1
|
Ding R, Yu J, Ke W, Du L, Cheng G, Hu S, Xu Y, Liu Y. TLR2 regulates Moraxella catarrhalis adhesion to and invasion into alveolar epithelial cells and mediates inflammatory responses. Virulence 2024; 15:2298548. [PMID: 38169345 PMCID: PMC10772937 DOI: 10.1080/21505594.2023.2298548] [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: 05/09/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Moraxella catarrhalis is a major cause of chronic obstructive pulmonary disease. Toll-like receptor 2 (TLR2) plays an important role in the inflammatory response in host respiratory epithelial cells. M. catarrhalis induces an inflammatory immune response in respiratory epithelial cells that is mostly dependent on TLR2. However, the mechanisms by which this pathogen adheres to and invades the respiratory epithelium are not well understood. The present study aimed to reveal the role of TLR2 in M. catarrhalis adhesion to and invasion into alveolar epithelial cells, using molecular techniques. Pretreatment with the TLR2 inhibitor TLR2-IN-C29 enhanced M. catarrhalis adhesion to A549 cells but reduced its invasion, whereas the agonist Pam3CSK4 reduced both M. catarrhalis adhesion and invasion into A549 cells. Similarly, M. catarrhalis 73-OR strain adhesion and invasion were significantly reduced in TLR2-/- A549 cells. Moreover, the lung clearance rate of the 73-OR strain was significantly higher in TLR2-/- C57/BL6J mice than in wild-type (WT) mice. Histological analysis showed that inflammatory responses were milder in TLR2-/- C57/BL6J mice than in WT mice, which was confirmed by a decrease in cytokine levels in TLR2-/- C57/BL6J mice. Overall, these results indicate that TLR2 promoted M. catarrhalis adhesion and invasion of A549 cells and lung tissues and mediated inflammatory responses in infected lungs. This study provides important insights into the development of potential therapeutic strategies against M. catarrhalis and TLR2-induced inflammatory responses.
Collapse
Affiliation(s)
- Rui Ding
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Jinhan Yu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weixin Ke
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lijun Du
- Department of Clinical Laboratory, Nanchong Central Hospital, the Second Clinical Medical College, North Sichuan Medical College, Nanchong, China
| | - Guixue Cheng
- Department of Clinical Laboratory, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Siqi Hu
- Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
| | - Yingchun Xu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Yali Liu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| |
Collapse
|
2
|
Budroni S, Taccone M, Stella M, Aprea S, Schiavetti F, Bardelli M, Lambert C, Rondini S, Weynants V, Contorni M, Wilkinson TMA, Brazzoli M, Rossi Paccani S. Cytokine Biomarkers of Exacerbations in Sputum From Patients With Chronic Obstructive Pulmonary Disease: A Prospective Cohort Study. J Infect Dis 2024; 230:e1112-e1120. [PMID: 38836471 PMCID: PMC11566228 DOI: 10.1093/infdis/jiae232] [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: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND We determined the relationships between cytokine expression in sputum and clinical data to characterize and understand chronic obstructive pulmonary disease (COPD) exacerbations in people with COPD. METHODS We measured 30 cytokines in 936 sputum samples, collected at stable state and exacerbation visits from 99 participants in the Acute Exacerbation and Respiratory InfectionS in COPD (AERIS) study (ClinicalTrials.gov NCT01360398). We determined their longitudinal expression and examined differential expression based on disease status or exacerbation type. RESULTS Of the cytokines, 17 were suitable for analysis. As for disease states, in exacerbation sputum samples, interleukin (IL) 17A, tumor necrosis factor alpha (TNF-α), IL-1β, and IL-10 were significantly increased compared to stable state sputum samples, but a logistic mixed model could not predict disease state. As for exacerbation types, bacteria-associated exacerbations showed higher expression of IL-17A, TNF-α, IL-1β, and IL-1α. IL-1α, IL-1β, and TNF-α were identified as suitable biomarkers for bacteria-associated exacerbation. Bacteria-associated exacerbations also formed a cluster separate from other exacerbation types in principal component analysis. CONCLUSIONS Measurement of cytokines in sputum from COPD patients could help identify bacteria-associated exacerbations based on increased concentrations of IL-1α, IL-1β, or TNF-α. This finding may provide a point-of-care assessment to distinguish a bacterial exacerbation of COPD from other exacerbation types.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Simona Rondini
- GSK, Siena, Italy
- GSK Vaccines Institute for Global Health, Siena, Italy
| | | | | | - Tom M A Wilkinson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton
- National Institute for Health Research Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | | | | |
Collapse
|
3
|
Wu J, Ma Y, Chen Y. Extracellular vesicles and COPD: foe or friend? J Nanobiotechnology 2023; 21:147. [PMID: 37147634 PMCID: PMC10161449 DOI: 10.1186/s12951-023-01911-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 04/25/2023] [Indexed: 05/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory airway disease characterized by progressive airflow limitation. The complex biological processes of COPD include protein hydrolysis tissue remodeling, innate immune inflammation, disturbed host-pathogen response, abnormal cellular phenotype conversion, and cellular senescence. Extracellular vesicles (EVs) (including apoptotic vesicles, microvesicles and exosomes), are released by almost all cell types and can be found in a variety of body fluids including blood, sputum and urine. EVs are key mediators in cell-cell communication and can be used by using their bioactive substances (DNA, RNA, miRNA, proteins and other metabolites) to enable cells in adjacent and distant tissues to perform a wide variety of functions, which in turn affect the physiological and pathological functions of the body. Thus, EVs is expected to play an important role in the pathogenesis of COPD, which in turn affects its acute exacerbations and may serve as a diagnostic marker for it. Furthermore, recent therapeutic approaches and advances have introduced EVs into the treatment of COPD, such as the modification of EVs into novel drug delivery vehicles. Here, we discuss the role of EVs from cells of different origins in the pathogenesis of COPD and explore their possible use as biomarkers in diagnosis, and finally describe their role in therapy and future prospects for their application. Graphical Abstract.
