01772-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 01/30/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Accumulating clinical evidence suggests that lung microbiome is closely linked to the progression of pulmonary diseases; however, it is still controversial which specimen type is preferred for the evaluation of lung microbiome. METHODS AND RESULTS To address this issue, we established a classical acute lung injury (ALI) mice model by intratracheal instillation of lipopolysaccharides (LPS). We found that the bacterial DNA obtained from the bronchoalveolar lavage fluid (BALF), intact lung tissue [Lung(i)], lung tissue after perfused [Lung(p)], and feces of one mouse were enough for 16S rRNA sequencing, except the BALF of mice treated with phosphate buffer saline (PBS), which might be due to the biomass of lung microbiome in the BALF were upregulated in the mice treated with LPS. Although the alpha diversity among the three specimens from lungs had minimal differences, Lung(p) had higher sample-to-sample variation compared with BALF and Lung(i). Consistently, PCoA analysis at phylum level indicated that BALF was similar to Lung(i), but not Lung(p), in the lungs of mice treated with LPS, suggesting that BALF and Lung(i) were suitable for the evaluation of lung microbiome in ALI. Importantly, Actinobacteria and Firmicutes were identified as the mostly changed phyla in the lungs and might be important factors involved in the gut-lung axis in ALI mice. Moreover, Actinobacteria and Proteobacteria might play indicative roles in the severity of lung injury. CONCLUSION This study shows both Lung(i) and BALF are suitable for the evaluation of murine lung microbiome in ALI, and several bacterial phyla, such as Actinobacteria, may serve as potential biomarkers for the severity of ALI. Video Abstract.
Collapse
Affiliation(s)
- Lijun Zheng
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Chengjun Liu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Hongjing Wang
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing, 400016, People's Republic of China
| | - Lejiao Mao
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xiaomei Dong
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Siyao Hu
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Na Li
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Dandan Pi
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Jingfu Qiu
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Feng Xu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing, 400016, People's Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
- Research Center for Environment and Human Health, School of Public Health, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
14
|
Mannion JM, McLoughlin RM, Lalor SJ. The Airway Microbiome-IL-17 Axis: a Critical Regulator of Chronic Inflammatory Disease. Clin Rev Allergy Immunol 2023; 64:161-178. [PMID: 35275333 PMCID: PMC10017631 DOI: 10.1007/s12016-022-08928-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2022] [Indexed: 02/07/2023]
Abstract
The respiratory tract is home to a diverse microbial community whose influence on local and systemic immune responses is only beginning to be appreciated. Increasing reports have linked changes in this microbiome to a range of pulmonary and extrapulmonary disorders, including asthma, chronic obstructive pulmonary disease and rheumatoid arthritis. Central to many of these findings is the role of IL-17-type immunity as an important driver of inflammation. Despite the crucial role played by IL-17-mediated immune responses in protection against infection, overt Th17 cell responses have been implicated in the pathogenesis of several chronic inflammatory diseases. However, our knowledge of the influence of bacteria that commonly colonise the respiratory tract on IL-17-driven inflammatory responses remains sparse. In this article, we review the current knowledge on the role of specific members of the airway microbiota in the modulation of IL-17-type immunity and discuss how this line of research may support the testing of susceptible individuals and targeting of inflammation at its earliest stages in the hope of preventing the development of chronic disease.
Collapse
Affiliation(s)
- Jenny M Mannion
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rachel M McLoughlin
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Stephen J Lalor
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
15
|
Wijsenbeek M, Suzuki A, Maher TM. Interstitial lung diseases. Lancet 2022; 400:769-786. [PMID: 35964592 DOI: 10.1016/s0140-6736(22)01052-2] [Citation(s) in RCA: 205] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 03/14/2022] [Accepted: 06/03/2022] [Indexed: 02/07/2023]
Abstract
Over 200 interstitial lung diseases, from ultra rare to relatively common, are recognised. Most interstitial lung diseases are characterised by inflammation or fibrosis within the interstitial space, the primary consequence of which is impaired gas exchange, resulting in breathlessness, diminished exercise tolerance, and decreased quality of life. Outcomes vary considerably for each of the different interstitial lung diseases. In some conditions, spontaneous reversibility or stabilisation can occur, but unfortunately in many people with interstitial lung disease, especially in those manifesting progressive pulmonary fibrosis, respiratory failure and death are a sad reality. Over the past 3 years, the field of interstitial lung disease has had important advances, with the approval of drugs to treat systemic sclerosis-associated interstitial lung disease, interstitial lung disease-associated pulmonary hypertension, and different forms of progressive pulmonary fibrosis. This Seminar provides an update on epidemiology, pathogenesis, presentation, diagnosis, disease course, and management of the interstitial lung diseases that are most frequently encountered in clinical practice. Furthermore, we describe how developments have led to a shift in the classification and treatment of interstitial lung diseases that exhibit progressive pulmonary fibrosis and summarise the latest practice-changing guidelines. We conclude with an outline of controversies, uncertainties, and future directions.
Collapse
Affiliation(s)
- Marlies Wijsenbeek
- Center for Interstitial Lung Diseases and Sarcoidosis, Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.
| | - Atsushi Suzuki
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Toby M Maher
- Hastings Centre for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
16
|
Chotirmall SH, Bogaert D, Chalmers JD, Cox MJ, Hansbro PM, Huang YJ, Molyneaux PL, O’Dwyer DN, Pragman AA, Rogers GB, Segal LN, Dickson RP. Therapeutic Targeting of the Respiratory Microbiome. Am J Respir Crit Care Med 2022; 206:535-544. [PMID: 35549655 PMCID: PMC9716896 DOI: 10.1164/rccm.202112-2704pp] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Sanjay H. Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore
| | - Debby Bogaert
- Center for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
- Department of Paediatric Immunology and Infectious Diseases, University Medical Center Utrecht, Utrecht, the Netherlands
| | - James D. Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, United Kingdom
| | - Michael J. Cox
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Philip M. Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Yvonne J. Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
| | - Philip L. Molyneaux
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - David N. O’Dwyer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Alexa A. Pragman
- Department of Medicine, Minneapolis Veterans Affairs Medical Center, Minneapolis, Minnesota
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Geraint B. Rogers
- Microbiome and Host Health, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Infection and Immunity, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Leopoldo N. Segal
- Division of Pulmonary, Critical Care, and Sleep Medicine, New York University Grossman School of Medicine, NYU Langone Health, New York, New York; and
| | - Robert P. Dickson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
- Weil Institute for Critical Care Research and Innovation, Ann Arbor, Michigan
| |
Collapse
|
17
|
Towards Treatable Traits for Pulmonary Fibrosis. J Pers Med 2022; 12:jpm12081275. [PMID: 36013224 PMCID: PMC9410230 DOI: 10.3390/jpm12081275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/24/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
Interstitial lung diseases (ILD) are a heterogeneous group of disorders, of which many have the potential to lead to progressive pulmonary fibrosis. A distinction is usually made between primarily inflammatory ILD and primarily fibrotic ILD. As recent studies show that anti-fibrotic drugs can be beneficial in patients with primarily inflammatory ILD that is characterized by progressive pulmonary fibrosis, treatment decisions have become more complicated. In this perspective, we propose that the ‘treatable trait’ concept, which is based on the recognition of relevant exposures, various treatable phenotypes (disease manifestations) or endotypes (shared molecular mechanisms) within a group of diseases, can be applied to progressive pulmonary fibrosis. These targets for medical intervention can be identified through validated biomarkers and are not necessarily related to specific diagnostic labels. Proposed treatable traits are: cigarette smoking, occupational, allergen or drug exposures, excessive (profibrotic) auto- or alloimmunity, progressive pulmonary fibrosis, pulmonary hypertension, obstructive sleep apnea, tuberculosis, exercise intolerance, exertional hypoxia, and anxiety and depression. There are also several potential traits that have not been associated with relevant outcomes or for which no effective treatment is available at present: air pollution, mechanical stress, viral infections, bacterial burden in the lungs, surfactant-related pulmonary fibrosis, telomere-related pulmonary fibrosis, the rs35705950 MUC5B promoter polymorphism, acute exacerbations, gastro-esophageal reflux, dyspnea, and nocturnal hypoxia. The ‘treatable traits’ concept can be applied in new clinical trials for patients with progressive pulmonary fibrosis and could be used for developing new treatment strategies.
