1
|
Higham A, Booth S, Dungwa J, Singh D. Histopathology of the small airways: Similarities and differences between ageing and COPD. Pulmonology 2025; 31:2430032. [PMID: 39883495 DOI: 10.1080/25310429.2024.2430032] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/19/2024] [Indexed: 01/31/2025] Open
Abstract
Age-related lung function decline is associated with small airway closure and gas trapping. The mechanisms which cause these changes are not fully understood. It has been suggested that COPD is caused by accelerated ageing. We have investigated pathological changes in the small airways during ageing, and evaluated whether the same or different processes exist in COPD. Histopathology and immunohistochemistry were used to examine small airway remodelling in healthy ageing, and then compare to age matched COPD patients. Ageing was associated with reduced alveolar attachment numbers (rho= -0.4 p = 0.049), increased epithelial area (rho = 0.5 p = 0.01), greater luminal narrowing due to epithelial expansion (rho = 0.5 p = 0.04) and increased alveolar septal neutrophils (rho = 0.6 p = 0.005). Compared to age matched controls, COPD small airways had 31% less alveolar attachments per airway (p = 0.02) and significantly more alveoalr septal neutrophils (p = 0.0007). Increased airway wall thickness was a feature of COPD but was not related to ageing in non-smokers. Alveolar attachment loss, accompanied by alveolar septum neutrophilic inflammation, and increased luminal narrowing due to epithelial expansion are major features of small airway remodelling during ageing. These features can explain the increased small airway narrowing and closure during ageing. Alveolar attachment loss is accelerated in COPD, likely due to increased neutrophilic inflammation.
Collapse
Affiliation(s)
- Andrew Higham
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester and Manchester University NHS Foundation Trust, Manchester, UK
| | - Sophie Booth
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester and Manchester University NHS Foundation Trust, Manchester, UK
- Medicines Evaluation Unit, Manchester, UK
| | | | - Dave Singh
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester and Manchester University NHS Foundation Trust, Manchester, UK
- Medicines Evaluation Unit, Manchester, UK
| |
Collapse
|
2
|
Cheng Z, Wu J, Xu C, Yan X. Mediating effects of gastroesophageal reflux disease and smoking behavior on the relationship between depression and chronic obstructive pulmonary disease: Trans-ethnic Mendelian randomization study. J Affect Disord 2025; 379:176-185. [PMID: 40074153 DOI: 10.1016/j.jad.2025.02.098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND This study seeks to elucidate the association between depression and the risk of chronic obstructive pulmonary disease (COPD) by Mendelian randomization (MR) analysis, motivated by prior observational studies indicating a potential link between these conditions. METHODS Data from individuals of European (EUR) and East Asian (EAS) ancestries diagnosed with major depressive disorder (MDD) were selected for analysis. The primary method utilized was inverse variance weighted (IVW) method, supplemented by a series of sensitivity analyses and false discovery rate (FDR) corrections. Subsequently, multivariable and mediation MR analyses were conducted to assess the impact of potential confounders and their mediating effects. RESULTS IVW revealed a significant causal relationship between MDD and COPD within EUR ancestry (OR 1.425, 95 % CI 1.243-1.633, P = 3.56 × 10-7, PFDR = 2.14 × 10-6). Additionally, replication datasets provided consistent evidence for these causal associations. Multivariable and mediation MR analyses identified gastroesophageal reflux disease (GORD) as a complete mediator (mediation effect: 98.97 %, P = 1.38 × 10-15), while smoking initiation (SI) (26.30 %, 5.54 × 10-9), age of smoking initiation (ASI) (18.73 %, 0.019), and cigarettes per day (CPD) (18.72 %, 0.004) were identified as partial mediators of this causal relationship. No causal association was detected in EAS ancestry, nor was reverse analysis. CONCLUSIONS This study established a causal relationship between MDD and COPD risk in EUR ancestry, identifying GORD and smoking as pivotal mediators. Future research involving larger cohorts is essential to validate the generalizability of these findings across other ancestries.
Collapse
Affiliation(s)
- Zewen Cheng
- Department of Thoracic Surgery, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou 215101, China
| | - Jian Wu
- Department of Thoracic Surgery, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou 215101, China
| | - Chun Xu
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Xiaokun Yan
- Department of Thoracic Surgery, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou 215101, China.
| |
Collapse
|
3
|
Labaki WW, Ram S, Namvar A, Bell AJ, Hoff BA, Kazerooni EA, Galban S, Martinez FJ, Hatt CR, Murray S, Mirkes EM, Gorban AN, Zinovyev A, Han MK, Galban CJ. Quantitative CT Scoring for Local COPD Severity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.09.25324951. [PMID: 40297437 PMCID: PMC12036413 DOI: 10.1101/2025.04.09.25324951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is complex, and its course is difficult to predict due to its diverse pathophysiology. Small airway disease (SAD), a key component of COPD and potential target for emerging therapeutics, may be reversible in mild COPD, but left unchecked, may worsen, leading to airway loss and emphysema. The dual nature of SAD complicates clinical management of COPD patients, necessitating more accurate monitoring methods. To meet this need, we developed elastic Parametric Response Mapping (ePRM), a tiered scoring system that classifies local lung volumes by the degree of PRM-derived SAD, normal, and emphysematous tissue. In individuals with or at risk for COPD, we demonstrate that chest CT ePRM can categorize local lung tissue into distinct tiers of disease severity that distinguish between tissue characterized by early reversible SAD and progressive destruction. This level of characterization is crucial to developing personalized treatment strategies for COPD.
Collapse
|
4
|
Brollo M, Marquant Q, Salvator H, Cohen J, Glorion M, Ferré A, Dres M, Roche N, Grassin‐Delyle S, Devillier P. Differences in the Inflammatory Response and Corticoid Responsiveness of Human Lung Macrophages and Parenchymal Explants Exposed to Cigarette Smoke Extracts. Basic Clin Pharmacol Toxicol 2025; 136:e70046. [PMID: 40344364 PMCID: PMC12061798 DOI: 10.1111/bcpt.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/18/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
Smoking is the main cause of chronic obstructive pulmonary disease (COPD) and is associated with corticosteroid resistance. Given the paucity of data on human lung preparations, macrophages (LMs), and parenchymal explants (LPEs) were exposed to cigarette smoke extracts (CSE) in the presence or absence of lipopolysaccharide (LPS). Moreover, LMs and LPEs were treated with budesonide prior exposure to CSE or LPS. The levels of cytokines (TNF-α, IL-6) and chemokines (CCL2, CCL4, CXCL1, CXCL5, and CXCL8) in the supernatants were measured using ELISAs. In LMs, exposure to CSE was not associated with significant difference in the production of cytokines and chemokines, with the notable exception of greater CXCL8 production. The results were generally the same for LPEs. CSE exposure did not potentiate the LPS-induced production of the cytokines and chemokines and even tended to reduce this production in LMs and LPEs. Lastly, CSE exposure inhibited budesonide's anti-inflammatory activity in LMs but not in LPEs. This study extends the data on the CSE inflammatory effects and its inhibition of corticosteroid efficacy in human lung preparations. Our findings question the relevance of these preparations with regard to the long-term toxicity of smoking and the corticosteroid resistance observed in smokers and in patients with COPD.
Collapse
Affiliation(s)
- Marion Brollo
- Laboratory of Research in Respiratory Pharmacology, Molecular Immunology & Virology (VIM Suresnes), INRAE UMR‐0892Paris‐Saclay University, Foch HospitalSuresnesFrance
| | - Quentin Marquant
- Laboratory of Research in Respiratory Pharmacology, Molecular Immunology & Virology (VIM Suresnes), INRAE UMR‐0892Paris‐Saclay University, Foch HospitalSuresnesFrance
| | - Hélène Salvator
- Laboratory of Research in Respiratory Pharmacology, Molecular Immunology & Virology (VIM Suresnes), INRAE UMR‐0892Paris‐Saclay University, Foch HospitalSuresnesFrance
- Pneumology DepartmentFoch HospitalSuresnesFrance
- Faculty of Health Sciences Simone VeilVersailles Saint Quentin UniversityMontigny le BretonneuxFrance
| | | | - Matthieu Glorion
- Laboratory of Research in Respiratory Pharmacology, Molecular Immunology & Virology (VIM Suresnes), INRAE UMR‐0892Paris‐Saclay University, Foch HospitalSuresnesFrance
- Thoracic Surgery DepartmentFoch HospitalSuresnesFrance
| | - Alexis Ferré
- Intensive Care UnitVersailles HospitalLe ChesnayFrance
| | - Martin Dres
- INSERM, UMRS_1158 Neurophysiologie Respiratoire Expérimentale et CliniqueSorbonne UniversitéParisFrance
- Department of Medical Intensive CareAPHP, Groupe Hospitalier Pitié SalpêtrièreParisFrance
| | - Nicolas Roche
- Pneumology DepartmentCochin Hospital, AP‐HP, Paris Cité University, UMR 1016, Institut CochinParisFrance
| | - Stanislas Grassin‐Delyle
- Laboratory of Research in Respiratory Pharmacology, Molecular Immunology & Virology (VIM Suresnes), INRAE UMR‐0892Paris‐Saclay University, Foch HospitalSuresnesFrance
- Faculty of Health Sciences Simone VeilVersailles Saint Quentin UniversityMontigny le BretonneuxFrance
- Infection & Inflammation, Health Biotechnology Department, INSERM U1173, Faculty of Health Sciences Simone VeilVersailles Saint Quentin University, Paris‐Saclay UniversityMontigny‐le‐BretonneuxFrance
| | - Philippe Devillier
- Laboratory of Research in Respiratory Pharmacology, Molecular Immunology & Virology (VIM Suresnes), INRAE UMR‐0892Paris‐Saclay University, Foch HospitalSuresnesFrance
- Faculty of Health Sciences Simone VeilVersailles Saint Quentin UniversityMontigny le BretonneuxFrance
| |
Collapse
|
5
|
Chen Y, Latisenko R, Lynch DA, Ciet P, Charbonnier JP, Tiddens HAWM. Effect of inspiratory lung volume on bronchial and arterial dimensions and ratios on chest computed tomography in patients with chronic obstructive pulmonary disease. Eur Radiol 2025; 35:2990-2998. [PMID: 39613958 PMCID: PMC12081482 DOI: 10.1007/s00330-024-11126-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/26/2024] [Accepted: 09/13/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND The assessment of bronchus-artery (BA) metrics on chest CT is important for detecting airway abnormalities. It is less clear how BA metrics are dependent on lung volume. METHODS CTs were obtained from a COPDGene substudy investigating the impact of radiation dose on lung density. Patients with chronic obstructive pulmonary disease underwent a full-dose and a reduced-dose CT in the same imaging session. CTs were automatically analyzed by measuring diameters of the bronchial outer edge (Bout), bronchial inner wall (Bin), artery (A), and bronchial wall thickness (Bwt) from segmental (G0) and distal generations. BA ratios were computed: Bout/A, Bin/A, Bwt/A, and bronchial wall area/bronchial outer area (Bwa/Boa). The total lung volume of the CT (TLC-CT) was computed. Differences between the volumes between the two CTs were expressed as % of the highest TLC-CT (ΔTLC-CT%). For the BA metrics of each CT, we computed the median of measurements in G1-6. Mixed-effect models were used to investigate the influence of TLC-CT on BA metrics adjusted for dose protocol. RESULTS One thousand three hundred nineteen patients with a mean (SD) age of 64.4 (8.7) years were included. Three hundred twenty-nine (124) BA pairs were analyzed per CT. No significant difference was found for TLC-CT in relation to dose (p = 0.17). A ΔTLC-CT% of >10% (found in 121, 9%) led to 0.03 and 0.05 decreases in Bout/A and Bin/A and 0.008 and 0.11 decrease in log (Bwt/A) and log (Bwa/Boa), and a 0.03 increase in Bin and 0.06, 0.12, and 0.04 decrease in Bout, log (Bwt), and log (A) (all p < 0.001). CONCLUSIONS Variations in TLC over 10% between time points significantly influence bronchial dimensions, affecting BA metrics. Standardizing volumes is recommended for sensitive tracking of airway disease changes over time. KEY POINTS Question Are BA metrics dependent on total lung capacity (TLC), and if so, how? Findings TLC variations over 10% between time points significantly influence bronchial dimensions, affecting BA metrics. Variations below 10% between CT scans have little effect on BA metrics. Clinical relevance Small lung volume differences between chest CTs have little impact on bronchus and artery metrics; it is imperative to standardize chest CT lung volumes to ensure precise diagnosis and monitoring of airway disease.
Collapse
Affiliation(s)
- Yuxin Chen
- Division of Respiratory Medicine and Allergology, Department of Paediatrics, Sophia Children's Hospital, Erasmus MC, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | - David A Lynch
- Department of Radiology, National Jewish Health, Denver, CO, USA
| | - Pierluigi Ciet
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Radiology and Medical Science, University of Cagliari, Cagliari, Italy
| | | | - Harm A W M Tiddens
- Division of Respiratory Medicine and Allergology, Department of Paediatrics, Sophia Children's Hospital, Erasmus MC, Rotterdam, The Netherlands.
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
- Thirona, Nijmegen, The Netherlands.
| |
Collapse
|
6
|
Toumpanakis D, Bartziokas K, Bakakos A, Fouka E, Bakakos P, Loukides S, Steiropoulos P, Papaioannou AI. Monoclonal Antibodies for the Treatment of Chronic Obstructive Pulmonary Disease. Pulm Ther 2025; 11:177-193. [PMID: 40123030 PMCID: PMC12102449 DOI: 10.1007/s41030-025-00291-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common and complex disease characterized by persistent airflow limitation and the presence of exacerbations, resulting in significant morbidity and mortality. Although the pathogenesis of COPD is multifactorial, airway inflammation plays a significant role in disease progression. Despite the advantages of non-pharmaceutical and pharmaceutical interventions that have significantly improved the symptom burden and exacerbation frequency in COPD, there is a lack of disease-modifying therapies that target the underlying disease mechanisms. Monoclonal antibodies (mAbs), a drug class that has improved treatment in severe asthma by blocking mediators of the type 2 (Th2) and allergic inflammatory cascades, are currently under investigation for their efficacy in COPD. Our review summarizes the evidence for the use of monoclonal antibodies in COPD and discusses current limitations and promising advances. Although targeting Th1 inflammation has failed to improve COPD outcomes, recent clinical trials have shown beneficial effects of monoclonal antibodies targeting Th2 inflammation, providing evidence for a personalized approach in COPD treatment.