Collapse
Affiliation(s)
- Jiankang Wu
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Yiming Ma
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| | - Yan Chen
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
4
|
Arnold HK, Hanselmann R, Duke SM, Sharpton TJ, Beechler BR. Chronic clinical signs of upper respiratory tract disease associate with gut and respiratory microbiomes in a cohort of domestic felines. PLoS One 2022; 17:e0268730. [PMID: 36454958 PMCID: PMC9714858 DOI: 10.1371/journal.pone.0268730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 11/18/2022] [Indexed: 12/05/2022] Open
Abstract
Feline upper respiratory tract disease (FURTD), often caused by infections etiologies, is a multifactorial syndrome affecting feline populations worldwide. Because of its highly transmissible nature, infectious FURTD is most prevalent anywhere cats are housed in groups such as animal shelters, and is associated with negative consequences such as decreasing adoption rates, intensifying care costs, and increasing euthanasia rates. Understanding the etiology and pathophysiology of FURTD is thus essential to best mitigate the negative consequences of this disease. Clinical signs of FURTD include acute respiratory disease, with a small fraction of cats developing chronic sequelae. It is thought that nasal mucosal microbiome changes play an active role in the development of acute clinical signs, but it remains unknown if the microbiome may play a role in the development and progression of chronic clinical disease. To address the knowledge gap surrounding how microbiomes link to chronic FURTD, we asked if microbial community structure of upper respiratory and gut microbiomes differed between cats with chronic FURTD signs and clinically normal cats. We selected 8 households with at least one cat exhibiting chronic clinical FURTD, and simultaneously collected samples from cohabitating clinically normal cats. Microbial community structure was assessed via 16S rDNA sequencing of both gut and nasal microbiome communities. Using a previously described ecophylogenetic method, we identified 136 and 89 microbial features within gut and nasal microbiomes respectively that significantly associated with presence of active FURTD clinical signs in cats with a history of chronic signs. Overall, we find that nasal and gut microbial community members associate with the presence of chronic clinical course, but more research is needed to confirm our observations.
Collapse
Affiliation(s)
- Holly Kristin Arnold
- Department of Microbiology, Oregon State University, Corvallis, Oregon, United States of America
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, United States of America
- * E-mail:
| | - Rhea Hanselmann
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, California, United States of America
| | - Sarah M. Duke
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, United States of America
| | - Thomas J. Sharpton
- Department of Microbiology, Oregon State University, Corvallis, Oregon, United States of America
- Department of Statistics, Oregon State University, Corvallis, Oregon, United States of America
| | - Brianna R. Beechler
- Department of Microbiology, Oregon State University, Corvallis, Oregon, United States of America
- Department of Biomedical Sciences, Oregon State University, Corvallis, Oregon, United States of America
| |
Collapse
|
5
|
Sethi S. Is It the Heart or the Lung? Sometimes It Is Both. J Am Heart Assoc 2022; 11:e027112. [DOI: 10.1161/jaha.122.027112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
6
|
Joean O, Welte T. Vaccination and modern management of chronic obstructive pulmonary disease - a narrative review. Expert Rev Respir Med 2022; 16:605-614. [PMID: 35713962 DOI: 10.1080/17476348.2022.2092099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) carries a tremendous societal and individual burden, posing significant challenges for public health systems worldwide due to its high morbidity and mortality. Due to aging and multimorbidity but also in the wake of important progress in deciphering the heterogeneous disease endotypes, an individualized approach to the prevention and management of COPD is necessary. AREAS COVERED This article tackles relevant immunization strategies that are available or still under development with a focus on the latest evidence but also controversies around different regional immunization approaches. Further, we present the crossover between chronic lung inflammation and lung microbiome disturbance as well as its role in delineating COPD endotypes. Moreover, the article attempts to underline endotype-specific treatment approaches. Lastly, we highlight non-pharmacologic prevention and management programs in view of the challenges and opportunities of the COVID-19 era. EXPERT OPINION Despite the remaining challenges, personalized medicine has the potential to offer tailored approaches to prevention and therapy and promises to improve the care of patients living with COPD.