Collapse
|
18
|
Campbell CD, Barnett C, Sulaiman I. A clinicians’ review of the respiratory microbiome. Breathe (Sheff) 2022; 18:210161. [PMID: 36338247 PMCID: PMC9584600 DOI: 10.1183/20734735.0161-2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/02/2022] [Indexed: 11/25/2022] Open
Abstract
The respiratory microbiome and its impact in health and disease is now well characterised. With the development of next-generation sequencing and the use of other techniques such as metabolomics, the functional impact of microorganisms in different host environments can be elucidated. It is now clear that the respiratory microbiome plays an important role in respiratory disease. In some diseases, such as bronchiectasis, examination of the microbiome can even be used to identify patients at higher risk of poor outcomes. Furthermore, the microbiome can aid in phenotyping. Finally, development of multi-omic analysis has revealed interactions between the host and microbiome in some conditions. This review, although not exhaustive, aims to outline how the microbiome is investigated, the healthy respiratory microbiome and its role in respiratory disease. The respiratory microbiome encompasses bacterial, fungal and viral communities. In health, it is a dynamic structure and dysbiotic in disease. Dysbiosis can be related to disease severity and may be utilised to predict patients at clinical risk.https://bit.ly/3pNSgnA
Collapse
|
19
|
胡 彬, 陈 佳, 李 为, 汪 景, 李 晓. [An updated review of the mechanism of fibrosis in acquired laryngotrachealstenosis]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2022; 36:310-314. [PMID: 35511628 PMCID: PMC10128174 DOI: 10.13201/j.issn.2096-7993.2022.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Indexed: 04/30/2023]
Abstract
Acquired laryngotracheal stenosis is a laryngeal obstruction disease due to pathologic scar formation. Although acquired laryngotracheal stenosis is hypothesized to be related to fibrosis, its specific mechanisms have yet to be characterized. This article reviews the latest research progress on the mechanisms of laryngotracheal fibrosis, including metabolic changes, immune cell dysregulation, extracellular matrix changes and microbiota.
Collapse
Affiliation(s)
- 彬 胡
- 上海市儿童医院 上海交通大学附属儿童医院耳鼻喉科(上海, 200333)
| | - 佳瑞 陈
- 上海市儿童医院 上海交通大学附属儿童医院耳鼻喉科(上海, 200333)
| | | | - 景 汪
- 上海市儿童医院 上海交通大学附属儿童医院耳鼻喉科(上海, 200333)
| | - 晓艳 李
- 上海市儿童医院 上海交通大学附属儿童医院耳鼻喉科(上海, 200333)
| |
Collapse
|
20
|
McElroy AN, Invernizzi R, Laskowska JW, O'Neill A, Doroudian M, Moghoofei M, Mostafaei S, Li F, Przybylski AA, O'Dwyer DN, Bowie AG, Fallon PG, Maher TM, Hogaboam CM, Molyneaux PL, Hirani N, Armstrong ME, Donnelly SC. Candidate Role for Toll-like Receptor 3 L412F Polymorphism and Infection in Acute Exacerbation of Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2022; 205:550-562. [PMID: 34985402 PMCID: PMC12042197 DOI: 10.1164/rccm.202010-3880oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/04/2022] [Indexed: 11/16/2022] Open
Abstract
Rationale: The Toll-like receptor 3 Leu412Phe (TLR3 L412F) polymorphism attenuates cellular antiviral responses and is associated with accelerated disease progression in idiopathic pulmonary fibrosis (IPF). The role of TLR3 L412F in bacterial infection in IPF or in acute exacerbations (AE) has not been reported. Objectives: To characterize the association between TLR3 L412F and AE-related death in IPF. To determine the effect of TLR3 L412F on the lung microbiome and on antibacterial TLR responses of primary lung fibroblasts from patients with IPF. Methods: TLR-mediated antibacterial and antiviral responses were quantitated in L412F wild-type and 412F-heterozygous primary lung fibroblasts from patients with IPF using ELISA, Western blot analysis, and quantitative PCR. Hierarchical heatmap analysis was employed to establish bacterial and viral clustering in nasopharyngeal lavage samples from patients with AE-IPF. 16S ribosomal RNA quantitative PCR and pyrosequencing were used to determine the effect of TLR3 L412F on the IPF lung microbiome. Measurements and Main Results: A significant increase in AE-related death in patients with 412F-variant IPF was reported. We established that 412F-heterozygous IPF lung fibroblasts have reduced antibacterial TLR responses to LPS (TLR4), Pam3CYSK4 (TLR1/2), flagellin (TLR5), and FSL-1 (TLR6/1) and have reduced responses to live Pseudomonas aeruginosa infection. Using 16S ribosomal RNA sequencing, we demonstrated that 412F-heterozygous patients with IPF have a dysregulated lung microbiome with increased frequencies of Streptococcus and Staphylococcus spp. Conclusions: This study reveals that TLR3 L412F dysregulates the IPF lung microbiome and reduces the responses of IPF lung fibroblasts to bacterial TLR agonists and live bacterial infection. These findings identify a candidate role for TLR3 L412F in viral- and bacterial-mediated AE death.
Collapse
Affiliation(s)
- Aoife N McElroy
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Rachele Invernizzi
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Joanna W Laskowska
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Andrew O'Neill
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Mohammad Doroudian
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | | | - Shayan Mostafaei
- Department of Biostatistics, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Feng Li
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Alexander A Przybylski
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - David N O'Dwyer
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, and
| | - Padraic G Fallon
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland
| | - Toby M Maher
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Royal Brompton Hospital, London, United Kingdom
| | - Cory M Hogaboam
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Philip L Molyneaux
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Royal Brompton Hospital, London, United Kingdom
| | - Nik Hirani
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Lung Fibrosis Clinic, Royal Infirmary Edinburgh, Edinburgh, United Kingdom; and
| | - Michelle E Armstrong
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Seamas C Donnelly
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Tallaght University Hospital, Tallaght, Dublin, Ireland
| |
Collapse
|
21
|
Knudsen KS, Lehmann S, Nielsen R, Tangedal S, Haaland I, Hiemstra PS, Eagan TM. The lower airways microbiome and antimicrobial peptides in idiopathic pulmonary fibrosis differ from chronic obstructive pulmonary disease. PLoS One 2022; 17:e0262082. [PMID: 34990493 PMCID: PMC8735599 DOI: 10.1371/journal.pone.0262082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 12/19/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The lower airways microbiome and host immune response in chronic pulmonary diseases are incompletely understood. We aimed to investigate possible microbiome characteristics and key antimicrobial peptides and proteins in idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD). METHODS 12 IPF patients, 12 COPD patients and 12 healthy controls were sampled with oral wash (OW), protected bronchoalveolar lavage (PBAL) and right lung protected sterile brushings (rPSB). The antimicrobial peptides and proteins (AMPs), secretory leucocyte protease inhibitor (SLPI) and human beta defensins 1 and 2 (hBD-1 & hBD-2), were measured in PBAL by enzyme linked immunosorbent assay (ELISA). The V3V4 region of the bacterial 16S rDNA gene was sequenced. Bioinformatic analyses were performed with QIIME 2. RESULTS hBD-1 levels in PBAL for IPF were lower compared with COPD. The predominant phyla in IPF were Firmicutes, Bacteroides and Actinobacteria; Proteobacteria were among top three in COPD. Differential abundance analysis at genus level showed significant differences between study groups for less abundant, mostly oropharyngeal, microbes. Alpha diversity was lower in IPF in PBAL compared to COPD (p = 0.03) and controls (p = 0.01), as well as in rPSB compared to COPD (p = 0.02) and controls (p = 0.04). Phylogenetic beta diversity showed significantly more similarity for IPF compared with COPD and controls. There were no significant correlations between alpha diversity and AMPs. CONCLUSIONS IPF differed in microbial diversity from COPD and controls, accompanied by differences in antimicrobial peptides. Beta diversity similarity between OW and PBAL in IPF may indicate that microaspiration contributes to changes in its microbiome.
Collapse
Affiliation(s)
- Kristel S. Knudsen
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Sverre Lehmann
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Rune Nielsen
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Solveig Tangedal
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ingvild Haaland
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pieter S. Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, Netherlands
| | - Tomas M. Eagan
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
22
|
Hoffman TW, van Moorsel CHM, Kazemier KM, Biesma DH, Grutters JC, van Kessel DA. Humoral Immune Status in Relation to Outcomes in Patients with Idiopathic Pulmonary Fibrosis. Lung 2021; 199:667-676. [PMID: 34714393 DOI: 10.1007/s00408-021-00488-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/22/2021] [Indexed: 11/27/2022]
Abstract
PURPOSE Idiopathic pulmonary fibrosis (IPF) is a severe fibrotic lung disease, in which inflammation is thought to only play a secondary role. Several factors associated with acute exacerbations of IPF (AE-IPF) have been identified, including infections. This study investigated whether humoral immunodeficiency or increased inflammatory markers at diagnosis were associated with AE-IPF and survival. METHODS Four-hundred-and-nine patients diagnosed with IPF between 2011 and 2017 were retrospectively included. Immune status investigations at diagnosis included measurement of serum immunoglobulins (available in 38%), leukocyte and lymphocyte subsets in blood and bronchoalveolar lavage (BAL) fluid (available in 58%), as well as response to pneumococcal vaccination (available in 64%). RESULTS Serum immunoglobulins or IgG subclass levels were below the lower limit of normal in 6%. The response to pneumococcal vaccination was severely impaired in 1%. Thirteen percent of patients developed an AE-IPF (4.7% per year). AE-IPF were associated with elevated lymphocytes in BAL fluid at diagnosis (p = 0.03). Higher serum IgA and IgG at diagnosis were associated with worse survival (p = 0.01; and p = 0.04), as were an increased BAL lymphocyte percentage (p = 0.005), and higher blood leukocytes and neutrophils (p = 0.01; and p = 0.0005). In a multivariate model, only BAL lymphocyte count retained statistical significance (p = 0.007). CONCLUSION The prevalence of humoral immunodeficiencies was low in patients with IPF and not associated with AE-IPF or survival. Elevated lymphocytes in BAL were associated with the development of AE-IPF and worse survival. Higher serum immunoglobulins and immune cells in blood were also associated with worse survival. The local immune response in the lungs may be a target for future therapies.