Collapse
Affiliation(s)
- Dimitrios Toumpanakis
- 2Nd Department of Critical Care, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Konstantinos Bartziokas
- 2Nd Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Agamemnon Bakakos
- 1St Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Diseases Hospital, Athens, Greece
| | - Evangelia Fouka
- Respiratory Medicine Department, Aristotle University of Thessaloniki, G Papanikolaou Hospital, Thessaloniki, Greece
| | - Petros Bakakos
- 1St Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Diseases Hospital, Athens, Greece
| | - Stelios Loukides
- 2Nd Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Paschalis Steiropoulos
- Department of Pneumonology, Medical School, Democritus University of Thrace, 68100, Alexandroupolis, Greece.
| | - Andriana I Papaioannou
- 1St Respiratory Medicine Department, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Diseases Hospital, Athens, Greece
| |
Collapse
|
7
|
Sun S, Zhang Z, Zhao H. Identification and validation of USP15 and CUL2 as ubiquitination related biomarker in chronic obstructive pulmonary disease. Hereditas 2025; 162:86. [PMID: 40413536 PMCID: PMC12103031 DOI: 10.1186/s41065-025-00460-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025] Open
Abstract
PURPOSE Ubiquitination is one of the important epigenetic modifications, influencing the development of various diseases. The objective of this study is to investigate the ubiquitination related genes in chronic obstructive pulmonary disease (COPD). METHODS The gene microarray dataset from COPD patients and ubiquitination related genes were analyzed. Venn diagram analysis was used to intersect differentially expressed genes and ubiquitination related genes. The functional enrichment analysis of Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Enrichment Analysis (GSEA) were performed on differentially expressed ubiquitination related genes. Finally, we confirmed the expression of hub genes through qPCR and western blot experiments in clinical COPD patients and cell lines. RESULTS We identified 2,932 differentially expressed genes and 96 differentially expressed ubiquitination related genes. GO analysis indicated that the differentially expressed ubiquitination related genes were mainly enriched in post-translational protein modification and ubiquitin ligase complex. KEGG analysis showed that ubiquitination related genes were mainly involved in ubiquitin mediated proteolysis and TNF signaling pathway. GSEA analysis suggested that some hub genes are involved in allograft rejection, IL6/JAK/STAT3 signaling and inflammatory response. Our qPCR and western blot experimental results indicate that the expression of USP15 and CUL2 is higher in COPD group compared to the control group, consistent with the bioinformatics analysis. CONCLUSION Our bioinformatics analysis and experimental results suggest that USP15 and CUL2 may contribute to the progression of COPD through ubiquitination modification. To our knowledge, this is the first study to demonstrate the involvement of USP15 and CUL2 in COPD. Our results may provide new insights into the diagnosis and treatment of COPD.
Collapse
Affiliation(s)
- Shulei Sun
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Zhaoxiong Zhang
- Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Haiyan Zhao
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
8
|
Zhang Q, Zhou L, Yuan L, Zhang R, Pan S, Wang X, Yi L, Yuan F, Guo X, Gu M, Wang Y, Jia X. scRNA-seq and scATAC-seq analyses highlight the role of TNF signaling pathway in chronic obstructive pulmonary disease model mice. PLoS One 2025; 20:e0322538. [PMID: 40343927 PMCID: PMC12063857 DOI: 10.1371/journal.pone.0322538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 03/24/2025] [Indexed: 05/11/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent and progressive form of respiratory disease in which patients exhibit persistent respiratory damage affecting the alveoli and/or airway due to exposure to toxic gases or particulate matter. C57BL/6 mice were exposed to cigarette smoke and lipopolysaccharide to establish a COPD model mice, followed by scATAC (Assay for Transposase Accessible Chromatin) sequencing and scRNA sequencing of lung tissue samples. The resultant data revealed consistent findings between scATAC-seq and scRNA-seq regarding cell types, differentially expressed genes, and signaling pathways in COPD model mice. Tumor necrosis factor (TNF) signaling pathway enrichment was evident in the scRNA-seq and scATAC-seq datasets, with similar trends in monocytes/macrophages, dendritic cells, and B cells. In COPD model mice, significant tumor necrosis factor receptor 1 (TNFR1) upregulation and high levels of activity related to cellular communication were observed, and significant increases in Il1b, Csf1, and Bcl3 site accessibility were evident in cells. These findings suggest that the TNF signaling pathway maybe associated with COPD.
Collapse
MESH Headings
- Animals
- Pulmonary Disease, Chronic Obstructive/genetics
- Pulmonary Disease, Chronic Obstructive/metabolism
- Pulmonary Disease, Chronic Obstructive/pathology
- Signal Transduction
- Mice
- Disease Models, Animal
- Mice, Inbred C57BL
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Male
- Lung/metabolism
- Lung/pathology
- Lipopolysaccharides
- RNA-Seq
- Sequence Analysis, RNA
- Single-Cell Gene Expression Analysis
Collapse
Affiliation(s)
- Qiang Zhang
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Li Zhou
- BGI-Qingdao, BGI-Shenzhen, Qingdao, China
- Beijing Easyresearch Technology Limited, Beijing, China
| | - Lindong Yuan
- Department of Respiratory and Critical Care Medicine, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Ruihua Zhang
- BGI-Qingdao, BGI-Shenzhen, Qingdao, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | | | - Xizi Wang
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Lili Yi
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Fengjiao Yuan
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Xianchao Guo
- Harbin Inji Technology Co., Ltd., Harbin, Heilongjiang, China
- GenVista Technology Co., Ltd, Harbin, Heilongjiang, China
| | - Mingliang Gu
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Xiaodong Jia
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, P.R.China
| |
Collapse
|
9
|
Vilches LF, Dickinson JD. Start SPREADing the News: Biosensors Detect Ripples of Extracellular Signal-regulated Kinase Signaling in Airway Epithelial Cells. Am J Respir Cell Mol Biol 2025; 72:469-471. [PMID: 39586020 PMCID: PMC12051931 DOI: 10.1165/rcmb.2024-0547ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024] Open
Affiliation(s)
- Luis F Vilches
- Department of Internal Medicine University of Nebraska Medical Center Omaha, Nebraska
| | - John D Dickinson
- Department of Internal Medicine University of Nebraska Medical Center Omaha, Nebraska
| |
Collapse
|
10
|
Schedel M, Heimel V, Taube C. Type 2 inflammation, a common denominator in chronic airway disease? Curr Opin Pulm Med 2025; 31:302-309. [PMID: 40104899 DOI: 10.1097/mcp.0000000000001159] [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: 03/20/2025]
Abstract
PURPOSE OF REVIEW This review addresses the growing understanding that a specific subset of patients with a respiratory disease, including asthma, chronic obstructive pulmonary disease (COPD), or bronchiectasis may have one thing in common: type 2 inflammation. In the era of personalized medicine, we need to refine clinical markers combined with molecular and cellular endotyping to improve patient outcomes. RECENT FINDINGS Recent literature reveals that type 2 markers such as blood eosinophils, fractional exhaled nitric oxide (FeNO), and immunglobulin E (IgE), can provide valuable insights into disease progression, exacerbation risk, and treatment response, but their stability remains to be investigated. Treating asthma and COPD patients with biologics to target IL-4/IL-13, IL-5, and alarmins have shown potential, although efficacy varied. In bronchiectasis, a subset of patients with type 2 inflammation may benefit from corticosteroid therapy, despite broader concerns regarding its use. SUMMARY This underscores the importance of improved disease endotyping to better characterize patients who may benefit from targeted therapies. In clinical practice, personalized treatment based on inflammatory profiles has been shown to improve outcomes in heterogeneous lung diseases. Future research needs to focus on validating reliable biomarkers and optimizing clinical trial designs to advance therapeutic strategies in respiratory diseases.
Collapse
Affiliation(s)
- Michaela Schedel
- Translational Pulmonology, Department of Pulmonary Medicine, University Medical Center Essen - Ruhrlandklinik
- Translational Pulmonology, Department of Pulmonary Medicine, University Medical Center
| | - Victoria Heimel
- Translational Pulmonology, Department of Pulmonary Medicine, University Medical Center Essen - Ruhrlandklinik
| | - Christian Taube
- Department of Pulmonary Medicine, University Medical Center Essen - Ruhrlandklinik, Essen, Germany
| |
Collapse
|
11
|
Kocot N, Pękala E, Koczurkiewicz-Adamczyk P, Chłoń-Rzepa G, Łapa A, Wójcik-Pszczoła K. Airway and cardiovascular remodeling in chronic obstructive pulmonary disease (COPD) as a target for transient receptor potential ankyrin 1 (TRPA1) channel modulators. Bioorg Chem 2025; 158:108301. [PMID: 40058223 DOI: 10.1016/j.bioorg.2025.108301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/06/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by chronic inflammation, which leads to airway remodeling (AR). AR refers to various structural changes occurring in the airway wall, resulting in thickening, and narrowing of the airways. Apart from airways, and lung tissue, pulmonary vasculature also undergoes remodeling. Thus, the pressure in vascular bed is increased, leading to pulmonary hypertension and further right and left ventricle hypertrophy, as well as myocardial fibrosis. Currently, there is lack of effective treatment directly targeting airway and cardiovascular remodeling in the course of COPD. Due to a lot of research showing involvement of transient receptor potential ankyrin 1 (TRPA1) in respiratory disorders, it seems reasonable to consider this ion channel as a molecular target in treatment of remodeling consequences of COPD. The aim of this review is to summarize current knowledge of its role in this case and to identify areas requiring further research. Moreover, we provide few patented structures intended to treat chronic respiratory diseases, which may be worth investigating in the context of airway remodeling.
Collapse
Affiliation(s)
- Natalia Kocot
- Jagiellonian University, Doctoral School of Medical and Health Sciences, Łazarza 16, 31-530 Kraków, Poland; Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmaceutical Biochemistry, Medyczna 9, 30-688 Kraków, Poland.
| | - Elżbieta Pękala
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmaceutical Biochemistry, Medyczna 9, 30-688 Kraków, Poland.
| | - Paulina Koczurkiewicz-Adamczyk
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmaceutical Biochemistry, Medyczna 9, 30-688 Kraków, Poland.
| | - Grażyna Chłoń-Rzepa
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, Medyczna 9, 30-688 Kraków, Poland.
| | - Aleksandra Łapa
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmaceutical Biochemistry, Medyczna 9, 30-688 Kraków, Poland.
| | - Katarzyna Wójcik-Pszczoła
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmaceutical Biochemistry, Medyczna 9, 30-688 Kraków, Poland.
| |
Collapse
|
12
|
Kelly MP, Nikolaev VO, Gobejishvili L, Lugnier C, Hesslinger C, Nickolaus P, Kass DA, Pereira de Vasconcelos W, Fischmeister R, Brocke S, Epstein PM, Piazza GA, Keeton AB, Zhou G, Abdel-Halim M, Abadi AH, Baillie GS, Giembycz MA, Bolger G, Snyder G, Tasken K, Saidu NEB, Schmidt M, Zaccolo M, Schermuly RT, Ke H, Cote RH, Mohammadi Jouabadi S, Roks AJM. Cyclic nucleotide phosphodiesterases as drug targets. Pharmacol Rev 2025; 77:100042. [PMID: 40081105 DOI: 10.1016/j.pharmr.2025.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/13/2025] [Indexed: 03/15/2025] Open
Abstract
Cyclic nucleotides are synthesized by adenylyl and/or guanylyl cyclase, and downstream of this synthesis, the cyclic nucleotide phosphodiesterase families (PDEs) specifically hydrolyze cyclic nucleotides. PDEs control cyclic adenosine-3',5'monophosphate (cAMP) and cyclic guanosine-3',5'-monophosphate (cGMP) intracellular levels by mediating their quick return to the basal steady state levels. This often takes place in subcellular nanodomains. Thus, PDEs govern short-term protein phosphorylation, long-term protein expression, and even epigenetic mechanisms by modulating cyclic nucleotide levels. Consequently, their involvement in both health and disease is extensively investigated. PDE inhibition has emerged as a promising clinical intervention method, with ongoing developments aiming to enhance its efficacy and applicability. In this comprehensive review, we extensively look into the intricate landscape of PDEs biochemistry, exploring their diverse roles in various tissues. Furthermore, we outline the underlying mechanisms of PDEs in different pathophysiological conditions. Additionally, we review the application of PDE inhibition in related diseases, shedding light on current advancements and future prospects for clinical intervention. SIGNIFICANCE STATEMENT: Regulating PDEs is a critical checkpoint for numerous (patho)physiological conditions. However, despite the development of several PDE inhibitors aimed at controlling overactivated PDEs, their applicability in clinical settings poses challenges. In this context, our focus is on pharmacodynamics and the structure activity of PDEs, aiming to illustrate how selectivity and efficacy can be optimized. Additionally, this review points to current preclinical and clinical evidence that depicts various optimization efforts and indications.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Neurobiology, Center for Research on Aging, University of Maryland School of Medicine, Baltimore, Maryland
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leila Gobejishvili
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, Louisville
| | - Claire Lugnier
- Translational CardioVascular Medicine, CRBS, UR 3074, Strasbourg, France
| | | | - Peter Nickolaus
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Rodolphe Fischmeister
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Orsay, France
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, Connecticut
| | - Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, Connecticut
| | - Gary A Piazza
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Adam B Keeton
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - George S Baillie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark A Giembycz
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Gretchen Snyder
- Molecular Neuropharmacology, Intra-Cellular Therapies Inc (ITI), New York, New York
| | - Kjetil Tasken
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nathaniel E B Saidu
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics and National Institute for Health and Care Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Ralph T Schermuly
- Department of internal Medicine, Justus Liebig University of Giessen, Giessen, Germany
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina
| | - Rick H Cote
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire
| | - Soroush Mohammadi Jouabadi
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Anton J M Roks
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
13
|
Zhou Y, Wu F, Deng Z, Wang Z, Tian H, Huang P, Zheng Y, Yang H, Zhao N, Dai C, Yang C, Yu S, Tian J, Cui J, Liu S, Wang D, Wang X, Lu J, Zhong N, Ran P. Lung function decline and incidence of chronic obstructive pulmonary disease in participants with spirometry-defined small airway dysfunction: a 15-year prospective cohort study in China. Respir Res 2025; 26:169. [PMID: 40296032 PMCID: PMC12039187 DOI: 10.1186/s12931-025-03244-3] [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: 04/12/2024] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Small airway dysfunction (SAD) is common but little is known about the longitudinal prognosis of spirometry-defined SAD. Therefore, we aimed to evaluate the risk of lung function decline and incident chronic obstructive pulmonary disease (COPD) of spirometry-defined SAD. METHODS It was a population-based prospective cohort study conducted in Guangdong, China. Participants were enrolled in the years 2002, 2008, 2012, 2017, and 2019, and those who completed baseline demographic data, a standardized epidemiological questionnaire for COPD, and spirometry were included. Follow-up visits were conducted every three years after enrolment, with a maximum follow-up time of 15 years and a minimum follow-up time of 3 years. Spirometry-defined SAD was defined as having at least two out of three parameters (maximal mid-expiratory flow, forced expiratory flow 50%, and forced expiratory flow 75%) below 65% of the predicted value. Non-obstructive SAD and obstructive SAD were further differentiated based on the presence of airflow obstruction (forced expiratory volume in one second [FEV1]/forced vital capacity [FVC] < 0.70). Pre- and post-bronchodilator spirometry measurements were analyzed separately. RESULTS Pre-bronchodilator spirometry dataset included 4680 participants (mean age 55.3 [10.8] years, 2194 [46.9%] males). Participants with pre-bronchodilator SAD had a significantly faster annual decline of FEV1 % of predicted value (0.31 ± 0.05 vs. 0.20 ± 0.03 %/year; difference: 0.12 [95% confidence interval: 0.01-0.23]; P = 0.023), FVC, and FVC % of predicted value compared to those without pre-bronchodilator SAD. The annual decline of lung function in participants with pre-bronchodilator non-obstructive SAD was not significantly different from that in pre-bronchodilator healthy controls, but they were more likely to progress to spirometry-defined COPD (adjusted hazard ratio: 2.92 [95% confidence interval: 2.28-3.76], P < 0.001). Post-bronchodilator spirometry dataset yielded similar results. CONCLUSIONS Individuals with spirometry-defined SAD have a faster decline in lung function compared to those without SAD, and non-obstructive SAD is more likely to progress to spirometry-defined COPD. TRIAL REGISTRATION Chinese Clinical Trials Registration ChiCTR1900024643. Registered on 19 July 2019.