Collapse
Affiliation(s)
- Oana Joean
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease, Member of the German Center for Lung Research, Hannover, Germany
| |
Collapse
|
7
|
Hoult G, Gillespie D, Wilkinson TMA, Thomas M, Francis NA. Biomarkers to guide the use of antibiotics for acute exacerbations of COPD (AECOPD): a systematic review and meta-analysis. BMC Pulm Med 2022; 22:194. [PMID: 35549921 PMCID: PMC9101830 DOI: 10.1186/s12890-022-01958-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 04/18/2022] [Indexed: 11/30/2022] Open
Abstract
Background Antibiotics are frequently prescribed for acute exacerbations of COPD (AECOPD) even though most do not have a bacterial aetiology. Biomarkers may help clinicians target antibiotic use by identifying AECOPD caused by bacterial pathogens. We aimed to summarise current evidence on the diagnostic accuracy of biomarkers for detecting bacterial versus non-bacterial AECOPD. Methods We searched Embase and Medline using a search strategy including terms for COPD, biomarkers and bacterial infection. Data regarding diagnostic accuracy for each biomarker in predicting bacterial cause of exacerbation were extracted and summarised. We used to QUADAS-2 tool to assess risk of bias. Results Of 509 papers identified, 39 papers evaluating 61 biomarkers were eligible for inclusion. Moderate quality evidence was found for associations between serum C-reactive protein (CRP), serum procalcitonin (PCT), sputum interleukin (IL)-8 and sputum tumour necrosis factor alpha (TNF-α), and the presence of bacterial pathogens in the sputum of patients with AECOPD. Having bacterial pathogens was associated with a mean difference (higher) CRP and PCT of 29.44 mg/L and 0.76 ng/mL respectively. There was inconsistent or weak evidence for associations between bacterial AECOPD and higher levels of sputum IL-1β, IL-6, myeloperoxidase (MPO) and neutrophil elastase (NE). We did not find any consistent evidence of diagnostic value for other biomarkers. Conclusions There is moderate evidence from heterogeneous studies that serum CRP and PCT are of value in differentiating bacterial from non-bacterial AECOPD, and little evidence for other biomarkers. Further high-quality research on the role of biomarkers in identifying bacterial exacerbations is needed. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-01958-4.
Collapse
Affiliation(s)
- George Hoult
- Cardiff University School of Medicine, UHW Main Building, Heath Park, Cardiff, CF14 4XN, UK
| | - David Gillespie
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK.,Centre for Trials Research, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Tom M A Wilkinson
- Clinical and Experimental Sciences, Faculty of Medicine, Southampton General Hospital, Southampton University, Mailpoint 810, Level F, South Block, Southampton, SO16 6YD, UK.,NIHR Southampton BRC - Respiratory Theme, Faculty of Medicine, Southampton General Hospital, Southampton University, Mailpoint 810, Level F, South Block, Southampton, SO16 6YD, UK
| | - Mike Thomas
- Primary Care Research Centre, School of Primary Care, Population Sciences and Medical Education, Aldermoor Health Centre, Faculty of Medicine, University of Southampton, Aldermoor Close, Southampton, SO16 5ST, UK
| | - Nick A Francis
- Primary Care Research Centre, School of Primary Care, Population Sciences and Medical Education, Aldermoor Health Centre, Faculty of Medicine, University of Southampton, Aldermoor Close, Southampton, SO16 5ST, UK.
| |
Collapse
|
8
|
Inhaled Antibiotics and Airway Bacterial Decolonization for Patients with Chronic Obstructive Pulmonary Disease: The Rationale and Future. J Transl Int Med 2022; 10:181-184. [PMID: 36776240 PMCID: PMC9901548 DOI: 10.2478/jtim-2022-0005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
9
|
Pázmány P, Soós A, Hegyi P, Dohos D, Kiss S, Szakács Z, Párniczky A, Garami A, Péterfi Z, Molnár Z. Inflammatory Biomarkers Are Inaccurate Indicators of Bacterial Infection on Admission in Patients With Acute Exacerbation of Chronic Obstructive Pulmonary Disease-A Systematic Review and Diagnostic Accuracy Network Meta-Analysis. Front Med (Lausanne) 2021; 8:639794. [PMID: 34869399 PMCID: PMC8636902 DOI: 10.3389/fmed.2021.639794] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 10/21/2021] [Indexed: 01/02/2023] Open
Abstract
Introduction: The value of inflammatory biomarkers in the diagnosis of bacterial infection induced acute exacerbation of chronic obstructive pulmonary disease (AECOPD) is currently unclear. Our objective was to investigate the diagnostic accuracy of on-admission inflammatory biomarkers in differentiating bacterial origin in AECOPD. Methods: Systematic literature search was performed to include cross-sectional studies on AECOPD patients with microbiological culture results as gold standard, and at least one on-admission inflammatory biomarker determined from serum: C-reactive protein (CRP), procalcitonin (PCT), neutrophil/lymphocyte ratio, eosinophil percentage, CD64index; or sputum: neutrophil elastase, tumor necrosis factor alfa, interleukin-1-beta (IL-1b), interleukin-8, sputum color, as index tests. We ranked index tests by superiority indices in a network meta-analysis and also calculated pooled sensitivity and specificity. Results: Altogether, 21 eligible articles reported data on 2,608 AECOPD patients (44% bacterial). Out of the 14 index tests, sputum IL-1b showed the highest diagnostic performance with a pooled sensitivity of 74% (CI: 26-97%) and specificity of 65% (CI: 19-93%). Pooled sensitivity for CRP and PCT were: 67% (CI: 54-77%) and 54% (CI: 39-69%); specificity 62% (CI: 52-71%) and 71% (CI: 59-79%), respectively. Conclusion: Admission inflammatory biomarkers are inaccurate indicators of bacterial infection in AECOPD. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/#myprospero, identifier: 42020161301.