Collapse
Affiliation(s)
- T W Hoffman
- Department of Pulmonology, St. Antonius Hospital, Koekoekslaan 1, 3435CM, Nieuwegein, The Netherlands.
| | - C H M van Moorsel
- Department of Pulmonology, St. Antonius Hospital, Koekoekslaan 1, 3435CM, Nieuwegein, The Netherlands
- Division of Heart and Lungs, University Medical Centre, Utrecht, The Netherlands
| | - K M Kazemier
- Department of Pulmonology, St. Antonius Hospital, Koekoekslaan 1, 3435CM, Nieuwegein, The Netherlands
- Division of Heart and Lungs, University Medical Centre, Utrecht, The Netherlands
- Centre for Translational Immunology, University Medical Centre, Utrecht, The Netherlands
| | - D H Biesma
- Department of Internal Medicine, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - J C Grutters
- Department of Pulmonology, St. Antonius Hospital, Koekoekslaan 1, 3435CM, Nieuwegein, The Netherlands
- Division of Heart and Lungs, University Medical Centre, Utrecht, The Netherlands
| | - D A van Kessel
- Department of Pulmonology, St. Antonius Hospital, Koekoekslaan 1, 3435CM, Nieuwegein, The Netherlands
- Division of Heart and Lungs, University Medical Centre, Utrecht, The Netherlands
| |
Collapse
|
23
|
Chen CY, Chen CH, Wang CY, Lai CC, Chao CM, Wei YF. The effect of additional antimicrobial therapy on the outcomes of patients with idiopathic pulmonary fibrosis: a systematic review and meta-analysis. Respir Res 2021; 22:243. [PMID: 34526011 PMCID: PMC8442344 DOI: 10.1186/s12931-021-01839-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/06/2021] [Indexed: 11/16/2022] Open
Abstract
Background The effect of additional antimicrobial agents on the clinical outcomes of patients with idiopathic pulmonary fibrosis (IPF) is unclear. Methods We performed comprehensive searches of randomized control trials (RCTs) that compared the clinical efficacy of additional antimicrobial agents to those of placebo or usual care in the treatment of IPF patients. The primary outcome was all-cause mortality, and the secondary outcomes were changes in forced vital capacity (FVC), diffusing capacity of the lung for carbon monoxide (DLCO), and the risk of adverse events (AEs). Results Four RCTs including a total of 1055 patients (528 receiving additional antibiotics and 527 receiving placebo or usual care) were included in this meta-analysis. Among the study group, 402 and 126 patients received co-trimoxazole and doxycycline, respectively. The all-cause mortality rates were 15.0% (79/528) and 14.0% (74/527) in the patients who did and did not receive additional antibiotics, respectively (odds ratio [OR] 1.07; 95% confidence interval [CI] 0.76 to 1.51; p = 0.71). No significant difference was observed in the changes in FVC (mean difference [MD], 0.01; 95% CI − 0.03 to 0.05; p = 0.56) and DLCO (MD, 0.05; 95% CI − 0.17 to 0.28; p = 0.65). Additional use of antimicrobial agents was also associated with an increased risk of AEs (OR 1.65; 95% CI 1.19 to 2.27; p = 0.002), especially gastrointestinal disorders (OR 1.54; 95% CI 1.10 to 2.15; p = 0.001). Conclusions In patients with IPF, adding antimicrobial therapy to usual care did not improve mortality or lung function decline but increased gastrointestinal toxicity. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01839-0.
Collapse
Affiliation(s)
- Ching-Yi Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chao-Hsien Chen
- Division of Pulmonary, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Cheng-Yi Wang
- Department of Internal Medicine, Cardinal Tien Hospital and School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chih-Cheng Lai
- Department of Internal Medicine, Kaohsiung Veterans General Hospital, Tainan Branch, Tainan, Taiwan
| | - Chien-Ming Chao
- Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| | - Yu-Feng Wei
- Department of Internal Medicine, E-Da Cancer Hospital, Yan-Chao District, No. 21, Yida Road, Jiao-su Village, Kaohsiung, 824, Taiwan. .,School of Medicine for International Students, College of Medicine, and Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung, Taiwan.
| |
Collapse
|
24
|
Abstract
The healthy lung was long thought of as sterile, but recent advances using molecular sequencing approaches have detected bacteria at low levels. Healthy lung bacteria largely reflect communities present in the upper respiratory tract that enter the lung via microaspiration, which is balanced by mechanical and immune clearance and likely involves limited local replication. The nature and dynamics of the lung microbiome, therefore, differ from those of ecological niches with robust self-sustaining microbial communities. Aberrant populations (dysbiosis) have been demonstrated in many pulmonary diseases not traditionally considered microbial in origin, and potential pathways of microbe-host crosstalk are emerging. The question now is whether and how dysbiotic microbiota contribute to initiation or perpetuation of injury. The fungal microbiome and virome are less well studied. This Review highlights features of the lung microbiome, unique considerations in studying it, examples of dysbiosis in selected disease, emerging concepts in lung microbiome-host interactions, and critical areas for investigation.
Collapse
|
25
|
Baker JM, Dickson RP. Is the lung microbiome alive? Lessons from Antarctic soil. Eur Respir J 2021; 58:58/1/2100321. [PMID: 34326174 DOI: 10.1183/13993003.00321-2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 01/22/2023]
Affiliation(s)
- Jennifer M Baker
- Division of Pulmonary and Critical Care Medicine, Dept of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.,Dept of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Robert P Dickson
- Division of Pulmonary and Critical Care Medicine, Dept of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA .,Dept of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.,Michigan Center for Integrative Research in Critical Care, Ann Arbor, MI, USA
| |
Collapse
|
26
|
Seixas S, Kolbe AR, Gomes S, Sucena M, Sousa C, Vaz Rodrigues L, Teixeira G, Pinto P, Tavares de Abreu T, Bárbara C, Semedo J, Mota L, Carvalho AS, Matthiesen R, Marques PI, Pérez-Losada M. Comparative analysis of the bronchoalveolar microbiome in Portuguese patients with different chronic lung disorders. Sci Rep 2021; 11:15042. [PMID: 34294826 PMCID: PMC8298389 DOI: 10.1038/s41598-021-94468-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/09/2021] [Indexed: 01/04/2023] Open
Abstract
The lung is inhabited by a diverse microbiome that originates from the oropharynx by a mechanism of micro-aspiration. Its bacterial biomass is usually low; however, this condition shifts in lung cancer (LC), chronic obstructive pulmonary disease (COPD) and interstitial lung disease (ILD). These chronic lung disorders (CLD) may coexist in the same patient as comorbidities and share common risk factors, among which the microbiome is included. We characterized the microbiome of 106 bronchoalveolar lavages. Samples were initially subdivided into cancer and non-cancer and high-throughput sequenced for the 16S rRNA gene. Additionally, we used a cohort of 25 CLD patients where crossed comorbidities were excluded. Firmicutes, Proteobacteria and Bacteroidetes were the most prevalent phyla independently of the analyzed group. Streptococcus and Prevotella were associated with LC and Haemophilus was enhanced in COPD versus ILD. Although no significant discrepancies in microbial diversity were observed between cancer and non-cancer samples, statistical tests suggested a gradient across CLD where COPD and ILD displayed the highest and lowest alpha diversities, respectively. Moreover, COPD and ILD were separated in two clusters by the unweighted UniFrac distance (P value = 0.0068). Our results support the association of Streptoccocus and Prevotella with LC and of Haemophilus with COPD, and advocate for specific CLD signatures.