Collapse
Affiliation(s)
- Yumin Zhou
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, 510120, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Fan Wu
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, 510120, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Zhishan Deng
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, 510120, China
| | - Zihui Wang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, 510120, China
| | - Heshen Tian
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, 510120, China
| | - Peiyu Huang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, 510120, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Youlan Zheng
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, 510120, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Huajing Yang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, 510120, China
| | - Ningning Zhao
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, 510120, China
| | - Cuiqiong Dai
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, 510120, China
| | - Changli Yang
- Department of Pulmonary and Critical Care Medicine, Wengyuan County People's Hospital, Shaoguan, China
| | - Shuqing Yu
- Lianping County People's Hospital, Lianping County Hospital of Traditional Chinese Medicine, Heyuan, China
| | - Jia Tian
- Department of Pulmonary and Critical Care Medicine, The Second People's Hospital of Hunan Province, Changsha, China
| | - Jiangyu Cui
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, 510120, China
| | - Shengming Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dali Wang
- The Second Hospital of Liwan District of Guangzhou, Guangzhou, China
| | - Xiaoping Wang
- The First Municipal People Hospital of Shaoguan, Shaoguan, China
| | - Jiachun Lu
- State Key Laboratory of Respiratory Disease, Institute of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, 510120, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Pixin Ran
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 195 Dongfeng Xi Road, Guangzhou, 510120, China.
- Guangzhou National Laboratory, Guangzhou, China.
| |
Collapse
|
14
|
Kunchur NN, Poole JJA, Levine J, Hackett TL, Thornhill R, Mostaço-Guidolin LB. Classification of collagen remodeling in asthma using second-harmonic generation imaging, supervised machine learning and texture-based analysis. FRONTIERS IN BIOINFORMATICS 2025; 5:1539936. [PMID: 40313867 PMCID: PMC12043662 DOI: 10.3389/fbinf.2025.1539936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/19/2025] [Indexed: 05/03/2025] Open
Abstract
Airway remodeling is present in all stages of asthma severity and has been linked to reduced lung function, airway hyperresponsiveness and increased deposition of fibrillar collagens. Traditional histological staining methods used to visualize the fibrotic response are poorly suited to capture the morphological traits of extracellular matrix (ECM) proteins in their native state, hindering our understanding of disease pathology. Conversely, second harmonic generation (SHG), provides label-free, high-resolution visualization of fibrillar collagen; a primary ECM protein contributing to the loss of asthmatic lung elasticity. From a cohort of 13 human lung donors, SHG-imaged collagen belonging to non-asthmatic (control) and asthmatic donors was evaluated through a custom textural classification pipeline. Integrated with supervised machine learning, the pipeline enables the precise quantification and characterization of collagen, delineating amongst control and remodeled airways. Collagen distribution is quantified and characterized using 80 textural features belonging to the Gray Level Cooccurrence Matrix (GLCM), Gray Level Size Zone Matrix (GLSZM), Gray Level Run Length Matrix (GLRLM), Gray Level Dependence Matrix (GLDM) and Neighboring Gray Tone Difference Matrix (NGTDM). To denote an accurate subset of features reflective of fibrillar collagen formation; filter, wrapper, embedded and novel statistical methods were applied as feature refinement. Textural feature subsets of high predictor importance trained a support vector machine model, achieving an AUC-ROC of 94% ± 0.0001 in the classification of remodeled airway collagen vs. control lung tissue. Combined with detailed texture analysis and supervised ML, we demonstrate that morphological variation amongst remodeled SHG-imaged collagen in lung tissue can be successfully characterized.
Collapse
Affiliation(s)
- Natasha N. Kunchur
- Department of Systems and Computer Engineering at Carleton University, Ottawa, ON, Canada
| | - Joshua J. A. Poole
- Department of Systems and Computer Engineering at Carleton University, Ottawa, ON, Canada
| | - Jesse Levine
- Department of Systems and Computer Engineering at Carleton University, Ottawa, ON, Canada
| | - Tillie-Louise Hackett
- Anesthesiology, Pharmacology and Therapeutics Department at the University of British Columbia, Medical Sciences, Vancouver, BC, Canada
| | - Rebecca Thornhill
- Department of Radiology, Radiation Oncology, and Medical Physics at the University of Ottawa, Ottawa, ON, Canada
- Department of Medical Imaging at the Ottawa Hospital, Ottawa, ON, Canada
| | | |
Collapse
|
15
|
Šileikienė V, Jurgauskienė L. Role of Regulatory T Cells in Pulmonary Ageing and COPD Development. Int J Mol Sci 2025; 26:3721. [PMID: 40332320 PMCID: PMC12027511 DOI: 10.3390/ijms26083721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is recognized as a long-term inflammatory lung condition, predominantly resulting from smoking tobacco. While all smokers exhibit some level of pulmonary inflammation, only about 15-20% go on to develop significant COPD, indicating that specific individual factors may enhance these inflammatory responses and contribute to the disease's progression. T regulatory cell (Treg) activity is crucial in mediating pulmonary inflammation in COPD. With accumulating evidence supporting the autoimmune characteristics of COPD, there has been an increasing focus on the role Treg cells play in the disease's initiation and development. This article aims to review the existing literature regarding Treg cells and their influence on COPD pathogenesis and lung ageing. Treg-mediated suppression is a critical mechanism in the negative regulation of immune-related inflammation, which is significant in various disorders, including autoimmunity, allergies, infections (both acute and chronic), and cancer. The lungs of ageing individuals often resemble those affected by COPD, leading to the perception of COPD as a condition that accelerates lung ageing. Changes in Treg cells with age correspond to decreased adaptive immune responses and a higher likelihood of immune-related disorders. The increased presence of Treg cells in older adults may help explain several immunological conditions commonly associated with ageing, which include malignancies, infections, and COPD.
Collapse
Affiliation(s)
- Virginija Šileikienė
- Clinic of Chest Diseases, Immunology and Allergology, Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
| | - Laimutė Jurgauskienė
- Clinic of Cardiac and Vascular Diseases, Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania;
| |
Collapse
|
16
|
Makimoto K, Singh GV, Kirby M. Advances in detecting small airway disease with medical imaging. Eur Respir J 2025; 65:2500212. [PMID: 40154561 DOI: 10.1183/13993003.00212-2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/13/2025] [Indexed: 04/01/2025]
Affiliation(s)
| | | | - Miranda Kirby
- Toronto Metropolitan University, Toronto, ON, Canada
| |
Collapse
|
17
|
Mohamady YK, Geudens V, De Fays C, Zapata M, Hagrass O, Aversa L, Vermant M, Jin X, Willems L, Gyselinck I, Hooft C, Vermaut A, Beeckmans H, Kerckhof P, Aerts G, Aelbrecht C, Verhaegen J, Higham A, Coudyzer W, Cortesi EE, Vanstapel A, McDonough JE, Carlon MS, Quarck R, Boone MN, Dupont L, Everaerts S, Van Raemdonck DE, Ceulemans LJ, Hackett TL, Vos R, Abuouf Y, Jacob J, Wuyts WA, Hogg JC, Filoche M, Gayan-Ramirez G, Janssens W, Vanaudenaerde BM. Computational fluid dynamics of small airway disease in chronic obstructive pulmonary disease. EBioMedicine 2025; 114:105670. [PMID: 40174553 PMCID: PMC11999283 DOI: 10.1016/j.ebiom.2025.105670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Small airways (<2 mm diameter) are major sites of airflow obstruction in chronic obstructive pulmonary disease (COPD). This study aimed to quantify the impact of small airway disease, characterized by narrowing, occlusion, and obliteration, on airflow parameters in smokers and end-stage patients with COPDs. METHODS We performed computational fluid dynamics (CFD) simulations of inspiratory airflow in three lung groups: control non-used donor lungs (no smoking/emphysema history), non-used donor lungs with a smoking history and emphysema, and explanted end-stage COPD lungs. Each group included four lungs, with two tissue cylinders. Micro-CT-scanned small airways were segmented into 3D models for CFD simulations to quantify pressure, resistance, and shear stress. CFD results were benchmarked against simplified linear and Weibel models. FINDINGS CFD simulations showed higher pressures in COPD vs. controls (p = 0.0091) and smokers (p = 0.015), along with increased resistance (p = 0.0057 vs. controls; p = 0.0083 vs. smokers) and up to a tenfold rise in shear stress (p = 0.010 vs. controls). Narrowing and occlusion were shown to independently increase pressure, resistance, and shear stress, which were validated through segmentation corrections. Pressures and resistance assessed with simplified models were up to seven-fold higher for smokers and even 72 higher for COPD compared with CFD values. INTERPRETATION These findings show that increased airflow parameters can explain the association between small airway disease and airflow limitation in COPD, underscoring small airway vulnerability. Additionally, they highlight the limitations of theoretical models in accurately capturing small airway disease. FUNDING Supported by the KU Leuven (C16/19/005).
Collapse
Affiliation(s)
- Yousry K Mohamady
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Vincent Geudens
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Charlotte De Fays
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium; Pneumology Lab, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium.
| | - Marta Zapata
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Omar Hagrass
- Department of Operations Research and Financial Engineering, Princeton University, USA.
| | - Lucia Aversa
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Marie Vermant
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Xin Jin
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Lynn Willems
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Iwein Gyselinck
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Charlotte Hooft
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Astrid Vermaut
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Hanne Beeckmans
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Pieterjan Kerckhof
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Gitte Aerts
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Celine Aelbrecht
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Janne Verhaegen
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Andrew Higham
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Manchester University, UK.
| | | | - Emanuela E Cortesi
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Arno Vanstapel
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - John E McDonough
- Firestone Institute for Respiratory Health, St Joseph's Healthcare Hamilton, McMaster University, Hamilton, Canada.
| | - Marianne S Carlon
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Rozenn Quarck
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Matthieu N Boone
- Dept of Physics and Astronomy, UGCT, Radiation Physics, Ghent University, Gent, Belgium.
| | - Lieven Dupont
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Stephanie Everaerts
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Dirk E Van Raemdonck
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Laurens J Ceulemans
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium; Translational Cell and Tissue Research, KU Leuven and UZ Leuven, Leuven, Belgium.
| | - Tillie-Louise Hackett
- Center for Heart Lung Innovation, The University of British Columbia, Vancouver, Canada.
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Yasser Abuouf
- Department of Mechanical Engineer, Alexandria University, Egypt.
| | - Joseph Jacob
- Hawkes Institute, Department of Computer Science, UCL, London, UK; UCL Respiratory, UCL, London, UK.
| | - Wim A Wuyts
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - James C Hogg
- Center for Heart Lung Innovation, The University of British Columbia, Vancouver, Canada.
| | - Marcel Filoche
- Physique de la Matière Condensée, Ecole Polytechnique, Palaiseau, France.
| | - Ghislaine Gayan-Ramirez
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Wim Janssens
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Bart M Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery, BREATHE, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| |
Collapse
|
18
|
Shao W, Guo H, Sun J, Liu Z, Chang Z, Hao Y, Li H, Li B, Chen F, Liu J. Effects of different degrees of anthracosis on pleural structure: A preliminary exploratory study. Respir Med 2025; 240:108043. [PMID: 40090525 DOI: 10.1016/j.rmed.2025.108043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/22/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND Pleural anthracosis (PA) appears as superficial black discoloration or scattered focal black spots on the surface of the pleura. Few studies have investigated the relationship between PA and changes in pleural structure. The aim of this study is to perform a detailed morphometric characterization of pleura in patients with different degrees of PA at both macroscopic and microscopic levels. METHODS Preoperative, intraoperative, and postoperative data were collected for all patients. Lung tissue (far away from the tumor) was obtained from patients with pulmonary nodules who required surgical treatment. According to different degrees of PA, the pleura were classified into four degrees. In addition to using grade data, we also calculated the ratio of PA. Slices were stained with Hematoxylin-eosin (HE) and D2-40 to obtain the results of pleural structures. RESULTS We prospectively evaluated 48 subjects between March 12, 2024 and August 17, 2024. There was no significant difference in PA levels between people living in urban and rural areas (P = 0.32). However, the proportion of rural population in patients with PA level 3 was 2.6 times that of urban population. Patients who were male demonstrated significantly higher level of PA (P = 0.01). With the increase of the smoking index, the patients with PA levels increased. PA levels increased with age (P = 0.02). With the decrease of the FEV1/FVC, the patients with PA levels also increased (P = 0.03), and the patients with PA ratio also increased (P = 0.007). With the continuous improvement of PA ratio and upgrading of PA level, the pleural thickness continued to decrease(R = -0.75, P < 0.01), the number of subpleural lymphatic vessels (SLVs) increased (P < 0.01), cross-sectional area of SLVs increased (P < 0.01), length of SLVs increased (P < 0.01), and SLVs density increased (P < 0.01). CONCLUSIONS The current study demonstrated that the proportion of PA is negatively correlated with pleural thickness and positively correlated with the number of SLVs, cross-sectional area of SLVs, length of SLVs, and SLVs density. This study provides us with new evidence for the subpleural lymphatic metastasis pathways.