Collapse
Affiliation(s)
- Piroska Pázmány
- Medical School, Institute for Translational Medicine, University of Pécs, Pécs, Hungary
- Department of General Medicine and Pulmonology, Heim Pál National Institute for Pediatrics, Budapest, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
| | - Alexandra Soós
- Medical School, Institute for Translational Medicine, University of Pécs, Pécs, Hungary
| | - Péter Hegyi
- Medical School, Institute for Translational Medicine, University of Pécs, Pécs, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
- János Szentágothai Research Center, University of Pécs, Pécs, Hungary
- Centre for Translational Medicine, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| | - Dóra Dohos
- Medical School, Institute for Translational Medicine, University of Pécs, Pécs, Hungary
| | - Szabolcs Kiss
- Medical School, Institute for Translational Medicine, University of Pécs, Pécs, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
| | - Zsolt Szakács
- Medical School, Institute for Translational Medicine, University of Pécs, Pécs, Hungary
- János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Andrea Párniczky
- Medical School, Institute for Translational Medicine, University of Pécs, Pécs, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
- Department of Gastroenterology, Heim Pál National Institute for Pediatrics, Budapest, Hungary
| | - András Garami
- Medical School, Institute for Translational Medicine, University of Pécs, Pécs, Hungary
| | - Zoltán Péterfi
- Department of Internal Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Zsolt Molnár
- Medical School, Institute for Translational Medicine, University of Pécs, Pécs, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
- Centre for Translational Medicine, Semmelweis University, Faculty of Medicine, Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Medical Faculty, Poznan University for Medical Sciences, Poznan, Poland
- Department of Anaesthesiology and Intensive Therapy, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| |
Collapse
|
10
|
Doğan NÖ, Varol Y, Köktürk N, Aksay E, Alpaydın AÖ, Çorbacıoğlu ŞK, Aksel G, Baha A, Akoğlu H, Karahan S, Şen E, Ergan B, Bayram B, Yılmaz S, Gürgün A, Polatlı M. 2021 Guideline for the Management of COPD Exacerbations: Emergency Medicine Association of Turkey (EMAT) / Turkish Thoracic Society (TTS) Clinical Practice Guideline Task Force. Turk J Emerg Med 2021; 21:137-176. [PMID: 34849428 PMCID: PMC8593424 DOI: 10.4103/2452-2473.329630] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 01/18/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an important public health problem that manifests with exacerbations and causes serious mortality and morbidity in both developed and developing countries. COPD exacerbations usually present to emergency departments, where these patients are diagnosed and treated. Therefore, the Emergency Medicine Association of Turkey and the Turkish Thoracic Society jointly wanted to implement a guideline that evaluates the management of COPD exacerbations according to the current literature and provides evidence-based recommendations. In the management of COPD exacerbations, we aim to support the decision-making process of clinicians dealing with these patients in the emergency setting.
Collapse
Affiliation(s)
- Nurettin Özgür Doğan
- Department of Emergency Medicine, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Yelda Varol
- Department of Pulmonology, Dr. Suat Seren Chest Diseases and Chest Surgery Training and Research Hospital, University of Health Sciences, İzmir, Turkey
| | - Nurdan Köktürk
- Department of Pulmonology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Ersin Aksay
- Department of Emergency Medicine, Dokuz Eylül University, İzmir, Turkey
| | - Aylin Özgen Alpaydın
- Department of Pulmonology, Faculty of Medicine, Dokuz Eylül University, İzmir, Turkey
| | - Şeref Kerem Çorbacıoğlu
- Department of Emergency Medicine, Keçiören Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Gökhan Aksel
- Department of Emergency Medicine, Ümraniye Training and Research Hospital, University of Health Sciences, İstanbul, Turkey
| | - Ayşe Baha
- Department of Pulmonology, Near East University, Nicosia, TRNC
| | - Haldun Akoğlu
- Department of Emergency Medicine, Faculty of Medicine, Marmara University, İstanbul, Turkey
| | - Sevilay Karahan
- Department of Biostatistics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Elif Şen
- Department of Pulmonology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Begüm Ergan
- Department of Pulmonology, Faculty of Medicine, Dokuz Eylül University, İzmir, Turkey
| | - Başak Bayram
- Department of Emergency Medicine, Dokuz Eylül University, İzmir, Turkey
| | - Serkan Yılmaz
- Department of Emergency Medicine, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Alev Gürgün
- Department of Pulmonology, Faculty of Medicine, Ege University, İzmir, Turkey
| | - Mehmet Polatlı
- Department of Pulmonology, Faculty of Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| |
Collapse
|
11
|