Collapse
Affiliation(s)
- Susana Seixas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal. .,Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.
| | - Allison R Kolbe
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Sílvia Gomes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal
| | - Maria Sucena
- Pneumology Department, Centro Hospitalar de São João (CHSJ), Porto, Portugal
| | - Catarina Sousa
- Pneumology Department, Centro Hospitalar de São João (CHSJ), Porto, Portugal
| | - Luís Vaz Rodrigues
- Department of Pneumology, Unidade Local de Saúde da Guarda (USLGuarda), Guarda, Portugal
| | - Gilberto Teixeira
- Department of Pneumology, Centro Hospitalar Do Baixo Vouga (CHBV), Aveiro, Portugal
| | - Paula Pinto
- Unidade de Pneumologia de Intervenção, Hospital Pulido Valente, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon, Portugal.,Instituto de Saúde Ambiental, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Tiago Tavares de Abreu
- Unidade de Pneumologia de Intervenção, Hospital Pulido Valente, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon, Portugal
| | - Cristina Bárbara
- Unidade de Pneumologia de Intervenção, Hospital Pulido Valente, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon, Portugal.,Instituto de Saúde Ambiental, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Júlio Semedo
- Unidade de Pneumologia de Intervenção, Hospital Pulido Valente, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon, Portugal
| | - Leonor Mota
- Unidade de Pneumologia de Intervenção, Hospital Pulido Valente, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon, Portugal
| | - Ana Sofia Carvalho
- Computational and Experimental Biology Group, CEDOC, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Rune Matthiesen
- Computational and Experimental Biology Group, CEDOC, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Patrícia Isabel Marques
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal
| | - Marcos Pérez-Losada
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA.,CIBIO-InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, Campus Agrário de Vairão, Vairão, Portugal
| |
Collapse
|
27
|
Duckworth A, Longhurst HJ, Paxton JK, Scotton CJ. The Role of Herpes Viruses in Pulmonary Fibrosis. Front Med (Lausanne) 2021; 8:704222. [PMID: 34368196 PMCID: PMC8339799 DOI: 10.3389/fmed.2021.704222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 06/24/2021] [Indexed: 12/29/2022] Open
Abstract
Pulmonary fibrosis (PF) is a serious lung disease which can result from known genetic or environmental exposures but is more commonly idiopathic (IPF). In familial PF (FPF), the majority of identified causal genes play key roles in the maintenance of telomeres, the protective end structures of chromosomes. Recent evidence suggests that short telomeres may also be implicated causally in a significant proportion of idiopathic cases. The possible involvement of herpes viruses in PF disease incidence and progression has been examined for many years, with some studies showing strong, statistically significant associations and others reporting no involvement. Evidence is thus polarized and remains inconclusive. Here we review the reported involvement of herpes viruses in PF in both animals and humans and present a summary of the evidence to date. We also present several possible mechanisms of action of the different herpes viruses in PF pathogenesis, including potential contributions to telomere attrition and cellular senescence. Evidence for antiviral treatment in PF is very limited but suggests a potential benefit. Further work is required to definitely answer the question of whether herpes viruses impact PF disease onset and progression and to enable the possible use of targeted antiviral treatments to improve clinical outcomes.
Collapse
Affiliation(s)
- Anna Duckworth
- College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Hilary J. Longhurst
- Department of Medicine, University of Auckland, Auckland, New Zealand
- Dyskeratosis Congenita (DC) Action, London, United Kingdom
| | - Jane K. Paxton
- Dyskeratosis Congenita (DC) Action, London, United Kingdom
| | - Chris J. Scotton
- College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
28
|
Trachalaki A, Tsitoura E, Mastrodimou S, Invernizzi R, Vasarmidi E, Bibaki E, Tzanakis N, Molyneaux PL, Maher TM, Antoniou K. Enhanced IL-1β Release Following NLRP3 and AIM2 Inflammasome Stimulation Is Linked to mtROS in Airway Macrophages in Pulmonary Fibrosis. Front Immunol 2021; 12:661811. [PMID: 34220810 PMCID: PMC8248801 DOI: 10.3389/fimmu.2021.661811] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/21/2021] [Indexed: 12/20/2022] Open
Abstract
Fibrotic Interstitial lung diseases (ILDs) are complex disorders of variable clinical behaviour. The majority of them cause significant morbidity, whilst Idiopathic Pulmonary Fibrosis (IPF) is recognised as the most relentless. NLRP3, AIM2, and NLRC4 inflammasomes are multiprotein complexes driving IL-1β release; a proinflammatory and profibrotic cytokine. Several pathogenetic factors associated with IPF are identified as inflammasome activators, including increases in mtROS and bacterial burden. Mitochondrial oxidation and alterations in bacterial burden in IPF and other ILDs may lead to augmented inflammasome activity in airway macrophages (AMs). IPF (n=14), non-IPF-ILDs (n=12) patients and healthy subjects (n=12) were prospectively recruited and AMs were isolated from bronchoalveolar lavage. IL-1β release resulting from NLRP3, AIM2 and NLRC4 inflammasomes stimulation in AMs were determined and baseline levels of mitochondrial ROS and microbial burden were also measured. Our results showed that NLRP3 was more inducible in IPF and other ILDs compared to controls. Additionally, following AIM2 activation IL-1β release was significantly higher in IPF compared to controls, whereas similar trends were observed in Non-IPF-ILDs. NLRC4 activation was similar across groups. mtROS was significantly associated with heightened NLRP3 and AIM2 activation, and mitochondrial antioxidant treatment limited inflammasome activation. Importantly, microbial burden was linked to baseline IL-1β release and AIM2 and IL-18 relative expression independently of mtROS. In conclusion, the above findings suggested a link between the overactivation of NLRP3 and AIM2 inflammasomes, driven by mitochondrial oxidation, in the pathogenesis of lung fibrosis while changes in the microbiota may prime the inflammasome in the lungs.
Collapse
Affiliation(s)
- Athina Trachalaki
- Laboratory of Molecular and Cellular Pneumonology, Respiratory Medicine Department, School of Medicine, University of Crete, Heraklion, Greece.,National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Royal Brompton Hospital, London, United Kingdom
| | - Eliza Tsitoura
- Laboratory of Molecular and Cellular Pneumonology, Respiratory Medicine Department, School of Medicine, University of Crete, Heraklion, Greece
| | - Semeli Mastrodimou
- Laboratory of Molecular and Cellular Pneumonology, Respiratory Medicine Department, School of Medicine, University of Crete, Heraklion, Greece
| | - Rachele Invernizzi
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Eirini Vasarmidi
- Laboratory of Molecular and Cellular Pneumonology, Respiratory Medicine Department, School of Medicine, University of Crete, Heraklion, Greece
| | - Eleni Bibaki
- Laboratory of Molecular and Cellular Pneumonology, Respiratory Medicine Department, School of Medicine, University of Crete, Heraklion, Greece
| | - Nikolaos Tzanakis
- Laboratory of Molecular and Cellular Pneumonology, Respiratory Medicine Department, School of Medicine, University of Crete, Heraklion, Greece
| | - Philip L Molyneaux
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Royal Brompton Hospital, London, United Kingdom
| | - Toby M Maher
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Royal Brompton Hospital, London, United Kingdom
| | - Katerina Antoniou
- Laboratory of Molecular and Cellular Pneumonology, Respiratory Medicine Department, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
29
|
Baker JM, Hinkle KJ, McDonald RA, Brown CA, Falkowski NR, Huffnagle GB, Dickson RP. Whole lung tissue is the preferred sampling method for amplicon-based characterization of murine lung microbiota. MICROBIOME 2021; 9:99. [PMID: 33952355 PMCID: PMC8101028 DOI: 10.1186/s40168-021-01055-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/22/2021] [Indexed: 05/06/2023]
Abstract
BACKGROUND Low-biomass microbiome studies (such as those of the lungs, placenta, and skin) are vulnerable to contamination and sequencing stochasticity, which obscure legitimate microbial signal. While human lung microbiome studies have rigorously identified sampling strategies that reliably capture microbial signal from these low-biomass microbial communities, the optimal sampling strategy for characterizing murine lung microbiota has not been empirically determined. Performing accurate, reliable characterization of murine lung microbiota and distinguishing true microbial signal from noise in these samples will be critical for further mechanistic microbiome studies in mice. RESULTS Using an analytic approach grounded in microbial ecology, we compared bacterial DNA from the lungs of healthy adult mice collected via two common sampling approaches: homogenized whole lung tissue and bronchoalveolar lavage (BAL) fluid. We quantified bacterial DNA using droplet digital PCR, characterized bacterial communities using 16S rRNA gene sequencing, and systematically assessed the quantity and identity of bacterial DNA in both specimen types. We compared bacteria detected in lung specimens to each other and to potential source communities: negative (background) control specimens and paired oral samples. By all measures, whole lung tissue in mice contained greater bacterial signal and less evidence of contamination than did BAL fluid. Relative to BAL fluid, whole lung tissue exhibited a greater quantity of bacterial DNA, distinct community composition, decreased sample-to-sample variation, and greater biological plausibility when compared to potential source communities. In contrast, bacteria detected in BAL fluid were minimally different from those of procedural, reagent, and sequencing controls. CONCLUSIONS An ecology-based analytical approach discriminates signal from noise in this low-biomass microbiome study and identifies whole lung tissue as the preferred specimen type for murine lung microbiome studies. Sequencing, analysis, and reporting of potential source communities, including negative control specimens and contiguous biological sites, are crucial for biological interpretation of low-biomass microbiome studies, independent of specimen type. Video abstract.