Collapse
Affiliation(s)
- Weipeng Shao
- Department of Thoracic Surgical Ward Ⅱ, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, China.
| | - Hongbo Guo
- Department of Thoracic Surgical Ward Ⅱ, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, China
| | - Jian Sun
- Department of Thoracic Surgical Ward Ⅱ, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, China
| | - Zhan Liu
- Department of Thoracic Surgical Ward Ⅱ, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, China
| | - Ziyi Chang
- Department of Pathology, China-Japan Friendship Hospital, Beijing, China
| | - Yang Hao
- Department of General Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Hui Li
- Department of Thoracic Surgical Ward Ⅱ, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, China
| | - Bobo Li
- Department of Thoracic Surgical Ward Ⅱ, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, China
| | - Feng Chen
- Department of Thoracic Surgical Ward Ⅱ, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, China
| | - Jie Liu
- Department of Thoracic Surgical Ward Ⅱ, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, 250117, Shandong, China
| |
Collapse
|
19
|
Arebro J, Pournaras N, Ramos-Ramírez P, Cardenas EI, Bandeira E, Che KF, Brundin B, Bossios A, Karimi R, Nyrén S, Stjärne P, Sköld M, Lindén A. Nasal production of IL-26 involving T cells in smokers with and without chronic obstructive pulmonary disease. J Allergy Clin Immunol 2025:S0091-6749(25)00332-X. [PMID: 40158635 DOI: 10.1016/j.jaci.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/28/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Novel specific therapy in chronic obstructive pulmonary disease (COPD) will require accessible targets for endotyping to identify responsive patients. It is therefore of interest that IL-26 in the bronchoalveolar space is enhanced and associates with bronchoalveolar pathology among long-term smokers (LTS) with and without COPD. OBJECTIVE We determined whether IL-26 in the nasal cavity can be produced by T cells and associates with bronchoalveolar pathology and clinical symptoms in LTS with and without COPD. METHODS We characterized LTS with and without COPD plus healthy nonsmokers by radiology, spirometry, modified Medical Research Council scale, and St George Respiratory Questionnaire. We determined extracellular IL-26 concentrations (via ELISA) in nasal (NAL) and bronchoalveolar lavage (BAL) samples, BAL neutrophil counts, and NAL IL-26+ T-cell expression (via flow cytometry). RESULTS The NAL IL-26 concentrations were higher in LTS with COPD than in healthy nonsmokers. These enhanced IL-26 concentrations displayed a positive correlation with forced expiratory volume in 1 second/forced vital capacity ratio. The IL-26 protein was expressed in CD4+ and CD8+ T cells, but only a small portion of these cells coexpressed IL-15, IL-17A, or IL-22 in LTS with COPD. In this group, IL-26+ CD3+ T cells displayed a negative correlation with forced expiratory volume in 1 second, as did with extracellular NAL IL-26 concentrations. The relative mean fluorescence intensity for CD8+ T cells displayed a negative correlation with modified Medical Research Council and St George Respiratory Questionnaire score. CONCLUSION In the nasal cavity, IL-26 can be produced by local T cells. This IL-26 reflects bronchoalveolar pathology and clinical symptoms, thereby constituting an accessible target with potential for clinically relevant endotyping in COPD.
Collapse
Affiliation(s)
- Julia Arebro
- Division of Ear, Nose, and Throat (ENT) Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden; Department of ENT Diseases, Karolinska University Hospital, Stockholm, Sweden.
| | - Nikolaos Pournaras
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska Severe COPD Center, Karolinska University Hospital, Stockholm, Sweden
| | - Patricia Ramos-Ramírez
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eduardo I Cardenas
- Division of Ear, Nose, and Throat (ENT) Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden; Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elga Bandeira
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karlhans Fru Che
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bettina Brundin
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Apostolos Bossios
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska Severe COPD Center, Karolinska University Hospital, Stockholm, Sweden
| | - Reza Karimi
- Division for Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Sven Nyrén
- Division of Radiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Pär Stjärne
- Division of Ear, Nose, and Throat (ENT) Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden; Department of ENT Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Sköld
- Department of Respiratory Medicine and Allergy, Karolinska Severe COPD Center, Karolinska University Hospital, Stockholm, Sweden; Division for Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Anders Lindén
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska Severe COPD Center, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
20
|
Roth D, Şahin AT, Ling F, Tepho N, Senger CN, Quiroz EJ, Calvert BA, van der Does AM, Güney TG, Glasl S, van Schadewijk A, von Schledorn L, Olmer R, Kanso E, Nawroth JC, Ryan AL. Structure and function relationships of mucociliary clearance in human and rat airways. Nat Commun 2025; 16:2446. [PMID: 40069153 PMCID: PMC11897160 DOI: 10.1038/s41467-025-57667-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025] Open
Abstract
Mucociliary clearance is a vital defense mechanism of the human airways, protecting against harmful particles and infections. When this process fails, it contributes to respiratory diseases like chronic obstructive pulmonary disease (COPD) and asthma. While advances in single-cell transcriptomics have revealed the complexity of airway composition, much of what we know about how airway structure impacts clearance relies on animal studies. This limits our ability to create accurate human-based models of airway diseases. Here we show that the airways in female rats and in humans exhibit species-specific differences in the distribution of ciliated and secretory cells as well as in ciliary beat, resulting in significantly higher clearance effectiveness in humans. We further reveal that standard lab-grown cultures exhibit lower clearance effectiveness compared to human airways, and we identify the underlying structural differences. By combining diverse experiments and physics-based modeling, we establish universal benchmarks to assess human airway function, interpret preclinical models, and better understand disease-specific impairments in mucociliary clearance.
Collapse
Affiliation(s)
- Doris Roth
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Ayşe Tuğçe Şahin
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Feng Ling
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Niels Tepho
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Christiana N Senger
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Erik J Quiroz
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ben A Calvert
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anne M van der Does
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tankut G Güney
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Sarah Glasl
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Annemarie van Schadewijk
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Laura von Schledorn
- Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
- Biomedical Research in End stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Ruth Olmer
- Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
- Biomedical Research in End stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Eva Kanso
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Janna C Nawroth
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany.
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany.
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany.
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany.
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, USA.
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, USA.
| | - Amy L Ryan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, USA.
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
21
|
Wei Y, Ma J. Effective-Component Compatibility of Bufei Yishen Formula (ECC-BYF) III Inhibits Mucus Hypersecretion by BEAS-2B Cells via miR-146a-5p-Mediated Regulation of the EGFR/MEK/ERK Pathway. Int J Chron Obstruct Pulmon Dis 2025; 20:623-639. [PMID: 40092321 PMCID: PMC11908394 DOI: 10.2147/copd.s498477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 02/18/2025] [Indexed: 03/19/2025] Open
Abstract
Purpose To explore the role of the miR-146a-5p-mediated regulation of the EGFR/MEK/ERK pathway in the effect of effective-component compatibility of Bufei Yishen Formula III (ECC-BYF III) on ameliorating mucus hypersecretion by bronchial epithelial cells (BEAS-2B cells). Methods BEAS-2B cells exposed to cigarette smoke extract (CSE) were used to establish a mucus hypersecretion model of BEAS-2B cells. The optimal intervention concentration of ECC-BYF III was screened by CCK-8, qRT-PCR and ELISA, the effects of ECC-BYF III on MUC5AC, MUC5B, IL-4, IL-8, TNF-α, IL-1α, miR-146a-5p and EGFR/MEK/ERK pathway expression were assessed. Furthermore, dual luciferase reporter gene was used to verify the relationship between miR-146a-5p and EGFR/MEK/ERK, and to observe the effect of down-regulating miR-146a-5p on ECC-BYF III ameliorating mucus hypersecretion and EGFR/MEK/ERK pathway. Results ECC-BYF III reduced the expression of MUC5AC and MUC5B, decreased the mRNA expression of IL-1α, IL-8 and TNF-α, increased the mRNA expression of IL-4, and decreased the protein expression of TNF-α. Moreover, ECC-BYF III ameliorated CSE induced mucus hypersecretion in BEAS-2B cells through EGFR/MEK/ERK pathway. Finally, our results indicated that ECC-BYF III ameliorated the model by targeting miR-146a-5p and downregulating the EGFR/MEK/ERK pathway. Conclusion ECC-BYF III can ameliorate CSE induced mucus hypersecretion by BEAS-2B cells and reduce the inflammatory response. The underlying mechanism may be related to the regulation of miR-146a-5p and the EGFR/MEK/ERK pathway. ECC-BYF III can inhibit activation of the EGFR/MEK/ERK pathway by upregulating the expression of miR-146a-5p, thereby ameliorating mucus hypersecretion by BEAS-2B cells.
Collapse
Affiliation(s)
- Yumeng Wei
- Traditional Chinese Medicine (Zhongjing) School, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Jindi Ma
- Traditional Chinese Medicine (Zhongjing) School, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People’s Republic of China, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| |
Collapse
|
22
|
Jia S, Chen Q, Huang W, Wang P, Zeng Y. Relationship between systemic immune response index (SIRI) and COPD: a cross-sectional study based on NHANES 2007-2012. Sci Rep 2025; 15:7887. [PMID: 40050308 PMCID: PMC11885421 DOI: 10.1038/s41598-025-90947-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/17/2025] [Indexed: 03/09/2025] Open
Abstract
Although the link between inflammation and chronic obstructive pulmonary disease (COPD) is increasingly recognized, the correlation between systemic immune response index (SIRI), a novel marker of inflammation, and COPD is unknown. This cross-sectional study used data from patients with complete lung function in NHANES 2007-2012 to explore the relationship between SIRI and COPD. We performed a series of statistical analyses on a total of 5056 participants, including multiple linear regression, smoothed curve fitting, ROC curve analysis, and subgroup analysis. In the fully corrected model, the logistic multiple regression showed that SIRI was associated with a high risk of COPD (OR1.350, 95% CI:1.220,1.493). The ROC curve showed that SIRI (AUC = 0.596) was significantly more efficient than other inflammatory factors in predicting COPD. Smoothed curve fit effect and threshold effect analyses showed a linear correlation between SIRI COPD prevalence, and subgroup analyses showed that the effect of SIRI on COPD was more pronounced in still smokers (OR 1.58, 95% CI: 1.34, 1.86) versus men (OR 1.62, 95% CI: 1.44, 1.83). The results of the interaction test provide evidence supporting SIRI as an independent risk factor for COPD.
Collapse
Affiliation(s)
- Shengqi Jia
- Department of Respiratory and Critical Care Medicine, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuying Chen
- Department of Respiratory and Critical Care Medicine, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijia Huang
- Department of Geriatrics, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Wang
- Department of Respiratory and Critical Care Medicine, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yulan Zeng
- Department of Respiratory and Critical Care Medicine, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
23
|
Hussain MS, Goyal A, Goyal K, S. RJ, Nellore J, Shahwan M, Rekha A, Ali H, Dhanasekaran M, MacLoughlin R, Dua K, Gupta G. Targeting CXCR2 signaling in inflammatory lung diseases: neutrophil-driven inflammation and emerging therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025. [DOI: 10.1007/s00210-025-03970-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 02/20/2025] [Indexed: 05/04/2025]
|
24
|
Rasmusen SKH, Mortensen J. Can the single-breath alveolar volume be adjusted to estimate true total lung capacity? Eur Clin Respir J 2025; 12:2470002. [PMID: 40034812 PMCID: PMC11873942 DOI: 10.1080/20018525.2025.2470002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
Background Total lung capacity (TLC) measured with single-breath gas diffusion (TLCsb) is systematically lower than TLC measured with whole-body plethysmography (TLCwbp) especially in patients with obstructive defects. We aimed to develop and validate a regression correction equation to reduce the discrepancy between the two measurements of TLC. Second, we compared the ability to detect restriction (reduced TLC) from adjusted TLC measured by single-breath (TLCsbadj) with gold standard TLCwbp. Methods Lung function data from 800 consecutive patients were analysed with multivariable linear regression. A group of 530 were included for model development, and 270 were used for model validation. Results TLCsb was found to be on average 1.1 L lower than TLCwbp (p < 0.001). This difference increased with degree of airway obstruction. After adjustment TLCsbadj did not significantly differ from TLCwbp in obstructive and mixed obstructive-restrictive subjects. TLCsbadj had a sensitivity of 70% and a specificity of 99% to predict restriction on an individual basis, with a 95% confidence interval (CI) of [-19.6%; 17.7%] percentage when comparing adjusted values of TLCsb with the true TLCwbp value. Conclusions After adjustment TLCsb was no longer significantly underestimated in obstructive and mixed restrictive-obstructive groups compared to TLCwbp. The adjustment can be used on individual subjects to estimate restriction via the TLCsb, thereby making the single-breath gas diffusion method a more valid alternative than without adjustment, when compared with the gold standard whole-body plethysmography to measure TLC.
Collapse
Affiliation(s)
- Simon Kristoffer Høgh Rasmusen
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jann Mortensen
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Medicine, The National Hospital, Torshavn, Faroe Island
| |
Collapse
|
25
|
Bartziokas K, Papaioannou AI, Rovina N, Papaiakovou G, Loukides S, Steiropoulos P. Εosinophilic Chronic Obstructive Pulmonary Disease. What Do We Know So Far? Pulm Ther 2025; 11:7-24. [PMID: 39661227 PMCID: PMC11861475 DOI: 10.1007/s41030-024-00280-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
The latest advances in asthma treatment have highlighted the significance of eosinophilia and the possible role of some pro-eosinophilic mediators, like interleukins (IL) IL-5, IL-4/IL-13, and IL-33 in the disease's pathogenesis. Considering that a subgroup of patients with chronic obstructive pulmonary disease (COPD) may have blood eosinophilia akin to that seen in asthma, numerous studies in the last decade have suggested that eosinophilic COPD is a separate entity. While the exact role of blood eosinophils in the pathophysiology of COPD remains unclear, eosinophilia seems to increase the effectiveness of corticosteroid therapy. Currently, monoclonal antibodies targeting the interleukins (IL-5, IL-4, IL-13, and IL-33) or their receptors are being investigated in patients with COPD belonging in T2-high endotype. This review focuses on the mechanisms of eosinophilia in COPD, the effects of eosinophilia on disease outcome, and examines the most recent data on the use of peripheral blood eosinophilia in treating patients with COPD. Finally, we emphasize the current implication of monoclonal antibodies in COPD in the context of eosinophilic airway inflammation.