MacLeod M, Papi A, Contoli M, Beghé B, Celli BR, Wedzicha JA, Fabbri LM. Chronic obstructive pulmonary disease exacerbation fundamentals: Diagnosis, treatment, prevention and disease impact. Respirology 2021; 26:532-551. [PMID: 33893708 DOI: 10.1111/resp.14041] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In chronic obstructive pulmonary disease (COPD), exacerbations (ECOPD), characterized by an acute deterioration in respiratory symptoms, are fundamental events impacting negatively upon disease progression, comorbidities, wellbeing and mortality. ECOPD also represent the largest component of the socioeconomic burden of COPD. ECOPDs are currently defined as acute worsening of respiratory symptoms that require additional therapy. Definitions that require worsening of dyspnoea and sputum volume/purulence assume that acute infections, especially respiratory viral infections, and/or exposure to pollutants are the main cause of ECOPD. But other factors may contribute to ECOPD, such as the exacerbation of other respiratory diseases and non-respiratory diseases (e.g., heart failure, thromboembolism). The complexity of worsening dyspnoea has suggested a need to improve the definition of ECOPD using objective measurements such as blood counts and C-reactive protein to improve accuracy of diagnosis and a personalized approach to management. There are three time points when we can intervene to improve outcomes: acutely, to attenuate the length and severity of an established exacerbation; in the aftermath, to prevent early recurrence and readmission, which are common, and in the long-term, establishing preventative measures that reduce the risk of future events. Acute management includes interventions such as corticosteroids or antibiotics and measures to support the respiratory system, including non-invasive ventilation (NIV). Current therapies are broad and better understanding of clinical phenotypes and biomarkers may help to establish a more tailored approach, for example in relation to antibiotic prescription. Other unmet needs include effective treatment for viruses, which commonly cause exacerbations. Preventing early recurrence and readmission to hospital is important and the benefits of interventions such as antibiotics or anti-inflammatories in this period are not established. Domiciliary NIV in those patients who are persistently hypercapnic following discharge and pulmonary rehabilitation can have a positive impact. For long-term prevention, inhaled therapy is key. Dual bronchodilators reduce exacerbation frequency but in patients with continuing exacerbations, triple therapy should be considered, especially if blood eosinophils are elevated. Other options include phosphodiesterase inhibitors and macrolide antibiotics. ECOPD are a key component of the assessment of COPD severity and future outcomes (quality of life, hospitalisations, health care resource utilization, mortality) and are a central component in pharmacological management decisions. Targeted therapies directed towards specific pathways of inflammation are being explored in exacerbation prevention, and this is a promising avenue for future research.
Collapse
Affiliation(s)
- Mairi MacLeod
- National Heart and Lung Institute, Imperial College, London, UK
| | - Alberto Papi
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Marco Contoli
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Bianca Beghé
- Department of Respiratory Diseases, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | - Leonardo M Fabbri
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy.,Department of Respiratory Diseases, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
12
|
Bhat TA, Kalathil SG, Bogner PN, Lehmann PV, Thatcher TH, Sime PJ, Thanavala Y. AT-RvD1 Mitigates Secondhand Smoke-Exacerbated Pulmonary Inflammation and Restores Secondhand Smoke-Suppressed Antibacterial Immunity. THE JOURNAL OF IMMUNOLOGY 2021; 206:1348-1360. [PMID: 33558371 DOI: 10.4049/jimmunol.2001228] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/09/2021] [Indexed: 02/07/2023]
Abstract
Cigarette smoke is a potent proinflammatory trigger contributing to acute lung injury and the development of chronic lung diseases via mechanisms that include the impairment of inflammation resolution. We have previously demonstrated that secondhand smoke (SHS) exposure exacerbates bacterial infection-induced pulmonary inflammation and suppresses immune responses. It is now recognized that resolution of inflammation is a bioactive process mediated by lipid-derived specialized proresolving mediators that counterregulate proinflammatory signaling and promote resolution pathways. We therefore hypothesized that proresolving mediators could reduce the burden of inflammation due to chronic lung infection following SHS exposure and restore normal immune responses to respiratory pathogens. To address this question, we exposed mice to SHS followed by chronic infection with nontypeable Haemophilus influenzae (NTHI). Some groups of mice were treated with aspirin-triggered resolvin D1 (AT-RvD1) during the latter half of the smoke exposure period or during a period of smoking cessation and before infection. Treatment with AT-RvD1 markedly reduced the recruitment of neutrophils, macrophages, and T cells in lung tissue and bronchoalveolar lavage and levels of proinflammatory cytokines in the bronchoalveolar lavage. Additionally, treatment with AT-RvD1 improved Ab titers against the NTHI outer membrane lipoprotein Ag P6 following infection. Furthermore, treatment with AT-RvD1 prior to classically adjuvanted immunization with P6 increased Ag-specific Ab titers, resulting in rapid clearance of NTHI from the lungs after acute challenge. Collectively, we have demonstrated that AT-RvD1 potently reverses the detrimental effects of SHS on pulmonary inflammation and immunity and thus could be beneficial in reducing lung injury associated with smoke exposure and infection.