Collapse
Affiliation(s)
- Jennifer M Baker
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 6220 MSRB III/SPC 5642, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-5642, USA
| | - Kevin J Hinkle
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 6220 MSRB III/SPC 5642, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-5642, USA
| | - Roderick A McDonald
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 6220 MSRB III/SPC 5642, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-5642, USA
| | - Christopher A Brown
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 6220 MSRB III/SPC 5642, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-5642, USA
| | - Nicole R Falkowski
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 6220 MSRB III/SPC 5642, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-5642, USA
| | - Gary B Huffnagle
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 6220 MSRB III/SPC 5642, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-5642, USA
- Department of Molecular, Cellular, & Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Robert P Dickson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 6220 MSRB III/SPC 5642, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-5642, USA.
- Michigan Center for Integrative Research in Critical Care, Ann Arbor, MI, USA.
| |
Collapse
|
30
|
|
31
|
Invernizzi R, Wu BG, Barnett J, Ghai P, Kingston S, Hewitt RJ, Feary J, Li Y, Chua F, Wu Z, Wells AU, George PM, Renzoni EA, Nicholson AG, Rice A, Devaraj A, Segal LN, Byrne AJ, Maher TM, Lloyd CM, Molyneaux PL. The Respiratory Microbiome in Chronic Hypersensitivity Pneumonitis Is Distinct from That of Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2021; 203:339-347. [PMID: 32692582 PMCID: PMC7874329 DOI: 10.1164/rccm.202002-0460oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Rationale: Chronic hypersensitivity pneumonitis (CHP) is a condition that arises after repeated exposure and sensitization to inhaled antigens. The lung microbiome is increasingly implicated in respiratory disease, but, to date, no study has investigated the composition of microbial communities in the lower airways in CHP. Objectives: To characterize and compare the airway microbiome in subjects with CHP, subjects with idiopathic pulmonary fibrosis (IPF), and control subjects. Methods: We prospectively recruited individuals with a CHP diagnosis (n = 110), individuals with an IPF diagnosis (n = 45), and control subjects (n = 28). Subjects underwent BAL and bacterial DNA was isolated, quantified by quantitative PCR and the 16S ribosomal RNA gene was sequenced to characterize the bacterial communities in the lower airways. Measurements and Main Results: Distinct differences in the microbial profiles were evident in the lower airways of subjects with CHP and IPF. At the phylum level, the prevailing microbiota of both subjects with IPF and subjects with CHP included Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria. However, in IPF, Firmicutes dominated, whereas the percentage of reads assigned to Proteobacteria in the same group was significantly lower than the percentage found in subjects with CHP. At the genus level, the Staphylococcus burden was increased in CHP, and Actinomyces and Veillonella burdens were increased in IPF. The lower airway bacterial burden in subjects with CHP was higher than that in control subjects but lower than that of those with IPF. In contrast to IPF, there was no association between bacterial burden and survival in CHP. Conclusions: The microbial profile of the lower airways in subjects with CHP is distinct from that of IPF, and, notably, the bacterial burden in individuals with CHP fails to predict survival.
Collapse
Affiliation(s)
- Rachele Invernizzi
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Benjamin G Wu
- Division of Pulmonary and Critical Care Medicine, New York University, New York, New York; and
| | - Joseph Barnett
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Royal Brompton Hospital, London, United Kingdom
| | - Poonam Ghai
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Shaun Kingston
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Royal Brompton Hospital, London, United Kingdom
| | - Richard J Hewitt
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Royal Brompton Hospital, London, United Kingdom
| | - Johanna Feary
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Royal Brompton Hospital, London, United Kingdom
| | - Yonghua Li
- Division of Pulmonary and Critical Care Medicine, New York University, New York, New York; and
| | - Felix Chua
- Royal Brompton Hospital, London, United Kingdom
| | - Zhe Wu
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Royal Brompton Hospital, London, United Kingdom
| | | | | | | | - Andrew G Nicholson
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Royal Brompton Hospital, London, United Kingdom
| | | | - Anand Devaraj
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Royal Brompton Hospital, London, United Kingdom
| | - Leopoldo N Segal
- Division of Pulmonary and Critical Care Medicine, New York University, New York, New York; and
| | - Adam J Byrne
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Toby M Maher
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Clare M Lloyd
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Philip L Molyneaux
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Royal Brompton Hospital, London, United Kingdom
| |
Collapse
|
32
|
O'Dwyer DN, Garantziotis S. The Lung Microbiome in Health, Hypersensitivity Pneumonitis, and Idiopathic Pulmonary Fibrosis: A Heavy Bacterial Burden to Bear. Am J Respir Crit Care Med 2021; 203:281-283. [PMID: 32791003 PMCID: PMC7874306 DOI: 10.1164/rccm.202007-2822ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- David N O'Dwyer
- University of Michigan Medical School Ann Arbor, Michigan and
| | | |
Collapse
|
33
|
Pardo A, Selman M. The Interplay of the Genetic Architecture, Aging, and Environmental Factors in the Pathogenesis of Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2021; 64:163-172. [PMID: 32946290 DOI: 10.1165/rcmb.2020-0373ps] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic fibrosing lung disease of indeterminate etiology and limited therapeutic options. The initiation, development, and progression of IPF are influenced by genetic predisposition, aging, and host and environmental factors, but the magnitude of the contribution of each of them and the sequence of the pathogenic events are uncertain. Current evidence indicates that accumulated environmental exposures in a genetically predisposed individual, usually over 60 years of age, leads to phenotypic and functional alterations of the lung epithelium. Aberrant activation of epithelial cells results, through a complex release of numerous mediators, in the local expansion of peculiar subsets of aggressive fibroblasts and myofibroblasts, which are crucial effector cells of fibrotic remodeling and loss of the normal lung architecture and function. Progressive increase of the mechanical stiffness activates cell-autonomous and matrix-dependent processes contributing to the perpetuation of the fibrotic response. This Perspective provides an integral overview of the major risk factors underpinning the pathogenesis of IPF, including gene variants, aging alterations, environmental factors, host risk factors, and epigenetic reprogramming.
Collapse
Affiliation(s)
- Annie Pardo
- Facultad de Ciencias, Universidad Nacional Autónoma de México, México City, Mexico; and
| | - Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," México City, Mexico
| |
Collapse
|
34
|
Antoniou KM, Tsitoura E, Vasarmidi E, Symvoulakis EK, Aidinis V, Tzilas V, Tzouvelekis A, Bouros D. Precision medicine in idiopathic pulmonary fibrosis therapy: From translational research to patient-centered care. Curr Opin Pharmacol 2021; 57:71-80. [PMID: 33556824 DOI: 10.1016/j.coph.2020.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/22/2020] [Indexed: 12/15/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, irreversible fibrotic chronic lung disease affecting predominantly older adults, with a history of smoking. The current model of disease natural course is that recurrent injury of the alveolar epithelium in the context of advanced aging/cellular senescence is followed by defective re-epithelialization and scar tissue formation. Currently, two drugs, nintedanib and pirfenidone, that modify disease progression have been approved worldwide for the treatment of IPF. However, despite treatment, patients with IPF are not cured, and eventually, disease advances in most treated patients. Enhancing biogenomic and metabolic research output, its translation into clinical precision and optimal service delivery through patient-centeredness are key elements to support effective IPF care. In this review, we summarize therapeutic options currently investigated for IPF based on the major pathogenetic pathways and molecular targets that drive pulmonary fibrosis.
Collapse
Affiliation(s)
- Katerina M Antoniou
- Molecular & Cellular Pneumonology Laboratory, Department of Respiratory Medicine, Faculty of Medicine, University of Crete, Greece.