Collapse
Affiliation(s)
| | - Andriana I Papaioannou
- 1st Respiratory Medicine Department, Medical School, Sotiria Chest Diseases Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikoletta Rovina
- 1st Respiratory Medicine Department, Medical School, Sotiria Chest Diseases Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Papaiakovou
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Stelios Loukides
- 2nd Respiratory Medicine Department, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Paschalis Steiropoulos
- Department of Pneumonology, Medical School, Democritus University of Thrace, 68100, Alexandroupolis, Greece.
| |
Collapse
|
26
|
Jin KN, Lee HJ, Park H, Lee JK, Heo EY, Kim DK, Lee HW. Mucus Plugs as Precursors to Exacerbation and Lung Function Decline in COPD Patients. Arch Bronconeumol 2025; 61:138-146. [PMID: 39122616 DOI: 10.1016/j.arbres.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Mucus plugs identified through chest computed tomography (CT) scans have emerged as potential prognostic factors in chronic obstructive pulmonary disease (COPD). This 5-year longitudinal study investigated their impact on exacerbations and FEV1 decline. METHODS COPD patients with baseline chest CT and spirometric assessments were categorized based on mucus plug presence. Propensity-score matching yielded balanced groups. Exacerbation rates, time to exacerbation events, hazard ratio (HR) for exacerbations, and annual rates of FEV1 decline were evaluated. Sensitivity analysis was performed with stratification according to mucus plug scores of 0, 1-2, and ≥3. RESULTS Among 623 eligible patients, the mucus plug group was 44.3%. Through 1:1 propensity-score matching, each group was comprised of 187 individuals with balanced covariates. The mucus plug group showed higher rates of moderate-to-severe (0.51/year vs. 0.58/year, P=0.035), severe exacerbations (0.21/year vs. 0.24/year, P=0.032), and non-eosinophilic exacerbations (0.45/year vs. 0.52/year, P=0.008). Mucus plugs were associated with increased hazard of moderate-to-severe (adjusted HR=1.502 [95% CI 1.116-2.020]), severe (adjusted HR=2.106 [95% CI, 1.429-3.103]), and non-eosinophilic exacerbations (adjusted HR=1.551 [95% CI, 1.132-2.125]). Annual FEV1 decline was accelerated in the mucus plug group (β-coefficient=-62 [95% CI, -120 to -5], P=0.035). Sensitivity analysis showed higher risk of exacerbations and accelerated FEV1 decline in mucus plug score ≥3 compared to score 0. CONCLUSIONS Mucus plugs are associated with increased risks of exacerbations, particularly non-eosinophilic, and accelerated FEV1 declines over 5 years. Our study identified the potential prognostic value of mucus plugs on future exacerbation risks and lung function decline trajectories.
Collapse
Affiliation(s)
- Kwang Nam Jin
- Department of Radiology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyo Jin Lee
- Division of Respiratory and Critical Care, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Heemoon Park
- Division of Respiratory and Critical Care, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jung-Kyu Lee
- Division of Respiratory and Critical Care, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eun Young Heo
- Division of Respiratory and Critical Care, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Deog Kyeom Kim
- Division of Respiratory and Critical Care, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Woo Lee
- Division of Respiratory and Critical Care, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Izquierdo-Condoy JS, Gualpa Álvarez F, Morales-Lapo E, Arias Calvache WD, Bermúdez Moreira J, Quinga-Chiguano KM, Ortiz-Prado E. Adherence to Inhalation Therapy Among COPD Patients: A Cross-Sectional Study in a Tertiary Hospital in Quito, Ecuador. Int J Chron Obstruct Pulmon Dis 2025; 20:399-410. [PMID: 40008110 PMCID: PMC11853065 DOI: 10.2147/copd.s493992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Background Chronic Obstructive Pulmonary Disease (COPD), the third leading cause of death globally, poses a significant public health burden. Despite its high prevalence, underdiagnosis and poor treatment adherence remain major challenges, contributing to increased hospitalization and mortality. Purpose This study aimed to assess adherence to inhalation therapy among COPD patients treated at a specialty hospital in Quito, Ecuador. Patients and Methods A cross-sectional study was conducted on 85 patients diagnosed with COPD at a tertiary hospital in Quito. Data was collected through face-to-face surveys, utilizing the TAI-10 questionnaire to assess treatment adherence, along with demographic and clinical characteristics of the patients. Results Among the participants, 34.1% demonstrated good adherence to inhalation therapy, while 32.9% exhibited intermediate adherence, and 32.9% were non-adherent. The majority were older adults aged ≥76 years (67.1%) with a high prevalence of comorbidities (91.8%), particularly hypertension (58.8%). Non-adherence was primarily associated with forgetfulness and financial constraints. Higher BMI was significantly linked to better adherence, with obese patients showing higher odds of intermediate (OR=7.228, 95% CI 1.866-27.996) and good adherence (OR=9.966, 95% CI 2.538-39.139). Conclusion Approximately one-third of COPD patients in Ecuador demonstrate good adherence to inhalation therapy, while similar proportions show intermediate or poor adherence. Predominantly older adults and retirees with comorbidities, the population identified forgetfulness and financial constraints as key barriers. These findings emphasize the need for enhanced patient education, regular follow-ups, and tailored support for vulnerable groups.
Collapse
|
28
|
Roth D, Şahin AT, Ling F, Tepho N, Senger CN, Quiroz EJ, Calvert BA, van der Does AM, Güney TG, Glasl S, van Schadewijk A, von Schledorn L, Olmer R, Kanso E, Nawroth JC, Ryan AL. Structure and Function Relationships of Mucociliary Clearance in Human and Rat Airways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.24.572054. [PMID: 38187619 PMCID: PMC10769450 DOI: 10.1101/2023.12.24.572054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Mucociliary clearance is a vital defense mechanism of the human airways, protecting against harmful particles and infections. When this process fails, it contributes to respiratory diseases like chronic obstructive pulmonary disease (COPD) and asthma. While advances in single-cell transcriptomics have revealed the complexity of airway composition, much of what we know about how airway structure impacts clearance relies on animal studies. This limits our ability to create accurate human-based models of airway diseases. Here we show that the airways in female rats and in humans exhibit species-specific differences in the distribution of ciliated and secretory cells as well as in ciliary beat, resulting in significantly higher clearance effectiveness in humans. We further reveal that standard lab-grown cultures exhibit lower clearance effectiveness compared to human airways, and we identify the underlying structural differences. By combining diverse experiments and physics-based modeling, we establish universal benchmarks to assess human airway function, interpret preclinical models, and better understand disease-specific impairments in mucociliary clearance.
Collapse
Affiliation(s)
- Doris Roth
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Munich, D-81675, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Ayşe Tuğçe Şahin
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Munich, D-81675, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Feng Ling
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Munich, D-81675, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Niels Tepho
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Munich, D-81675, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Christiana N. Senger
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Erik J. Quiroz
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ben A. Calvert
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Anne M. van der Does
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tankut G. Güney
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Munich, D-81675, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Sarah Glasl
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Annemarie van Schadewijk
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Laura von Schledorn
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Hannover, D-30625, Germany
- Biomedical Research in End stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, D-30625, Germany
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, D-30625, Germany
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Hannover, D-30625, Germany
- Biomedical Research in End stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, D-30625, Germany
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, D-30625, Germany
| | - Eva Kanso
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Janna C. Nawroth
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Munich, D-81675, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, IA 52242, USA
| |
Collapse
|
29
|
Wein AN, Liu CR, Kreisel D. Bronchus-associated lymphoid tissue in lung transplantation: a facilitator of rejection or regulator of tolerance? Front Immunol 2025; 16:1553533. [PMID: 39975555 PMCID: PMC11835794 DOI: 10.3389/fimmu.2025.1553533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
The role of bronchus-associated lymphoid tissue (BALT) in the regulation of immune responses to transplanted lungs remains an area of interest and controversy. Early studies in a rat pulmonary transplant model suggested BALT may accelerate rejection of grafts by inducing a local and systemic inflammatory response. Such observations were corroborated in intrapulmonary tracheal transplant models in the rat. While some human studies have described the presence of BALT in grafts that have been chronically rejected, others did not observe an association between induction of BALT and adverse outcomes. More recent investigations have found that BALT, enriched in immunoregulatory cell populations, is induced in tolerant mouse lung allografts, suggesting that such structures may be protective against rejection. Thus, the role of BALT in lung transplantation biology is complex. Insights gained from studies that focus on the role of BALT in lung transplantation may be harnessed to develop new therapies.
Collapse
Affiliation(s)
- Alexander N. Wein
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Charles R. Liu
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| | - Daniel Kreisel
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
30
|
Lopes AJ. Pulmonary function tests: an integrated approach to interpreting results in the search for treatable traits. Expert Rev Respir Med 2025; 19:121-143. [PMID: 39855910 DOI: 10.1080/17476348.2025.2458607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
INTRODUCTION Technological advances have led to the proliferation of lung function assessment techniques beyond spirometry in most countries. At the same time, new knowledge of respiratory physiology has allowed an expansion of lung function parameters, requiring an integrated approach to interpreting results. AREAS COVERED This review addresses the major pulmonary function tests (PFTs) used in clinical practice, new concepts regarding reference values, and reformulations of terminology for defining standards of lung function impairment. It highlights the complexities and nuances inherent in the interpretation of PFT parameters, particularly in light of recent updates from the European Respiratory Society/American Thoracic Society. EXPERT OPINION In a new paradigm, PFTs should be used to classify the pathophysiology of treatable traits rather than to diagnose respiratory disease, given the considerable variation in the clinical patterns of PFTs. It is necessary to look not only at lung mechanics but also at lung volume, gas transfer, and small airway involvement to capture as much information as possible. In this context, it is also important to understand that racial/ethnic differences in lung function are not due to biological differences but may reflect socioeconomic status and represent health disparities.
Collapse
Affiliation(s)
- Agnaldo José Lopes
- Medical Sciences Post-Graduation Programme, School of Medical Sciences, State University of Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
- Rehabilitation Sciences Post-Graduation Programme, Augusto Motta University Centre (UNISUAM), Rio de Janeiro, Brazil
| |
Collapse
|
31
|
Matson SM, Choi J, Rorah D, Khan S, Trofimoff A, Kim T, Lee DH, Abdolijomoor A, Chen M, Azeem I, Ngo L, Bang TJ, Sachs P, Deane KD, Demoruelle MK, Castro M, Lee JS. Airways Abnormalities in a Prospective Cohort of Patients With Rheumatoid Arthritis. Chest 2025; 167:495-506. [PMID: 39343293 PMCID: PMC11867896 DOI: 10.1016/j.chest.2024.09.006] [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: 04/10/2024] [Revised: 07/02/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) affects roughly 1% of the population and commonly involves the lungs. Of lung involvement in RA, interstitial lung disease (ILD) is well known; however, airways disease in RA is relatively understudied. RESEARCH QUESTION What are the baseline airways abnormalities in a prospective cohort of patients with RA based on pulmonary function testing (PFT) results, high-resolution CT (HRCT) scans, and computational imaging analysis and are there associations between these abnormalities and respiratory symptoms? STUDY DESIGN AND METHODS In this single-center study, 188 patients with RA without a clinical diagnosis of ILD underwent HRCT imaging and PFT. Radiologists assessed HRCT scans for airway abnormalities. Computational imaging via VIDA Vision software and in-house quantitative CT imaging analysis was applied to 147 HRCT scans to quantify airway abnormalities. RESULTS Airways obstruction (FEV1 to FVC ratio < 0.7) was present in 20.7% of patients and was associated with older age, male sex, and higher smoking rate. Radiologists identified airway abnormalities in 61% of patients: 55% had bronchial wall thickening, 12% had bronchiectasis, and 5% had mosaic attenuation. These airways findings were associated with older age; male sex; lower FEV1, FVC, and FEV1 to FVC ratio; and higher rates of rheumatoid factor positivity. Prespecified quantitative CT scan metrics (wall thickening percentage and emphysema percentage) correlated with obstruction in PFT results and more severe respiratory symptoms, including shortness of breath and cough. INTERPRETATION High rates of airways abnormalities were found in this prospective RA cohort based on three methods of detection. Significant associations were identified between quantitative CT scan measures and respiratory symptoms. Airways disease may be an underrecognized extra-articular manifestation of RA and quantitative CT imaging may be a sensitive method to detect the clinical impact on respiratory symptoms.
Collapse
Affiliation(s)
- Scott M Matson
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Cente, Kansas City, KS.
| | - Jiwoong Choi
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Cente, Kansas City, KS
| | - Drayton Rorah
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Shamir Khan
- University of Kansas School of Medicine, Kansas City, KS
| | - Anna Trofimoff
- University of Kansas School of Medicine, Kansas City, KS
| | - Taewon Kim
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Cente, Kansas City, KS
| | - David H Lee
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Cente, Kansas City, KS
| | - Asma Abdolijomoor
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Cente, Kansas City, KS
| | - Maggie Chen
- University of Kansas School of Medicine, Kansas City, KS
| | - Imaan Azeem
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Cente, Kansas City, KS
| | - Linh Ngo
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Cente, Kansas City, KS
| | - Tami J Bang
- Department of Thoracic Radiology, National Jewish Hospital, Denver, CO
| | - Peter Sachs
- Department of Radiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Kevin D Deane
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - M Kristen Demoruelle
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Mario Castro
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Cente, Kansas City, KS
| | - Joyce S Lee
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
32
|
Dournes G, Benlala I. Breaking new ground in COPD imaging: 3D-PREFUL MRI versus traditional techniques. Eur Radiol 2025; 35:940-942. [PMID: 39136710 DOI: 10.1007/s00330-024-10994-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 02/01/2025]
Affiliation(s)
- Gaël Dournes
- Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, University of Bordeaux, INSERM, CIC 1401, F-33600, Pessac, France.
- CHU Bordeaux, Service d'Imagerie Thoracique et Cardiovasculaire, Service des Maladies Respiratoires, Service d'Exploration Fonctionnelle Respiratoire, paediatric Cystic Fibrosis Reference Center (CRCM), CIC 1401, F-33600, Pessac, France.
- Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, INSERM, CIC 1401, F-33600, Pessac, France.
| | - Ilyes Benlala
- Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, University of Bordeaux, INSERM, CIC 1401, F-33600, Pessac, France
- CHU Bordeaux, Service d'Imagerie Thoracique et Cardiovasculaire, Service des Maladies Respiratoires, Service d'Exploration Fonctionnelle Respiratoire, paediatric Cystic Fibrosis Reference Center (CRCM), CIC 1401, F-33600, Pessac, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, INSERM, CIC 1401, F-33600, Pessac, France
| |
Collapse
|
33
|
D R, Johnson P, Das S, Gr S. Serum Soluble Urokinase-Type Plasminogen Activator Receptor: A Promising Biomarker for Stable Chronic Obstructive Pulmonary Disease Patients. Cureus 2025; 17:e79594. [PMID: 40151757 PMCID: PMC11947502 DOI: 10.7759/cureus.79594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a condition in which airflow limitation becomes irreversible over time, often resulting from long-term exposure to environmental pollutants, harmful particles, smoke, and biomass fuel. Beyond FEV1, identifying a more specific biomarker to predict COPD progression remains a challenge. Soluble urokinase-type plasminogen activator receptor (suPAR) expression increases in the respiratory epithelial cells of COPD patients. This study aimed to evaluate serum suPAR levels across different grades of stable COPD patients. Methods Two hundred stable COPD patients (148 males and 52 females) were recruited after obtaining informed consent. Blood samples were collected, and serum suPAR levels were measured in all participants. Results Serum suPAR levels were elevated in COPD patients at Global Initiative for Obstructive Lung Disease (GOLD) stages IV and III (6.38 ± 0.05 ng/ml and 5.82 ± 0.18 ng/ml, respectively) compared to those at GOLD stages II and I (5.15 ± 0.25 ng/ml and 4.17 ± 0.29 ng/ml). A one-way ANOVA confirmed that the differences between groups were statistically significant (F = 428.83, p < 0.001). Conclusions This study suggests that serum suPAR levels can serve as a diagnostic marker for COPD. As low-grade pulmonary inflammation increases with disease severity, suPAR levels also rise. Additionally, this marker may be useful for monitoring the prognosis of stable COPD and assessing treatment response.
Collapse
Affiliation(s)
- Rekha D
- Physiology, Pondicherry Institute of Medical Sciences, Puducherry, IND
| | - Priscilla Johnson
- Physiology, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - Subhasis Das
- Physiology, Pondicherry Institute of Medical Sciences, Puducherry, IND
| | - Sathya Gr
- Physiology, Pondicherry Institute of Medical Sciences, Puducherry, IND
| |
Collapse
|
34
|
Petit LM, Saber Cherif L, Devilliers MA, Hatoum S, Ancel J, Delepine G, Durlach A, Dubernard X, Mérol JC, Ruaux C, Polette M, Deslée G, Perotin JM, Dormoy V. Glypican-3 is a key tuner of the Hedgehog pathway in COPD. Heliyon 2025; 11:e41564. [PMID: 39844999 PMCID: PMC11751517 DOI: 10.1016/j.heliyon.2024.e41564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/12/2024] [Accepted: 12/27/2024] [Indexed: 01/24/2025] Open
Abstract
Hedgehog (HH) pathway is involved in pulmonary development and lung homeostasis. It orchestrates airway epithelial cell (AEC) differentiation and contributes to respiratory pathogenesis. The core elements Gli2, Smo, and Shh were found altered in the bronchial epithelium of patients with chronic obstructive pulmonary disease (COPD). Here, we investigated the co-receptors to fully decipher the complex machinery of airway HH pathway activation in health and COPD. The core elements and co-receptors of HH signalling were investigated in lung cell populations using single-cell RNAseq analysis. The transcript levels of the principal co-receptor GPC3 were investigated on public RNAseq datasets and by RT-qPCR. The localisation of GPC3 was evaluated through immunofluorescent stainings on isolated bronchial AEC and tissues from non-COPD and COPD patients. GPC3 pharmacological modulation was achieved with Codrituzumab during AEC differentiation. We demonstrated that the core elements were not abundant in pulmonary cell populations. Focusing on co-receptors, GPC3 was the most expressed transcript in tracheobronchial epithelial cells. The decrease in GPC3 transcript levels correlated with the severity of airway obstrution in COPD patients. Finally, interfering with GPC3 signalling during AEC differentiation induced downregulation of the HH pathway attested by a decrease of Gli2 leading to reduced ciliogenesis and altered mucin production. GPC3 appears as a crucial co-receptor for the HH pathway in the respiratory context. The modulation of GPC3 may represent a novel experimental strategy to tune HH signalling in therapeutic perspectives.
Collapse
Affiliation(s)
- Laure M.G. Petit
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, Reims, France
| | - Lynda Saber Cherif
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, Reims, France
| | - Maëva A. Devilliers
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, Reims, France
| | - Sarah Hatoum
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, Reims, France
| | - Julien Ancel
- Université de Reims Champagne-Ardenne, INSERM, CHU de Reims, P3Cell UMR-S1250, Reims, France
| | - Gonzague Delepine
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, Reims, France
| | - Anne Durlach
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, Reims, France
| | - Xavier Dubernard
- Université de Reims Champagne-Ardenne, CHU de Reims, Reims, France
| | - Jean-Claude Mérol
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, Reims, France
| | - Christophe Ruaux
- Clinique Mutualiste La Sagesse, Département d’Otorhinolaryngologie, Rennes, France
| | - Myriam Polette
- Université de Reims Champagne-Ardenne, INSERM, CHU de Reims, P3Cell UMR-S1250, Reims, France
| | - Gaëtan Deslée
- Université de Reims Champagne-Ardenne, INSERM, CHU de Reims, P3Cell UMR-S1250, Reims, France
| | - Jeanne-Marie Perotin
- Université de Reims Champagne-Ardenne, INSERM, CHU de Reims, P3Cell UMR-S1250, Reims, France
| | - Valérian Dormoy
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, Reims, France
| |
Collapse
|
35
|
Liao SX, Wang YW, Sun PP, Xu Y, Wang TH. Prospects of neutrophilic implications against pathobiology of chronic obstructive pulmonary disease: Pharmacological insights and technological advances. Int Immunopharmacol 2025; 144:113634. [PMID: 39577220 DOI: 10.1016/j.intimp.2024.113634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/03/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent chronic inflammatory condition that affects the lungs globally. A key feature of this inflammatory response is the migration and aggregation of polymorphonuclear neutrophils (PMNs). The presence of neutrophilic inflammation within the airways is as distinguishing characteristic of COPD. As research advances, PMNs and their products emerge as central players in the airway inflammatory cascade of COPD patients. Their involvement in phagocytosis, degranulation, and the formation of neutrophil extracellular traps (NETs) significantly contributes to the pathogenesis of COPD. Moreover, studies have shown that excessive biological activities of neutrophils in the lungs can result in airway epithelial injury, emphysema, and mucus hypersecretion. Currently, there is growing empirical support for the moderate targeting neutrophils in the clinical management of COPD. This article delves into the pivotal role of neutrophils in COPD, emphasizing the urgency for novel therapeutic approaches that specifically target neutrophils. Additionally, it explores the potential of utilizing single-cell RNA sequencing to further investigate neutrophils and relevant risk genes as potential biomarkers for COPD treatment. By elucidating these mechanisms, this review aims to pave the way for future strategies to modulate neutrophil function, thereby addressing the pressing need for more effective COPD therapies.
Collapse
Affiliation(s)
- Shi-Xia Liao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Yan-Wen Wang
- West China Clinical Medical College, Sichuan University, Chengdu 610041, China
| | - Peng-Peng Sun
- Department of Osteopathy, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Yang Xu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Ting-Hua Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu 610041, China.
| |
Collapse
|
36
|
Li Y, Ge S, Liu J, Li R, Zhang R, Wang J, Pan J, Zhang Q, Zhang J, Zhang M. Peripheral Blood NMLR Can Predict 5-Year All-Cause Mortality in Patients with Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2025; 20:95-105. [PMID: 39802041 PMCID: PMC11725238 DOI: 10.2147/copd.s488877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 12/02/2024] [Indexed: 01/16/2025] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is characterized by pulmonary and systemic inflammation. The peripheral blood (neutrophil + monocyte)/lymphocyte ratio (NMLR) can predict the clinical outcomes of several inflammatory diseases. However, its prognostic value in COPD remains unknown. Methods This retrospective study included 870 patients with COPD due to acute exacerbation, and the 5-year all-cause mortality of these patients was recorded. The Kaplan-Meier method was used to compare the mortality risk of these patients according to their NMLR value. Multivariable COX hazard regression and restricted cubic spline model were used to assess the relationship between the NMLR and 5-year all-cause mortality of patients with COPD. Results The NMLR values of non-surviving patients with COPD were significantly increased compared to the survivors [3.88 (2.53-7.17) vs 2.95 (2.08-4.89), P=0.000]. The area under the NMLR receiver operating characteristic curve for predicting the 5-year all-cause mortality of COPD patients was 0.63. Kaplan-Meier survival curves showed that the 5-year all-cause mortality of COPD patients was significantly increased when the admission peripheral blood NMLR was ≥ 5.90 (27.3% vs 12.4%, P=0.000). The COX regression model showed that NMLR was an independent predictor of 5-year all-cause mortality in COPD patients (hazard ratio=1.84, 95% confidence interval: 1.28-2.64, P=0.001). Moreover, the restricted cubic spline model showed a non-linear relationship between NMLR and COPD death risk (Pnon-linear < 0.05). Conclusion The admission peripheral blood NMLR is a significant predictor of 5-year all-cause mortality in patients with COPD, and high NMLR values may indicate a poor clinical prognosis.
Collapse
Affiliation(s)
- Yuer Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Shaobo Ge
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Jin Liu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Rui Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Rui Zhang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Juan Wang
- Department of Ultrasound, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Jianli Pan
- Department of Special Need, Xi’an Children’s Hospital, Xi’an, Shaanxi, People’s Republic of China
| | - Qiuhong Zhang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Jie Zhang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Ming Zhang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| |
Collapse
|
37
|
Nadeem SA, Zhang X, Nagpal P, Hoffman EA, Chan KS, Comellas AP, Saha PK. Automated CT-based decoupling of the effects of airway narrowing and wall thinning on airway counts in chronic obstructive pulmonary disease. Br J Radiol 2025; 98:150-159. [PMID: 39447037 PMCID: PMC11652725 DOI: 10.1093/bjr/tqae211] [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: 10/20/2023] [Revised: 09/09/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024] Open
Abstract
OBJECTIVE We examine pathways of airway alteration due to wall thinning, narrowing, and obliteration in chronic obstructive pulmonary disease (COPD) using CT-derived airway metrics. METHODS Ex-smokers (N = 649; age mean ± std: 69 ± 6 years; 52% male) from the COPDGene Iowa cohort (September 2013-July 2017) were studied. Total airway count (TAC), peripheral TAC beyond 7th generation (TACp), and airway wall thickness (WT) were computed from chest CT scans using previously validated automated methods. Causal relationships among demographic, smoking, spirometry, COPD severity, airway counts, WT, and scanner variables were analysed using causal inference techniques including direct acyclic graphs to assess multi-pathway alterations of airways in COPD. RESULTS TAC, TACp, and WT were significantly lower (P < .0001) in mild, moderate, and severe COPD compared to the preserved lung function group. TAC (TACp) losses attributed to narrowing and obliteration of small airways were 4.59%, 13.29%, and 32.58% (4.64%, 17.82%, and 45.51%) in mild, moderate, and severe COPD, while the losses attributed to wall thinning were 8.24%, 17.01%, and 22.95% (12.79%, 25.66%, and 33.95%) in respective groups. CONCLUSIONS Different pathways of airway alteration in COPD are observed using CT-derived automated airway metrics. Wall thinning is a dominant contributor to both TAC and TACp loss in mild and moderate COPD while narrowing and obliteration of small airways is dominant in severe COPD. ADVANCES IN KNOWLEDGE This automated CT-based study shows that wall thinning dominates airway alteration in mild and moderate COPD while narrowing and obliteration of small airways leads the alteration process in severe COPD.
Collapse
Affiliation(s)
- Syed Ahmed Nadeem
- Department of Radiology, University of Iowa, Iowa City, IA 52242, United States
| | - Xinyu Zhang
- Department of Statistics and Actuarial Science, University of Iowa, Iowa City, IA 52242, United States
| | - Prashant Nagpal
- Department of Radiology, University of Wisconsin, Madison, WI 53792, United States
| | - Eric A Hoffman
- Department of Radiology, University of Iowa, Iowa City, IA 52242, United States
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, United States
| | - Kung-Sik Chan
- Department of Statistics and Actuarial Science, University of Iowa, Iowa City, IA 52242, United States
| | - Alejandro P Comellas
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, United States
| | - Punam K Saha
- Department of Radiology, University of Iowa, Iowa City, IA 52242, United States
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA 52242, United States
| |
Collapse
|
38
|
Teillaud JL, Regard L, Martin C, Sibéril S, Burgel PR. Exploring the Role of Tertiary Lymphoid Structures Using a Mouse Model of Bacteria-Infected Lungs. Methods Mol Biol 2025; 2864:281-297. [PMID: 39527228 DOI: 10.1007/978-1-0716-4184-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Animal models can be helpful tools for deciphering the generation, maintenance, and role of tertiary lymphoid structures (TLS) during infections or tumor development. We describe here the establishment of a persistent lung infection in immune-competent mice by intratracheal instillation of agarose beads containing Pseudomonas aeruginosa or Staphylococcus aureus bacteria. After instillation, animals develop a chronic pulmonary infection, marked by the presence of TLS. This experimental setting allows the study of the function of TLS induced by bacteria encountered in patients with cystic fibrosis (CF) as P. aeruginosa and S. aureus are the two main bacterial strains that infect the bronchi of adult CF patients. Additionally, we describe also how to manipulate the immune response in these infected animals by targeting immune cells involved in TLS function. Overall, this approach makes it possible to explore the role of chronic inflammation in the induction and maintenance of TLS in infected tissues.
Collapse
Affiliation(s)
- Jean-Luc Teillaud
- UMRS 1135 Sorbonne Université, Faculté de Santé Sorbonne Université, Paris, France.
- Inserm U1135, Paris, France.