Collapse
Affiliation(s)
- Tariq A Bhat
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | | | - Paul N Bogner
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | | | - Thomas H Thatcher
- Department of Medicine, University of Rochester, Rochester, NY 14620; and.,Department of Environmental Medicine, University of Rochester, Rochester, NY 14620
| | - Patricia J Sime
- Department of Medicine, University of Rochester, Rochester, NY 14620; and.,Department of Environmental Medicine, University of Rochester, Rochester, NY 14620
| | - Yasmin Thanavala
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263;
| |
Collapse
|
13
|
Smith DJ, Ellis PR, Turner AM. Exacerbations of Lung Disease in Alpha-1 Antitrypsin Deficiency. CHRONIC OBSTRUCTIVE PULMONARY DISEASES (MIAMI, FLA.) 2021; 8:162-176. [PMID: 33238089 PMCID: PMC8047608 DOI: 10.15326/jcopdf.2020.0173] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/18/2020] [Indexed: 01/13/2023]
Abstract
Alpha-1 antitrypsin deficiency (AATD) is an important risk factor for development of chronic obstructive pulmonary disease (COPD). Patients with AATD classically develop a different pattern of lung disease from those with usual COPD, decline faster and exhibit a range of differences in pathogenesis, all of which may be relevant to phenotype and/or impact of exacerbations. There are a number of definitions of exacerbation, with the main features being worsening of symptoms over at least 2 days, which may be associated with a change in treatment. In this article we review the literature surrounding exacerbations in AATD, focusing, in particular, on ways in which they may differ from such events in usual COPD, and the potential impact on clinical management.
Collapse
Affiliation(s)
- Daniel J. Smith
- Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
| | - Paul R. Ellis
- Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
| | - Alice M. Turner
- Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
- University Hospitals Birmingham, United Kingdom
| |
Collapse
|
14
|
Dong T, Santos S, Yang Z, Yang S, Kirkhus NE. Sputum and salivary protein biomarkers and point-of-care biosensors for the management of COPD. Analyst 2020; 145:1583-1604. [PMID: 31915768 DOI: 10.1039/c9an01704f] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) has become one of the most fatal diseases of the century considering mortality and morbidity levels worldwide. This disease is an inflammatory response to environmental stress and tobacco smoking. Although spirometry is the gold-standard diagnostic test administrated in primary and secondary care, it often exhibits low accuracy in cases of predicting disease worsening and possible bias due to the operator, patient, and conditions. Recent developments in proteomics research suggest that the presence of protein biomarkers can aid in the accurate diagnosis and prediction of disease outcomes. This review presents the cutting-edge research progress in the area of protein biomarkers towards the management of COPD. The literature review was confined to protein biomarkers in saliva and sputum because testing these bodily fluids shows great promise for point-of-care (POC) testing due to its practicality, non-invasiveness and inexpensive handling and sampling. Although it is conclusive that more studies on sputum and saliva are needed, this review studies the promising clinical value of interleukin (IL)-6 and IL-8, matrix metalloproteinase (MMP)-8 and MMP-9, C-reactive protein (CRP), tumor necrosis factor-alpha (TNF-α), and neutrophil elastase (NE). Following the critical analysis of salivary and sputum biomarkers, the recent development of POC biosensors for the multiplexed detection of biomarkers is also reported. Overall, the review aims to explore the possibility for the future development of POC sensors for chronic lung disease management utilizing clinically relevant biomarkers in saliva and sputum.
Collapse
Affiliation(s)
- Tao Dong
- Chongqing Key Laboratory of Micro-Nano Systems and Smart Transduction, Collaborative Innovation Center on Micro-Nano Transduction and Intelligent Eco-Internet of Things, Chongqing Key Laboratory of Colleges and Universities on Micro-Nano Systems Technology and Smart Transducing, National Research Base of Intelligent Manufacturing Service, Chongqing Technology and Business University, Nan'an District, Chongqing 400067, China and Department of Microsystems (IMS), Faculty of Technology, Natural Sciences and Maritime Sciences, University of South-Eastern Norway, Postboks 235, 3603 Kongsberg, Norway.
| | - Simão Santos
- Department of Microsystems (IMS), Faculty of Technology, Natural Sciences and Maritime Sciences, University of South-Eastern Norway, Postboks 235, 3603 Kongsberg, Norway.