| | - Eliza Tsitoura
- Molecular & Cellular Pneumonology Laboratory, Department of Respiratory Medicine, Faculty of Medicine, University of Crete, Greece
| | - Eirini Vasarmidi
- Molecular & Cellular Pneumonology Laboratory, Department of Respiratory Medicine, Faculty of Medicine, University of Crete, Greece
| | | | - Vassilis Aidinis
- Division of Immunology, Alexander Fleming Biomedical Sciences Research Center, Athens, Greece
| | - Vassilis Tzilas
- Center for Diseases of the Chest, Athens Medical Center, Athens, Greece
| | | | - Demosthenes Bouros
- Center for Diseases of the Chest, Athens Medical Center, Athens, Greece; Medical School, National and Kapodistrian University of Athens, Greece
| |
Collapse
|
35
|
Lung microbiota predict chronic rejection in healthy lung transplant recipients: a prospective cohort study. THE LANCET RESPIRATORY MEDICINE 2021; 9:601-612. [PMID: 33460570 DOI: 10.1016/s2213-2600(20)30405-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Alterations in the respiratory microbiome are common in chronic lung diseases, correlate with decreased lung function, and have been associated with disease progression. The clinical significance of changes in the respiratory microbiome after lung transplant, specifically those related to development of chronic lung allograft dysfunction (CLAD), are unknown. The aim of this study was to evaluate the effect of lung microbiome characteristics in healthy lung transplant recipients on subsequent CLAD-free survival. METHODS We prospectively studied a cohort of lung transplant recipients at the University of Michigan (Ann Arbor, MI, USA). We analysed characteristics of the respiratory microbiome in acellular bronchoalveolar lavage fluid (BALF) collected from asymptomatic patients during per-protocol surveillance bronchoscopy 1 year after lung transplantation. For our primary endpoint, we evaluated a composite of development of CLAD or death at 500 days after the 1-year surveillance bronchoscopy. Our primary microbiome predictor variables were bacterial DNA burden (total 16S rRNA gene copies per mL of BALF, quantified via droplet digital PCR) and bacterial community composition (determined by bacterial 16S rRNA gene sequencing). Patients' lung function was followed serially at least every 3 months by spirometry, and CLAD was diagnosed according to International Society of Heart and Lung Transplant 2019 guidelines. FINDINGS We analysed BALF from 134 patients, collected during 1-year post-transplant surveillance bronchoscopy between Oct 21, 2005, and Aug 25, 2017. Within 500 days of follow-up from the time of BALF sampling, 24 (18%) patients developed CLAD, five (4%) died before confirmed development of CLAD, and 105 (78%) patients remained CLAD-free with complete follow-up. Lung bacterial burden was predictive of CLAD development or death within 500 days of the surveillance bronchoscopy, after controlling for demographic and clinical factors, including immunosuppression and bacterial culture results, in a multivariable survival model. This relationship was evident when burden was analysed as a continuous variable (per log10 increase in burden, HR 2·49 [95% CI 1·38-4·48], p=0·0024) or by tertiles (middle vs lowest bacterial burden tertile, HR 4·94 [1·25-19·42], p=0·022; and highest vs lowest, HR 10·56 [2·53-44·08], p=0·0012). In patients who developed CLAD or died, composition of the lung bacterial community significantly differed to that in patients who survived and remained CLAD-free (on permutational multivariate analysis of variance, p=0·047 at the taxonomic level of family), although differences in community composition were associated with bacterial burden. No individual bacterial taxa were definitively associated with CLAD development or death. INTERPRETATION Among asymptomatic lung transplant recipients at 1-year post-transplant, increased lung bacterial burden is predictive of chronic rejection and death. The lung microbiome represents an understudied and potentially modifiable risk factor for lung allograft dysfunction. FUNDING US National Institutes of Health, Cystic Fibrosis Foundation, Brian and Mary Campbell and Elizabeth Campbell Carr research gift fund.
Collapse
|
36
|
Michalski JE, Schwartz DA. Genetic Risk Factors for Idiopathic Pulmonary Fibrosis: Insights into Immunopathogenesis. J Inflamm Res 2021; 13:1305-1318. [PMID: 33447070 PMCID: PMC7801923 DOI: 10.2147/jir.s280958] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis is an etiologically complex interstitial lung disease characterized by progressive scarring of the lungs with a subsequent decline in lung function. While much of the pathogenesis of IPF still remains unclear, it is now understood that genetic variation accounts for at least one-third of the risk of developing the disease. The single-most validated and most significant risk factor, genetic or otherwise, is a gain-of-function promoter variant in the MUC5B gene. While the functional impact of these IPF risk variants at the cellular and tissue levels are areas of active investigation, there is a growing body of evidence that these genetic variants may influence disease pathogenesis through modulation of innate immune processes.
Collapse
Affiliation(s)
- Jacob E Michalski
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - David A Schwartz
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
37
|
Wilson AM, Clark AB, Cahn T, Chilvers ER, Fraser W, Hammond M, Livermore DM, Maher TM, Parfrey H, Swart AM, Stirling S, Thickett DR, Whyte M. Effect of Co-trimoxazole (Trimethoprim-Sulfamethoxazole) vs Placebo on Death, Lung Transplant, or Hospital Admission in Patients With Moderate and Severe Idiopathic Pulmonary Fibrosis: The EME-TIPAC Randomized Clinical Trial. JAMA 2020; 324:2282-2291. [PMID: 33289822 PMCID: PMC7724556 DOI: 10.1001/jama.2020.22960] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
IMPORTANCE Idiopathic pulmonary fibrosis (IPF) has a poor prognosis and limited treatment options. Patients with IPF have altered lung microbiota, with bacterial burden within the lungs associated with mortality; previous studies have suggested benefit with co-trimoxazole (trimethoprim-sulfamethoxazole). OBJECTIVE To determine the efficacy of co-trimoxazole in patients with moderate and severe IPF. DESIGN, SETTING, AND PARTICIPANTS Double-blind, placebo-controlled, parallel randomized trial of 342 patients with IPF, breathlessness (Medical Research Council dyspnea scale score >1), and impaired lung function (forced vital capacity ≤75% predicted) conducted in 39 UK specialist interstitial lung disease centers between April 2015 (first patient visit) and April 2019 (last patient follow-up). INTERVENTIONS Study participants were randomized to receive 960 mg of oral co-trimoxazole twice daily (n = 170) or matched placebo (n = 172) for between 12 and 42 months. All patients received 5 mg of folic acid orally once daily. MAIN OUTCOMES AND MEASURES The primary outcome was time to death (all causes), lung transplant, or first nonelective hospital admission. There were 15 secondary outcomes, including the individual components of the primary end point respiratory-related events, lung function (forced vital capacity and gas transfer), and patient-reported outcomes (Medical Research Council dyspnea scale, 5-level EuroQol 5-dimension questionnaire, cough severity, Leicester Cough Questionnaire, and King's Brief Interstitial Lung Disease questionnaire scores). RESULTS Among 342 individuals who were randomized (mean age, 71.3 years; 46 [13%] women), 283 (83%) completed the trial. The median (interquartile range) duration of follow-up was 1.02 (0.35-1.73) years. Events per person-year of follow-up among participants randomized to the co-trimoxazole and placebo groups were 0.45 (84/186) and 0.38 (80/209), respectively, with a hazard ratio of 1.2 ([95% CI, 0.9-1.6]; P = .32). There were no statistically significant differences in other event outcomes, lung function, or patient-reported outcomes. Patients in the co-trimoxazole group had 696 adverse events (nausea [n = 89], diarrhea [n = 52], vomiting [n = 28], and rash [n = 31]) and patients in the placebo group had 640 adverse events (nausea [n = 67], diarrhea [n = 84], vomiting [n = 20], and rash [n = 20]). CONCLUSIONS AND RELEVANCE Among patients with moderate or severe IPF, treatment with oral co-trimoxazole did not reduce a composite outcome of time to death, transplant, or nonelective hospitalization compared with placebo. TRIAL REGISTRATION ISRCTN Identifier: ISRCTN17464641.
Collapse
Affiliation(s)
- Andrew M. Wilson
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, United Kingdom
- Department of Respiratory Medicine, Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, Norfolk, United Kingdom
| | - Allan B. Clark
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, United Kingdom
| | - Tony Cahn
- Department of Respiratory Medicine, Bedford Hospitals NHS Trust, South Wing, Bedford, United Kingdom
| | - Edwin R. Chilvers
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - William Fraser
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, United Kingdom
- Department of Endocrinology and Clinical Biochemistry, Norfolk and Norwich University Hospital NHS Foundation Trust, Colney Lane, Norwich, Norfolk, United Kingdom
| | - Matthew Hammond
- Norwich Clinical Trials Unit, University of East Anglia, Norwich Research Park, Norwich, Norfolk, United Kingdom
| | - David M. Livermore
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, United Kingdom
| | - Toby M. Maher
- Department of Endocrinology and Clinical Biochemistry, Norfolk and Norwich University Hospital NHS Foundation Trust, Colney Lane, Norwich, Norfolk, United Kingdom
- Hastings Centre for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Keck School of Medicine, University of Southern California, Los Angeles
- NIHR Respiratory Clinical Research Facility, Royal Brompton Hospital, London, United Kingdom
| | - Helen Parfrey
- Royal Papworth Hospital NHS Foundation Trust, Papworth Road, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Ann Marie Swart
- Norwich Clinical Trials Unit, University of East Anglia, Norwich Research Park, Norwich, Norfolk, United Kingdom
| | - Susan Stirling
- Norwich Clinical Trials Unit, University of East Anglia, Norwich Research Park, Norwich, Norfolk, United Kingdom
| | - David R. Thickett
- Institute of Inflammation and Aging, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Moira Whyte
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | | |
Collapse
|
38
|
Valenzi E, Yang H, Sembrat JC, Yang L, Winters S, Nettles R, Kass DJ, Qin S, Wang X, Myerburg MM, Methé B, Fitch A, Alder JK, Benos PV, McVerry BJ, Rojas M, Morris A, Kitsios GD. Topographic heterogeneity of lung microbiota in end-stage idiopathic pulmonary fibrosis: the Microbiome in Lung Explants-2 (MiLEs-2) study. Thorax 2020; 76:239-247. [PMID: 33268457 DOI: 10.1136/thoraxjnl-2020-214770] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 09/18/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Lung microbiota profiles in patients with early idiopathic pulmonary fibrosis (IPF) have been associated with disease progression; however, the topographic heterogeneity of lung microbiota and their roles in advanced IPF are unknown. METHODS We performed a retrospective, case-control study of explanted lung tissue obtained at the time of lung transplantation or rapid autopsy from patients with IPF and other chronic lung diseases (connective tissue disease-associated interstitial lung disease (CTD-ILD), cystic fibrosis (CF), COPD and donor lungs unsuitable for transplant from Center for Organ Recovery and Education (CORE)). We sampled subpleural tissue and airway-based specimens (bronchial washings and airway tissue) and quantified bacterial load and profiled communities by amplification and sequencing of the 16S rRNA gene. FINDINGS Explants from 62 patients with IPF, 15 patients with CTD-ILD, 20 patients with CF, 20 patients with COPD and 20 CORE patients were included. Airway-based samples had higher bacterial load compared with distal parenchymal tissue. IPF basilar tissue had much lower bacterial load compared with CF and CORE lungs (p<0.001). No microbial community differences were found between parenchymal tissue samples from different IPF lobes. Dirichlet multinomial models revealed an IPF cluster (29%) with distinct composition, high bacterial load and low alpha diversity, exhibiting higher odds for acute exacerbation or death. INTERPRETATION IPF explants had low biomass in the distal parenchyma of all three lobes with higher bacterial load in the airways. The discovery of a distinct subgroup of patients with IPF with higher bacterial load and worse clinical outcomes supports investigation of personalised medicine approaches for microbiome-targeted interventions.