- Team "Immune Microenvironment and Immunotherapy", Center of Immunology and Microbial Infections (CIMI), Paris, France.
| | - Lucile Regard
- Université Paris Cité, Institut Cochin, Inserm U1016, Paris, France
- Respiratory Medicine and Cystic Fibrosis National Reference Center, Hôpital Cochin, AP-HP, Université Paris Cité, Paris, France
- European Reference Network on Rare Respiratory Diseases (ERN-Lung, Cystic Fibrosis Core Network), Frankfurt, Germany
| | - Clémence Martin
- Université Paris Cité, Institut Cochin, Inserm U1016, Paris, France
- Respiratory Medicine and Cystic Fibrosis National Reference Center, Hôpital Cochin, AP-HP, Université Paris Cité, Paris, France
- European Reference Network on Rare Respiratory Diseases (ERN-Lung, Cystic Fibrosis Core Network), Frankfurt, Germany
| | - Sophie Sibéril
- UMRS 1138, Sorbonne Université, Université Paris Cité, Paris, France
- Inserm U1138, Paris, France
- Team "Inflammation, Complement, and Cancer", Centre de Recherche des Cordeliers, Paris, France
| | - Pierre-Régis Burgel
- Université Paris Cité, Institut Cochin, Inserm U1016, Paris, France
- Respiratory Medicine and Cystic Fibrosis National Reference Center, Hôpital Cochin, AP-HP, Université Paris Cité, Paris, France
- European Reference Network on Rare Respiratory Diseases (ERN-Lung, Cystic Fibrosis Core Network), Frankfurt, Germany
| |
Collapse
|
39
|
Wu L, Zhang E, Tu Y, Chen Y, Wang C, Ren Y, Fang B. Inherent immunity and adaptive immunity: Mechanism and role in AECOPD. Innate Immun 2025; 31:17534259251322612. [PMID: 40017227 PMCID: PMC11869301 DOI: 10.1177/17534259251322612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 12/20/2024] [Accepted: 02/04/2025] [Indexed: 03/01/2025] Open
Abstract
Acute exacerbation of chronic obstructive pulmonary disease (AECOPD) is the leading cause of hospitalization and mortality in COPD patients. The occurrence of antibiotic resistance and the progression of non-infectious diseases contribute to poor patient outcomes. Thus, a comprehensive understanding of the mechanisms underlying AECOPD is essential for effective prevention. It is widely acknowledged that the immune system plays a fundamental role in pathogen clearance and the development of inflammation. Immune dysregulation, either due to deficiency or hyperactivity, has been implicated in AECOPD pathogenesis. Therefore, the purpose of this review is to investigate the possible mechanisms underlying dysregulated immune function and disease progression in COPD patients, specifically focusing on the innate and adaptive immune responses. The ultimate aim is to provide new insights for clinical prevention and treatment strategies targeting AECOPD.
Collapse
Affiliation(s)
- Linguangjin Wu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Emergency Department, Shanghai, China
| | - Erxin Zhang
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yadan Tu
- Chongqing Hospital of Traditional Chinese Medicine, Classic Department of Traditional Chinese Medicine, Chongqing, China
| | - Yong Chen
- Chongqing Hospital of Traditional Chinese Medicine, Classic Department of Traditional Chinese Medicine, Chongqing, China
| | - Chenghu Wang
- Chongqing Hospital of Traditional Chinese Medicine, Classic Department of Traditional Chinese Medicine, Chongqing, China
| | - Yi Ren
- Chongqing Hospital of Traditional Chinese Medicine, Classic Department of Traditional Chinese Medicine, Chongqing, China
| | - Bangjiang Fang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Emergency Department, Shanghai, China
| |
Collapse
|
40
|
Saccomanno J, Kilic L, Sgarbossa T, Neumann K, Stanzel F, Holland A, Grah C, Gesierich W, Krist J, Ficker JH, Eggeling S, Andreas S, Schmidt B, Eisenmann S, Schwick B, Franke KJ, Fertl A, Witzenrath M, Hübner RH. Clinical improvements after endoscopic lung volume reduction with valves in patients with advanced emphysema and a 6-min walk test ≤140 m at baseline. ERJ Open Res 2025; 11:00410-2024. [PMID: 39811547 PMCID: PMC11726540 DOI: 10.1183/23120541.00410-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/20/2024] [Indexed: 01/16/2025] Open
Abstract
Background Data regarding the effectiveness and safety of endoscopic lung volume reduction with valves (ELVR) in emphysema patients with a very low 6-min walk test (6MWT) are limited. Patients with severe emphysema and very low exercise capacity, as indicated by a 6MWT ≤140 m, are often excluded from clinical studies on ELVR, assuming limited therapeutic benefits and increased complication risk. Study designs and methods This study utilised data from the Lungenemphysemregister e.V., a large German national multi-centre prospective open-label clinical trial, and aimed to assess the outcomes of ELVR in patients with a baseline 6MWT ≤140 m and dyspnoea primarily attributed to hyperinflation. Results 54 patients with a baseline 6MWT ≤140 m and 365 patients with a baseline 6MWT between 140 and 450 m were included in the study. Baseline characteristics were representative for patients with advanced lung emphysema. Patients with a 6MWT ≤140 m at baseline had a lower forced expiratory volume in 1 s and diffusing capacity of the lung for carbon monoxide and higher symptom burden. In the 3-month follow-up, patients of both groups showed statistically significant improvements in lung function parameters, exercise capacity and quality of life parameters compared to baseline. Patients with a 6MWT ≤140 m at baseline showed significantly more 6MWT improvement compared to patients with baseline 6MWT between 140 and 450 m. Moreover, complication rates were similar in both groups. Interpretation In summary, the data indicate that ELVR may be an effective and safe treatment for emphysema patients with a very low 6MWT of ≤140 m if very limited exercise capacity is predominately caused by lung emphysema. Therefore future studies should include emphysema patients with a very low 6MWT.
Collapse
Affiliation(s)
- Jacopo Saccomanno
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lara Kilic
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Sgarbossa
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Konrad Neumann
- Institute of Biometry and Clinical Epidemiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Franz Stanzel
- Department of Respiratory Medicine, Lungenklinik Hemer, Hemer, Germany
| | - Angelique Holland
- Department of Respiratory Medicine, Universitätsklinikum Marburg, Marburg, Germany
| | - Christian Grah
- Department of Respiratory Medicine, Klinik Havelhöhe Berlin, Berlin, Germany
| | - Wolfgang Gesierich
- Department of Respiratory Medicine, Asklepios Fachkliniken München-Gauting, Gautingen, Germany
| | - Joanna Krist
- Department of Respiratory Medicine, Lungenklinik Heckeshorn, Helios Klinikum Emil von Behring, Berlin, Germany
| | - Joachim H. Ficker
- Department of Internal Medicine 3/Respiratory Medicine, Klinikum Nuernberg, Paracelsus Medical University, Nuernberg, Germany
| | - Stephan Eggeling
- Department of Thoracic Surgery, Vivantes-Klinikum Neukölln, Berlin, Germany
| | - Stefan Andreas
- Center of Respiratory Medicine, Lungenfachklinik Immenhausen, Immenhausen, Germany
| | - Bernd Schmidt
- Department of Internal Medicine – Respiratory and Sleep Medicine, DRK Kliniken Berlin Mitte, Berlin, Germany
| | - Stephan Eisenmann
- Department of Respiratory Medicine, Universitätsklinikum Halle, Halle, Germany
| | - Björn Schwick
- Department of Respiratory Medicine, Luisenhospital Aachen, Aachen, Germany
| | - Karl-Josef Franke
- Department of Respiratory Medicine and Critical Care Medicine, Klinikum Lüdenscheid, Lüdenscheid, Germany
- University Witten/Herdecke, Witten, Germany
| | - Andreas Fertl
- Department of Internal Medicine and Respiratory Medicine, Krankenhaus Martha-Maria München, München, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
- Capnetz Foundation, Hannover, Germany
| | - Ralf-Harto Hübner
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
41
|
Khedoe PPSJ, van 't Wout EFA. Buddy, bystander or betrayer: B cells in lymphoid aggregates in AATD emphysema. Eur Respir J 2025; 65:2402163. [PMID: 39746767 DOI: 10.1183/13993003.02163-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 01/04/2025]
Affiliation(s)
- P Padmini S J Khedoe
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Emily F A van 't Wout
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
42
|
Park JH. Clinical Characteristics of Chronic Obstructive Pulmonary Disease according to Smoking Status. Tuberc Respir Dis (Seoul) 2025; 88:14-25. [PMID: 39474731 PMCID: PMC11704726 DOI: 10.4046/trd.2024.0060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/16/2024] [Accepted: 10/28/2024] [Indexed: 01/07/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) can be caused by various factors, including lung infections, asthma, air pollution, childhood growth disorders, and genetic factors, though smoking is the predominant risk factor. The main pathological mechanisms in COPD involve small airway disease, emphysema, mucus hypersecretion, and vascular disorders. COPD in non-smokers is characterized by a normal 1-second forced expiratory volume decline, equal sex distribution, younger age of onset, fewer comorbidities, milder airflow obstruction, preserved diffusing capacity of the lungs for carbon monoxide, and radiological features such as more air-trapping and less severe emphysema compared to COPD in smokers. Nevertheless, non-smokers with COPD still experience a high prevalence of acute exacerbations, nearly equal to that of smokers with COPD. Moreover, COPD itself is an independent risk factor for developing lung cancer, regardless of smoking status. Given that COPD coexists with numerous comorbidities, effectively managing these comorbidities is crucial, requiring multifaceted efforts for comprehensive treatment.
Collapse
Affiliation(s)
- Joo Hun Park
- Department of Pulmonary and Critical Care Medicine, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
43
|
Baffetta F, Buonsanti C, Moraschini L, Aprea S, Canè M, Lombardi S, Contorni M, Rondini S, Arora AK, Bardelli M, Finco O, Serruto D, Paccani SR. Lung mucosal immunity to NTHi vaccine antigens: Antibodies in sputum of chronic obstructive pulmonary disease patients. Hum Vaccin Immunother 2024; 20:2343544. [PMID: 38655676 PMCID: PMC11057560 DOI: 10.1080/21645515.2024.2343544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/12/2024] [Indexed: 04/26/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common chronic respiratory illness in older adults. A major cause of COPD-related morbidity and mortality is acute exacerbation of COPD (AECOPD). Bacteria in the lungs play a role in exacerbation development, and the most common pathogen is non-typeable Haemophilus influenzae (NTHi). A vaccine to prevent AECOPD containing NTHi surface antigens was tested in a clinical trial. This study measured IgG and IgA against NTHi vaccine antigens in sputum. Sputum samples from 40 COPD patients vaccinated with the NTHi vaccine were collected at baseline and 30 days after the second dose. IgG and IgA antibodies against the target antigens and albumin were analyzed in the sputum. We compared antibody signals before and after vaccination, analyzed correlation with disease severity and between sputum and serum samples, and assessed transudation. Antigen-specific IgG were absent before vaccination and present with high titers after vaccination. Antigen-specific IgA before and after vaccination were low but significantly different for two antigens. IgG correlated between sputum and serum, and between sputum and disease severity. Sputum albumin was higher in patients with severe COPD than in those with moderate COPD, suggesting changes in transudation played a role. We demonstrated that immunization with the NTHi vaccine induces antigen-specific antibodies in sputum. The correlation between IgG from sputum and serum and the presence of albumin in the sputum of severe COPD patients suggested transudation of antibodies from the serum to the lungs, although local IgG production could not be excluded.Clinical Trial Registration: NCT02075541.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Simona Rondini
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
| | | | | | | | | | | |
Collapse
|
44
|
Pincikova T, Merikallio H, Kotortsi I, Karimi R, Li CX, Lappi-Blanco E, Lindén SK, Padra M, Wheelock ÅM, Nyrén S, Sköld CM, Kaarteenaho RL. Expression Levels of MUC5AC and MUC5B in Airway Goblet Cells Are Associated with Traits of COPD and Progression of Chronic Airflow Limitation. Int J Mol Sci 2024; 25:13653. [PMID: 39769414 PMCID: PMC11678853 DOI: 10.3390/ijms252413653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Mucins 5AC (MUC5AC) and 5B (MUC5B) are the major mucins providing the organizing framework for the airway's mucus gel. We retrieved bronchial mucosal biopsies and bronchial wash (BW) samples through bronchoscopy from patients with chronic obstructive pulmonary disease (n = 38), healthy never-smokers (n = 40), and smokers with normal lung function (n = 40). The expression of MUC5AC and MUC5B was assessed immunohistochemically. The mucin concentrations in BW were determined using the slot-blot technique. The immunohistochemical expression of MUC5AC and MUC5B was localized to goblet cells and submucosal glands. Smokers had higher MUC5AC and lower MUC5B goblet cell expression and higher concentrations of soluble MUC5AC in BW than never-smokers. The MUC5B expression in goblet cells correlated positively with expiratory air flows, diffusing capacity, and the dyspnoea score. Chronic bronchitis, emphysema, and the progression of chronic airflow limitation during a median follow-up time of 8.4 years were associated with higher MUC5AC and lower MUC5B expression in goblet cells. Sustainers, slow progressors, and rapid progressors of airflow obstruction differed in their MUC5B expression at baseline. Emphysema and bronchial wall thickening on CT at a follow-up visit were associated with lower MUC5B expression at baseline. Our findings strengthen the hypothesis that MUC5AC and MUC5B are yet another contributing factor to smoking-associated lung disease progression.
Collapse
Grants
- NA The Foundation of the Finnish Anti-Tuberculosis Association
- NA The Jalmari and Rauha Ahokas Foundation
- NA The Research Foundation of the Pulmonary Diseases
- NA A state subsidy of Oulu University Hospital
- CMS 20100435, 20130293, 20160418, 20180235, 20190320; SL 20230626 The Swedish Heart-Lung Foundation
- CMS 2016 The Swedish Respiratory Society
- CMS 2011, 2012, 2013 King Gustaf V's and Queen Victoria's Freemasons' Foundation
- CMS 2010 Hesselmans Foundation
- NA Karolinska Institutet
- 2017-01142, 2018-00520 and 2021-02542 The Swedish Research Council
- CMS 071217 The Swedish Cancer- and Allergy Foundation
- CMS 2013 Sandoz A/S
- CMS 20110434, 20130051, 20150061, 20170393 and 20200287 The Regional Agreement on Medical Training and Clinical Research (ALF) between Stockholm County Council and Karolinska Institutet
- MCF [7214]-2013 ERS-EU RESPIRE2
Collapse
Affiliation(s)
- Terezia Pincikova
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institute, 171 76 Stockholm, Sweden (Å.M.W.)