| | - Zhaochu Yang
- Chongqing Key Laboratory of Micro-Nano Systems and Smart Transduction, Collaborative Innovation Center on Micro-Nano Transduction and Intelligent Eco-Internet of Things, Chongqing Key Laboratory of Colleges and Universities on Micro-Nano Systems Technology and Smart Transducing, National Research Base of Intelligent Manufacturing Service, Chongqing Technology and Business University, Nan'an District, Chongqing 400067, China
| | - Shuai Yang
- Chongqing Key Laboratory of Micro-Nano Systems and Smart Transduction, Collaborative Innovation Center on Micro-Nano Transduction and Intelligent Eco-Internet of Things, Chongqing Key Laboratory of Colleges and Universities on Micro-Nano Systems Technology and Smart Transducing, National Research Base of Intelligent Manufacturing Service, Chongqing Technology and Business University, Nan'an District, Chongqing 400067, China
| | - Niels E Kirkhus
- Horten Kommune - Kommuneoverlege, Enhetsleder Legetjenester, Vestfold, Norway
| |
Collapse
|
15
|
Zhao D, Abbasi A, Rossiter HB, Su X, Liu H, Pi Y, Sang L, Zhong W, Yang Q, Guo X, Zhou Y, Li T, Casaburi R, Zhang N. Serum Amyloid A in Stable COPD Patients is Associated with the Frequent Exacerbator Phenotype. Int J Chron Obstruct Pulmon Dis 2020; 15:2379-2388. [PMID: 33061355 PMCID: PMC7535123 DOI: 10.2147/copd.s266844] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/31/2020] [Indexed: 11/24/2022] Open
Abstract
Background We sought to determine whether circulating inflammatory biomarkers were associated with the frequent exacerbator phenotype in stable COPD patients ie, those with two or more exacerbations in the previous year. Methods Eighty-eight stable, severe, COPD patients (4 females) were assessed for exacerbation frequency, pulmonary function, fraction of expired nitric oxide (FENO); inflammatory variables were measured in venous blood. Logistic regression assessed associations between the frequent exacerbator phenotype and systemic inflammation. Results Compared with infrequent exacerbators, frequent exacerbators (n=10; 11.4%) had greater serum concentration (median (25th-75th quartile)) of serum amyloid A (SAA; 134 (84–178) vs 71 (38–116) ng/mL; P=0.024), surfactant protein D (SP-D; 15.6 (9.0–19.3) vs 8.5 (3.6–14.9) ng/mL; P=0.049) and interleukin-4 (IL-4; 0.12 (0.08–1.44) vs 0.03 (0.01–0.10) pg/mL; P=0.001). SAA, SP-D and IL-4 were not significantly correlated with FEV1%predicted or FVC %predicted. After adjusting for sex, age, BMI, FEV1/FVC and smoking pack-years, only SAA remained independently associated with the frequent exacerbator phenotype (OR 1.49[1.09–2.04]; P=0.012). The odds of being a frequent exacerbator was 18-times greater in the highest SAA quartile (≥124.1 ng/mL) than the lowest SAA quartile (≤44.1 ng/mL) (OR 18.34[1.30–258.81]; P=0.031), and there was a significant positive trend of increasing OR with increasing SAA quartile (P=0.008). For SAA, the area under the receiver operating characteristic curve was 0.721 for identification of frequent exacerbators; an SAA cut-off of 87.0 ng/mL yielded an 80% sensitivity and 61.5% specificity. Conclusion In stable COPD patients, SAA was independently associated with the frequent exacerbator phenotype, suggesting that SAA may be a useful serum biomarker to inform progression or management in COPD.
Collapse
Affiliation(s)
- Dongxing Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China.,Rehabilitation Clinical Trials Center, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Asghar Abbasi
- Rehabilitation Clinical Trials Center, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Harry B Rossiter
- Rehabilitation Clinical Trials Center, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA.,Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Xiaofen Su
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Heng Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Yuhong Pi
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Li Sang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Weiyong Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Qifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Xiongtian Guo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Yanyan Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Tianyang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Richard Casaburi
- Rehabilitation Clinical Trials Center, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Nuofu Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, People's Republic of China
| |
Collapse
|
16
|
Sethi S, Aaron SD. Antibiotic Retreatment for Acute Exacerbations of Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2020; 202:481-482. [PMID: 32282222 PMCID: PMC7427405 DOI: 10.1164/rccm.202004-0896ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Affiliation(s)
- Sanjay Sethi
- Jacobs School of MedicineUniversity of BuffaloBuffalo, New Yorkand
| | - Shawn D Aaron
- The Ottawa Hospital Research InstituteUniversity of OttawaOttawa, Ontario, Canada
| |
Collapse
|
17
|
Mathioudakis AG, Janssens W, Sivapalan P, Singanayagam A, Dransfield MT, Jensen JUS, Vestbo J. Acute exacerbations of chronic obstructive pulmonary disease: in search of diagnostic biomarkers and treatable traits. Thorax 2020; 75:520-527. [PMID: 32217784 PMCID: PMC7279206 DOI: 10.1136/thoraxjnl-2019-214484] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/21/2020] [Accepted: 03/01/2020] [Indexed: 12/12/2022]
Abstract
Acute exacerbations of chronic obstructive pulmonary disease (COPD) are associated with a significant mortality, health and economic burden. Their diagnosis, assessment and management remain suboptimal and unchanged for decades. Recent clinical and translational studies revealed that the significant heterogeneity in mechanisms and outcomes of exacerbations could be resolved by grouping them etiologically. This is anticipated to lead to a better understanding of the biological processes that underlie each type of exacerbation and to allow the introduction of precision medicine interventions that could improve outcomes. This review summarises novel data on the diagnosis, phenotyping, targeted treatment and prevention of COPD exacerbations.