Collapse
Affiliation(s)
- Eleanor Valenzi
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Haopu Yang
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,School of Medicine, Tsinghua University, Beijing, China
| | - John C Sembrat
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Libing Yang
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,School of Medicine, Tsinghua University, Beijing, China
| | - Spencer Winters
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Bronson Adult Critical Care, Kalamazoo, Michigan, USA
| | - Rachel Nettles
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Daniel J Kass
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Shulin Qin
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xiaohong Wang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael M Myerburg
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Barbara Methé
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adam Fitch
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jonathan K Alder
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Panayiotis V Benos
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Bryan J McVerry
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mauricio Rojas
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Alison Morris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Georgios D Kitsios
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA .,Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
39
|
Verleden SE, Braubach P, Kuehnel M, Dickgreber N, Brouwer E, Tittmann P, Laenger F, Jonigk D. Molecular approach to the classification of chronic fibrosing lung disease-there and back again. Virchows Arch 2020; 478:89-99. [PMID: 33169196 DOI: 10.1007/s00428-020-02964-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/21/2020] [Accepted: 10/30/2020] [Indexed: 11/26/2022]
Abstract
Chronic diffuse parenchymal lung disease (DPLD) is an umbrella term for a very heterogeneous group of lung diseases. Over the last decades, clinical, radiological and histopathological criteria have been established to define and separate these entities. More recently the clinical utility of this approach has been challenged as a unifying concept of pathophysiological mechanisms and a shared response to therapy across the disease spectrum have been described. In this review, we discuss molecular motifs for subtyping and the prediction of prognosis focusing on genetics and markers found in the blood, lavage and tissue. As a purely molecular classification so far lacks sufficient sensitivity and specificity for subtyping, it is not routinely used and not implemented in international guidelines. However, a better molecular characterization of lung disease with a more precise identification of patients with, for example, a risk for rapid disease progression would facilitate more accurate treatment decisions and hopefully contribute to better patients' outcomes.
Collapse
Affiliation(s)
- Stijn E Verleden
- Institute of Pathology, Hannover Medical School, Hannover, Germany.
- BREATHE Lab, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Peter Braubach
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Mark Kuehnel
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Nicolas Dickgreber
- Department of Respiratory Medicine and Thoracic Oncology, Ibbenbueren General Hospital, Ibbenbueren, Germany
| | - Emily Brouwer
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Pauline Tittmann
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Florian Laenger
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Hannover, Germany
| |
Collapse
|
40
|
Lung Microbiome Differentially Impacts Survival of Patients with Non-Small Cell Lung Cancer Depending on Tumor Stroma Phenotype. Biomedicines 2020; 8:biomedicines8090349. [PMID: 32933105 PMCID: PMC7554830 DOI: 10.3390/biomedicines8090349] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 01/01/2023] Open
Abstract
The link between a lung tumor and the lung microbiome is a largely unexplored issue. To investigate the relationship between a lung microbiome and the phenotype of an inflammatory stromal infiltrate, we studied a cohort of 89 patients with non-small cell lung cancer. The microbiome was analyzed in tumor and adjacent normal tissue by 16S rRNA amplicon sequencing. Characterization of the tumor stroma was done using immunohistochemistry. We demonstrated that the bacterial load was higher in adjacent normal tissue than in a tumor (p = 0.0325) with similar patterns of taxonomic structure and alpha diversity. Lung adenocarcinomas did not differ in their alpha diversity from squamous cell carcinomas, although the content of Gram-positive bacteria increased significantly in the adenocarcinoma group (p = 0.0419). An analysis of an inflammatory infiltrate of tumor stroma showed a correlation of CD68, iNOS and FOXP3 with a histological type of tumor. For the first time we showed that high bacterial load in the tumor combined with increased iNOS expression is a favorable prognostic factor (HR = 0.1824; p = 0.0123), while high bacterial load combined with the increased number of FOXP3+ cells is a marker of poor prognosis (HR = 4.651; p = 0.0116). Thus, we established that bacterial load of the tumor has an opposite prognostic value depending on the status of local antitumor immunity.
Collapse
|
41
|
Lipinski JH, Moore BB, O'Dwyer DN. The evolving role of the lung microbiome in pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2020; 319:L675-L682. [PMID: 32877221 DOI: 10.1152/ajplung.00258.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mucosal surfaces are constantly exposed to a microbiome consisting of microorganisms that heavily influence human immunity and health. In the lung these microorganisms consist of bacteria, viruses, and fungi and exist in a relatively low biomass state. Bacterial communities of the lung modulate local inflammation and correlate with changes in pulmonary physiology and clinical outcomes in patients with lung disease. Instrumental to this progress has been the study of these bacterial communities in the pathogenesis of pulmonary fibrosis, a fatal and progressive disease culminating in respiratory failure. Key pathophysiological mechanisms in pulmonary fibrosis include recurrent idiopathic alveolar epithelial injury, unchecked collagen deposition, mucociliary dysfunction due to muc5b overexpression, hypoxia, and altered host defense. These key mechanisms and their related consequences promote severe progressive architectural lung destruction and loss of local homeostasis. As such, pulmonary fibrosis is an appropriate target disease for the study of the lung microbiome. Herein, we discuss recent advances in our understanding of the role of the lung microbiome in the pathogenesis of pulmonary fibrosis. We highlight fundamental clinical observations and mechanistic insights and identify crucial areas for further discovery science. An improved understanding of how the lung microbiome acts to influence outcomes in patients with pulmonary fibrosis will lead to enhanced therapies for this devastating lung disease.
Collapse
Affiliation(s)
- Jay H Lipinski
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
| | - David N O'Dwyer
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
42
|
Tielemans B, Dekoster K, Verleden SE, Sawall S, Leszczyński B, Laperre K, Vanstapel A, Verschakelen J, Kachelriess M, Verbeken E, Swoger J, Vande Velde G. From Mouse to Man and Back: Closing the Correlation Gap between Imaging and Histopathology for Lung Diseases. Diagnostics (Basel) 2020; 10:E636. [PMID: 32859103 PMCID: PMC7554749 DOI: 10.3390/diagnostics10090636] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023] Open
Abstract
Lung diseases such as fibrosis, asthma, cystic fibrosis, infection and cancer are life-threatening conditions that slowly deteriorate quality of life and for which our diagnostic power is high, but our knowledge on etiology and/or effective treatment options still contains important gaps. In the context of day-to-day practice, clinical and preclinical studies, clinicians and basic researchers team up and continuously strive to increase insights into lung disease progression, diagnostic and treatment options. To unravel disease processes and to test novel therapeutic approaches, investigators typically rely on end-stage procedures such as serum analysis, cyto-/chemokine profiles and selective tissue histology from animal models. These techniques are useful but provide only a snapshot of disease processes that are essentially dynamic in time and space. Technology allowing evaluation of live animals repeatedly is indispensable to gain a better insight into the dynamics of lung disease progression and treatment effects. Computed tomography (CT) is a clinical diagnostic imaging technique that can have enormous benefits in a research context too. Yet, the implementation of imaging techniques in laboratories lags behind. In this review we want to showcase the integrated approaches and novel developments in imaging, lung functional testing and pathological techniques that are used to assess, diagnose, quantify and treat lung disease and that may be employed in research on patients and animals. Imaging approaches result in often novel anatomical and functional biomarkers, resulting in many advantages, such as better insight in disease progression and a reduction in the numbers of animals necessary. We here showcase integrated assessment of lung disease with imaging and histopathological technologies, applied to the example of lung fibrosis. Better integration of clinical and preclinical imaging technologies with pathology will ultimately result in improved clinical translation of (therapy) study results.