- Stockholm CF-Center, Albatross, K56, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Heta Merikallio
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, 902 20 Oulu, Finland
- Center of Internal Medicine and Respiratory Medicine, Medical Research Center Oulu, Oulu University Hospital, 902 20 Oulu, Finland
| | - Ioanna Kotortsi
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institute, 171 76 Stockholm, Sweden (Å.M.W.)
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Reza Karimi
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institute, 171 76 Stockholm, Sweden (Å.M.W.)
| | - Chuan-Xing Li
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institute, 171 76 Stockholm, Sweden (Å.M.W.)
| | - Elisa Lappi-Blanco
- Cancer and Translational Medicine Research Unit, Department of Pathology, Oulu University Hospital and University of Oulu, 902 20 Oulu, Finland
| | - Sara K. Lindén
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (S.K.L.)
| | - Médea Padra
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (S.K.L.)
| | - Åsa M. Wheelock
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institute, 171 76 Stockholm, Sweden (Å.M.W.)
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Sven Nyrén
- Department of Molecular Medicine and Surgery, Division of Radiology, Karolinska Institute, Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
| | - Carl Magnus Sköld
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institute, 171 76 Stockholm, Sweden (Å.M.W.)
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Riitta L. Kaarteenaho
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, 902 20 Oulu, Finland
- Center of Internal Medicine and Respiratory Medicine, Medical Research Center Oulu, Oulu University Hospital, 902 20 Oulu, Finland
| |
Collapse
|
45
|
Lycan Jr TW, Norton DL, Ohar JA. COPD and Immune Checkpoint Inhibitors for Cancer: A Literature Review. Int J Chron Obstruct Pulmon Dis 2024; 19:2689-2703. [PMID: 39677829 PMCID: PMC11639883 DOI: 10.2147/copd.s490252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose Immune checkpoint inhibitors are a standard treatment option for many patients with cancer and are most frequently used to treat lung cancer. Chronic obstructive pulmonary disease (COPD) is the most common comorbidity of patients with lung cancer. As the cancer-specific survival of patients with lung cancer continues to increase with modern treatments, it is critical to optimize comorbidities to improve overall survival. This literature review aimed to summarize current research on the impact of COPD upon immunotherapy outcomes. Methods A comprehensive search was conducted in the PubMed database using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Inclusion criteria focused on peer-reviewed articles published between 2010 and 2024 that addressed COPD, cancer, and immune checkpoint inhibitors. The study team screened the studies for relevance and then synthesized them narratively. Results This review identified 37 studies that met the inclusion criteria. Findings suggest that COPD is predictive of improved efficacy but slightly worse toxicity from immune checkpoint inhibitor therapy. The chronic inflammation of COPD leads to immune exhaustion including the overexpression of immune checkpoints on T-cells. Particularly within "hot" tumors that have higher concentrations of tumor-infiltrating lymphocytes, the COPD-related increase in programmed cell death protein 1 (PD-1) signaling likely creates sensitivity to immune checkpoint inhibitors. However, COPD can also lead to respiratory dysfunction, debility, and interstitial lung disease; each of which increases the severity of immune-related adverse events. Conclusion COPD is a critical comorbidity that has a significant impact on many patients with cancer who receive treatment with immune checkpoint inhibitors. Future research is needed to design interventions to optimize COPD care in this high-risk patient population.
Collapse
Affiliation(s)
- Thomas W Lycan Jr
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Dustin L Norton
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jill A Ohar
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
46
|
Sen Chaudhuri A, Sun J. Lung-resident lymphocytes and their roles in respiratory infections and chronic respiratory diseases. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:214-223. [PMID: 39834580 PMCID: PMC11742555 DOI: 10.1016/j.pccm.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Indexed: 01/22/2025]
Abstract
Recent scientific breakthroughs have blurred traditional boundaries between innate and adaptive immunity, revealing a sophisticated network of tissue-resident cells that deliver immediate, localized immune responses. These lymphocytes not only provide rapid frontline defense but also present a paradoxical role in the pathogenesis of respiratory diseases such as asthma, chronic obstructive pulmonary disease, pulmonary fibrosis, and the long-term tissue consequences of viral infections including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). This review traverses the intricate landscape of lung-resident lymphocytes, delving into their origins, diverse functions, and their dualistic impact on pulmonary health. We dissect their interactions with the microenvironment and the regulatory mechanisms guiding their activity, with an emphasis on their contribution to both immune protection and immunopathology. This review aims to elucidate the complex narrative of these cells, enhancing our understanding of the development of precise therapeutic strategies to combat acute and chronic pulmonary diseases. Through this exploration, the review aspires to shed light on the potential of harnessing lung-resident lymphocytes for the treatment of respiratory conditions.
Collapse
Affiliation(s)
- Arka Sen Chaudhuri
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jie Sun
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
47
|
Xiao X, Ding Z, Shi Y, Zhang Q. Causal Role of Immune Cells in Chronic Obstructive Pulmonary Disease: A Two-Sample Mendelian Randomization Study. COPD 2024; 21:2327352. [PMID: 38573027 DOI: 10.1080/15412555.2024.2327352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/27/2024] [Indexed: 04/05/2024]
Abstract
Accumulating evidence has highlighted the importance of immune cells in the pathogenesis of chronic obstructive pulmonary disease (COPD). However, the understanding of the causal association between immunity and COPD remains incomplete due to the existence of confounding variables. In this study, we employed a two-sample Mendelian randomization (MR) analysis, utilizing the genome-wide association study database, to investigate the causal association between 731 immune-cell signatures and the susceptibility to COPD from a host genetics perspective. To validate the consistency of our findings, we utilized MR analysis results of lung function data to assess directional concordance. Furthermore, we employed MR-Egger intercept tests, Cochrane's Q test, MR-PRESSO global test, and "leave-one-out" sensitivity analyses to evaluate the presence of horizontal pleiotropy, heterogeneity, and stability, respectively. Inverse variance weighting results showed that seven immune phenotypes were associated with the risk of COPD. Analyses of heterogeneity and pleiotropy analysis confirmed the reliability of MR results. These results highlight the interactions between the immune system and the lungs. Further investigations into their mechanisms are necessary and will contribute to inform targeted prevention strategies for COPD.
Collapse
Affiliation(s)
- Xinru Xiao
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Ziqi Ding
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yujia Shi
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Qian Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| |
Collapse
|
48
|
Alsubait D, Rajani HF, Shan L, Koussih L, Halayko AJ, Lamkhioued B, Gounni AS. Expression of Semaphorin3E/PlexinD1 in human airway smooth muscle cells of patients with COPD. Am J Physiol Lung Cell Mol Physiol 2024; 327:L831-L838. [PMID: 39316678 DOI: 10.1152/ajplung.00284.2023] [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: 09/06/2023] [Revised: 08/15/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024] Open
Abstract
Semaphorin3E (Sema3E) is a member of axon guidance proteins that have emerged recently as essential regulators of cell migration and proliferation. It binds to PlexinD1 with high affinity and is expressed in different cell types, including immune, cancer, and epithelial cells. Recent work in our lab has revealed a critical immunoregulatory role of Sema3E in experimental allergic asthma; however, its role in chronic obstructive pulmonary disease (COPD) remains unclear. This study aimed to investigate the expression of Sema3E and its receptor, PlexinD1, in the airways of patients with COPD and whether Sema3E regulates airway smooth muscle (ASM) cell proliferation, a key feature of airway remodeling in COPD. We first demonstrate that human ASM cells obtained from COPD express Sema3E and PlexinD1 at both mRNA and protein levels. Also, bronchial sections from patients with COPD displayed immunoreactivity of Sema3E and its receptor PlexinD1, suggestive of functional contribution of Sema3E in airway remodeling. In contrast to ASM cells from healthy donors, Sema3E did not inhibit the platelet-derived growth factor (PDGF) induced cell proliferation in ASM cells of patients with COPD that were consistent with the binding of endogenous Sema3E to its receptors on the cell surface and the expression and release of p61KDa-Sema3E isoform. Our results support the Sema3E-PlexinD1 axis involvement in COPD airway smooth muscle remodeling.NEW & NOTEWORTHY Semaphorin3E (Sema3E), a protein guiding cell movement, is found in various cell types like neural, immune, cancer, and epithelial cells. This study examines Sema3E in chronic obstructive pulmonary disease (COPD) airways. In patients with COPD, airway smooth muscle cells express Sema3E and its receptor PlxD1. Unlike healthy cells, Sema3E does not hinder cell proliferation in COPD, indicating involvement in airway remodeling. These findings highlight the Sema3E-PlxD1 axis in COPD airway changes.
Collapse
Affiliation(s)
- Duaa Alsubait
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Prince Sultan Military College of Health Sciences, Ministry Agency for Excellence Services, General Administration of Health Services, Dammam, Kingdom of Saudi Arabia
| | - Huda Fatima Rajani
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lianyu Shan
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Latifa Koussih
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Department de Sciences Experimentales, Université de Saint-Boniface, Winnipeg, Manitoba, Canada
| | - Andrew J Halayko
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Bouchaib Lamkhioued
- Laboratoire d'Immunologie et de Biotechnologie, UR7509-IRMAIC, UFR de Pharmacie, Pôle-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Abdelilah S Gounni
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
49
|
Hermann EA, Sun Y, Hoffman EA, Allen NB, Ambale-Venkatesh B, Bluemke DA, Carr JJ, Kawut SM, Prince MR, Shah SJ, Smith BM, Watson KE, Lima JAC, Barr RG. Lung structure and longitudinal change in cardiac structure and function: the MESA COPD Study. Eur Respir J 2024; 64:2400820. [PMID: 39362671 PMCID: PMC12120402 DOI: 10.1183/13993003.00820-2024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 08/27/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Lung structure and cardiac structure and function are associated cross-sectionally. The classic literature suggests relationships of airways disease to cor pulmonale and emphysema to reduced cardiac output (CO) but longitudinal data are lacking. METHODS The Multi-Ethnic Study of Atherosclerosis (MESA) COPD Study was a multicentre longitudinal COPD case-control study of participants 50-79 years with ≥10 pack-years smoking without clinical cardiovascular disease. Segmental airway wall area (WA) and percent emphysema were measured on computed tomography. Right and left ventricle parameters were assessed on cardiac magnetic resonance imaging (cMRI) in exams 6 years apart. Longitudinal and period cross-sectional associations were evaluated with mixed models adjusted for demographics, body size and smoking. RESULTS The 187 participants with repeated cMRI were 67±7 years old; 42% had COPD; 22% currently smoked; and the race/ethnicity distribution was 54% White, 30% Black, 14% Hispanic and 3% Asian. Greater WA at enrolment was associated with longitudinal increase in right ventricular (RV) mass (3.5 (95% CI 1.1-5.9) g per 10 mm2 WA). Greater percent emphysema was associated with stably lower left ventricular (LV) end-diastolic volume (-7.8 (95% CI -10.3- -3.0) mL per 5% emphysema) and CO (-0.2 (95% CI -0.4- -0.1) L·min-1 per 5% emphysema). CONCLUSION Cardiac associations varied by lung structure over 6 years in this multi-ethnic study. Greater WA at enrolment was associated with longitudinal increases in RV mass, whereas greater percent emphysema was associated with stable decrements in LV filling and CO.
Collapse
Affiliation(s)
- Emilia A Hermann
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Yifei Sun
- Department of Biostatistics, Columbia University Medical Center, New York, NY, USA
| | - Eric A Hoffman
- Department of Radiology, University of Iowa, Iowa City, IA, USA
| | - Norrina B Allen
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | | | - David A Bluemke
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, USA
| | - J Jeffrey Carr
- Department of Radiology, Vanderbilt University, Nashville, TN, USA
| | - Steven M Kawut
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Martin R Prince
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Sanjiv J Shah
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Benjamin M Smith
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Karol E Watson
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Joao A C Lima
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - R Graham Barr
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- Department of Epidemiology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
50
|
Wang H, Zhang Y, Zhao C, Peng Y, Song W, Xu W, Wen X, Liu J, Yang H, Shi R, Zhao S. Serum IL-17A and IL-6 in paediatric Mycoplasma pneumoniae pneumonia: implications for different endotypes. Emerg Microbes Infect 2024; 13:2324078. [PMID: 38407218 PMCID: PMC10997354 DOI: 10.1080/22221751.2024.2324078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
Paediatric Mycoplasma pneumoniae pneumonia (MPP) is a heterogeneous disease with a diverse spectrum of clinical phenotypes. No studies have demonstrated the relationship between underlying endotypes and clinical phenotypes as well as prognosis about this disease. Thus, we conducted a multicentre prospective longitudinal study on children hospitalized for MPP between June 2021 and March 2023, with the end of follow-up in August 2023. Blood samples were collected and processed at multiple time points. Multiplex cytokine assay was performed to characterize serum cytokine profiles and their dynamic changes after admission. Cluster analysis based on different clinical phenotypes was conducted. Among the included 196 patients, the levels of serum IL-17A and IL-6 showed remarkable variabilities. Four cytokine clusters based on the two cytokines and four clinical groups were identified. Significant elevation of IL-17A mainly correlated with diffuse bronchiolitis and lobar lesion by airway mucus hypersecretions, while that of IL-6 was largely associated with lobar lesion which later developed into lung necrosis. Besides, glucocorticoid therapy failed to inhibit IL-17A, and markedly elevated IL-17A and IL-6 levels may correlate with lower airway obliterans. Our study provides critical relationship between molecular signatures (endotypes) and clustered clinical phenotypes in paediatric patients with MPP.
Collapse
Affiliation(s)
- Heng Wang
- Department II of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, People’s Republic of China
| | - Yanli Zhang
- Division of Pulmonology, Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Henan, People’s Republic of China
| | - Chengsong Zhao
- Department of Infectious Diseases, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, People’s Republic of China
| | - Yun Peng
- Department of Radiology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, People’s Republic of China
| | - Wenqi Song
- Department of Clinical Laboratory, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, People’s Republic of China
| | - Weihan Xu
- Department II of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, People’s Republic of China
| | - Xiaohui Wen
- Department II of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, People’s Republic of China
| | - Jinrong Liu
- Department II of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, People’s Republic of China
| | - Haiming Yang
- Department II of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, People’s Republic of China
| | - Ruihe Shi
- Division of Pulmonology, Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Henan, People’s Republic of China
| | - Shunying Zhao
- Department II of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, People’s Republic of China
| |
Collapse
|