Collapse
Affiliation(s)
- Birger Tielemans
- Department of Imaging and Pathology, KU Leuven, University of Leuven, 3000 Leuven, Belgium; (B.T.); (K.D.); (J.V.); (E.V.)
| | - Kaat Dekoster
- Department of Imaging and Pathology, KU Leuven, University of Leuven, 3000 Leuven, Belgium; (B.T.); (K.D.); (J.V.); (E.V.)
| | - Stijn E. Verleden
- Department of CHROMETA, BREATHE lab, KU Leuven, 3000 Leuven, Belgium; (S.E.V.); (A.V.)
| | - Stefan Sawall
- German Cancer Research Center (DKFZ), X-Ray Imaging and CT, Heidelberg University, 69117 Heidelberg, Germany; (S.S.); (M.K.)
| | - Bartosz Leszczyński
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 31-007 Kraków, Poland;
| | | | - Arno Vanstapel
- Department of CHROMETA, BREATHE lab, KU Leuven, 3000 Leuven, Belgium; (S.E.V.); (A.V.)
| | - Johny Verschakelen
- Department of Imaging and Pathology, KU Leuven, University of Leuven, 3000 Leuven, Belgium; (B.T.); (K.D.); (J.V.); (E.V.)
| | - Marc Kachelriess
- German Cancer Research Center (DKFZ), X-Ray Imaging and CT, Heidelberg University, 69117 Heidelberg, Germany; (S.S.); (M.K.)
| | - Erik Verbeken
- Department of Imaging and Pathology, KU Leuven, University of Leuven, 3000 Leuven, Belgium; (B.T.); (K.D.); (J.V.); (E.V.)
| | - Jim Swoger
- European Molecular Biology Laboratory (EMBL) Barcelona, 08003 Barcelona, Spain;
| | - Greetje Vande Velde
- Department of Imaging and Pathology, KU Leuven, University of Leuven, 3000 Leuven, Belgium; (B.T.); (K.D.); (J.V.); (E.V.)
| |
Collapse
|
43
|
Insights into the Role of Bioactive Food Ingredients and the Microbiome in Idiopathic Pulmonary Fibrosis. Int J Mol Sci 2020; 21:ijms21176051. [PMID: 32842664 PMCID: PMC7503951 DOI: 10.3390/ijms21176051] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 02/08/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic disease mainly associated with aging and, to date, its causes are still largely unknown. It has been shown that dietary habits can accelerate or delay the occurrence of aging-related diseases; however, their potential role in IPF development has been underestimated so far. The present review summarizes the evidence regarding the relationship between diet and IPF in humans, and in animal models of pulmonary fibrosis, in which we discuss the bioactivity of specific dietary food ingredients, including fatty acids, peptides, amino acids, carbohydrates, vitamins, minerals and phytochemicals. Interestingly, many animal studies reveal preventive and therapeutic effects of particular compounds. Furthermore, it has been recently suggested that the lung and gut microbiota could be involved in IPF, a relationship which may be linked to changes in immunological and inflammatory factors. Thus, all the evidence so far puts forward the idea that the gut-lung axis could be modulated by dietary factors, which in turn have an influence on IPF development. Overall, the data reviewed here support the notion of identifying food ingredients with potential benefits in IPF, with the ultimate aim of designing nutritional approaches as an adjuvant therapeutic strategy.
Collapse
|
44
|
De Sadeleer LJ, Goos T, Yserbyt J, Wuyts WA. Towards the Essence of Progressiveness: Bringing Progressive Fibrosing Interstitial Lung Disease (PF-ILD) to the Next Stage. J Clin Med 2020; 9:E1722. [PMID: 32503224 PMCID: PMC7355916 DOI: 10.3390/jcm9061722] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/19/2022] Open
Abstract
Although only recently introduced in the ILD community, the concept of progressive fibrosing interstitial lung disease (PF-ILD) has rapidly acquired an important place in the management of non-idiopathic pulmonary fibrosis fibrosing ILD (nonIPF fILD) patients. It confirms a clinical gut feeling that an important subgroup of nonIPF fILD portends a dismal prognosis despite therapeutically addressing the alleged triggering event. Due to several recently published landmark papers showing a treatment benefit with currently available antifibrotic drugs in PF-ILD patients, endorsing a PF-ILD phenotype has vital therapeutic consequences. Importantly, defining progressiveness is based on former progression, which has proven to be a rather moderate predictor of future progression. As fibrosis extent >20% and the presence of honeycombing have superior predictive properties regarding future progression, we advocate immediate initiation of antifibrotic treatment in the presence of these risk factors. In this perspective, we describe the historical context wherein PF-ILD has emerged, determine the currently employed PF-ILD criteria and their inherent limitations and propose new directions to mature its definition. Finally, while ascertaining progression in a nonIPF fILD patient clearly demonstrates the need for (additional) therapy, in the future, therapeutic decisions should be taken after assessing which pathway is ultimately driving the progression. Although not readily available, pathophysiological insight and diagnostic means are emergent to go full steam ahead in this novel direction.
Collapse
Affiliation(s)
- Laurens J. De Sadeleer
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, B-3000 Leuven, Belgium; (L.J.D.S.); (T.G.); (J.Y.)
- Unit of Interstitial Lung Diseases, Department of Respiratory Diseases, University Hospitals Leuven, B-3000 Leuven, Belgium
| | - Tinne Goos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, B-3000 Leuven, Belgium; (L.J.D.S.); (T.G.); (J.Y.)
- Unit of Interstitial Lung Diseases, Department of Respiratory Diseases, University Hospitals Leuven, B-3000 Leuven, Belgium
| | - Jonas Yserbyt
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, B-3000 Leuven, Belgium; (L.J.D.S.); (T.G.); (J.Y.)
| | - Wim A. Wuyts
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, B-3000 Leuven, Belgium; (L.J.D.S.); (T.G.); (J.Y.)
- Unit of Interstitial Lung Diseases, Department of Respiratory Diseases, University Hospitals Leuven, B-3000 Leuven, Belgium
| |
Collapse
|
45
|
Invernizzi R, Lloyd CM, Molyneaux PL. Respiratory microbiome and epithelial interactions shape immunity in the lungs. Immunology 2020; 160:171-182. [PMID: 32196653 PMCID: PMC7218407 DOI: 10.1111/imm.13195] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/17/2020] [Accepted: 02/17/2020] [Indexed: 12/11/2022] Open
Abstract
The airway epithelium represents a physical barrier to the external environment acting as the first line of defence against potentially harmful environmental stimuli including microbes and allergens. However, lung epithelial cells are increasingly recognized as active effectors of microbial defence, contributing to both innate and adaptive immune function in the lower respiratory tract. These cells express an ample repertoire of pattern recognition receptors with specificity for conserved microbial and host motifs. Modern molecular techniques have uncovered the complexity of the lower respiratory tract microbiome. The interaction between the microbiota and the airway epithelium is key to understanding how stable immune homeostasis is maintained. Loss of epithelial integrity following exposure to infection can result in the onset of inflammation in susceptible individuals and may culminate in lung disease. Here we discuss the current knowledge regarding the molecular and cellular mechanisms by which the pulmonary epithelium interacts with the lung microbiome in shaping immunity in the lung. Specifically, we focus on the interactions between the lung microbiome and the cells of the conducting airways in modulating immune cell regulation, and how defects in barrier structure and function may culminate in lung disease. Understanding these interactions is fundamental in the search for more effective therapies for respiratory diseases.
Collapse
Affiliation(s)
- Rachele Invernizzi
- Inflammation, Repair and Development SectionNational Heart and Lung InstituteImperial CollegeLondonUK
| | - Clare M. Lloyd
- Inflammation, Repair and Development SectionNational Heart and Lung InstituteImperial CollegeLondonUK
| | - Philip L. Molyneaux
- Inflammation, Repair and Development SectionNational Heart and Lung InstituteImperial CollegeLondonUK
- Department of Respiratory MedicineInterstitial Lung Disease UnitRoyal Brompton HospitalLondonUK
| |
Collapse
|
46
|
Dickson RP, Harari S, Kolb M. Making the case for causality: what role do lung microbiota play in idiopathic pulmonary fibrosis? Eur Respir J 2020; 55:55/4/2000318. [DOI: 10.1183/13993003.00318-2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 12/13/2022]
|