Collapse
Affiliation(s)
- Alexander G Mathioudakis
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK.,North West Lung Centre, University Hospital of South Manchester NHS Foundation Trust, Manchester, UK
| | - Wim Janssens
- Respiratory Division, Department of Clinical and Experimental Medicine, University Hospital Leuven & KU Leuven, Leuven, Belgium
| | - Pradeesh Sivapalan
- Section of Respiratory Medicine, Department of Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Aran Singanayagam
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Mark T Dransfield
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama School of Medicine, Birmingham, Alabama, USA
| | - Jens-Ulrik Stæhr Jensen
- Section of Respiratory Medicine, Department of Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.,Department of Clinical Medicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark.,PERSIMUNE&CHIP: Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Jørgen Vestbo
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK .,North West Lung Centre, University Hospital of South Manchester NHS Foundation Trust, Manchester, UK
| |
Collapse
|
18
|
López-López N, Euba B, Hill J, Dhouib R, Caballero L, Leiva J, Hosmer J, Cuesta S, Ramos-Vivas J, Díez-Martínez R, Schirra HJ, Blank LM, Kappler U, Garmendia J. Haemophilus influenzae Glucose Catabolism Leading to Production of the Immunometabolite Acetate Has a Key Contribution to the Host Airway-Pathogen Interplay. ACS Infect Dis 2020; 6:406-421. [PMID: 31933358 DOI: 10.1021/acsinfecdis.9b00359] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by abnormal inflammatory responses and impaired airway immunity, which provides an opportunistic platform for nontypeable Haemophilus influenzae (NTHi) infection. Clinical evidence supports that the COPD airways present increased concentrations of glucose, which may facilitate proliferation of pathogenic bacteria able to use glucose as a carbon source. NTHi metabolizes glucose through respiration-assisted fermentation, leading to the excretion of acetate, formate, and succinate. We hypothesized that such specialized glucose catabolism may be a pathoadaptive trait playing a pivotal role in the NTHi airway infection. To find out whether this is true, we engineered and characterized bacterial mutant strains impaired to produce acetate, formate, or succinate by inactivating the ackA, pflA, and frdA genes, respectively. While the inactivation of the pflA and frdA genes only had minimal physiological effects, the inactivation of the ackA gene affected acetate production and led to reduced bacterial growth, production of lactate under low oxygen tension, and bacterial attenuation in vivo. Moreover, bacterially produced acetate was able to stimulate the expression of inflammatory genes by cultured airway epithelial cells. These results back the notion that the COPD lung supports NTHi growth on glucose, enabling production of fermentative end products acting as immunometabolites at the site of infection. Thus, glucose catabolism may contribute not only to NTHi growth but also to bacterially driven airway inflammation. This information has important implications for developing nonantibiotic antimicrobials, given that airway glucose homeostasis modifying drugs could help prevent microbial infections associated with chronic lung disease.
Collapse
Affiliation(s)
| | - Begoña Euba
- Instituto de Agrobiotecnologı́a, CSIC-Gobierno Navarra, 31192 Mutilva, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Julian Hill
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Rabeb Dhouib
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Lucı́a Caballero
- Instituto de Agrobiotecnologı́a, CSIC-Gobierno Navarra, 31192 Mutilva, Spain
| | - José Leiva
- Servicio de Microbiologı́a, Clı́nica Universidad de Navarra, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Jennifer Hosmer
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Sergio Cuesta
- Instituto de Agrobiotecnologı́a, CSIC-Gobierno Navarra, 31192 Mutilva, Spain
| | - José Ramos-Vivas
- Servicio Microbiologı́a, Hospital Universitario Marqués de Valdecilla and Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain
- Red Española de Investigación en Patologı́a Infecciosa (REIPI), ISCIII, Madrid, Spain
| | - Roberto Díez-Martínez
- Telum Therapeutics, Centro Europeo de Empresas e Innovación de Navarra (CEIN), 31110 Noáin, Spain
| | - Horst Joachim Schirra
- Centre for Advanced Imaging, The University of Queensland, 4072 St Lucia, Queensland, Australia
| | - Lars M. Blank
- Institute of Applied Biotechnology, RWTH Aachen University, 52074 Aachen, Germany
| | - Ulrike Kappler
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Junkal Garmendia
- Instituto de Agrobiotecnologı́a, CSIC-Gobierno Navarra, 31192 Mutilva, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
19
|
Procalcitonin and antibiotics in moderate-severe acute exacerbation of chronic obstructive pulmonary disease: to use or not to use. Curr Opin Pulm Med 2020; 25:150-157. [PMID: 30418243 DOI: 10.1097/mcp.0000000000000548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW Chronic obstructive pulmonary disease (COPD) is a leading cause of morbidity and mortality worldwide. Acute exacerbations of COPD (AECOPD) are major driver for healthcare utilization with each exacerbation begetting the next exacerbation. It is, therefore, important to treat each episode effectively to prevent the next. However, this can be challenging as AECOPD result from complex interactions between host, environment and infective agents. The benefits of starting antibiotics in AECOPD, which are not life-threatening (e.g. not requiring mechanical ventilation) or not complicated by pneumonia remain controversial. RECENT FINDINGS The use of procalcitonin to guide antibiotic therapy in AECOPD has gained interest in recent years. The main advantage of this approach is a safe reduction in antibiotic use in a large group of patients, which may potentially translate to several other benefits. These include reduced antibiotic-related side-effects, reduced risk of developing antibiotic-resistant organisms and cost savings. This approach is associated with no increase in mortality or morbidity such as treatment failure, re-admission, admission to ICU. SUMMARY Procalcitonin-guided antibiotic therapy in AECOPD is a promising and safe approach, which may be ready for the prime time.
Collapse
|
20
|
Ogan N, Gunay E, Ekici B, Baha A, Gulensoy ES, Akpinar EE, Yuksel A. Morphological overview of cardiovascular comorbidities in chronic obstructive pulmonary disease: Frank's sign. Heart Lung 2020; 49:331-335. [PMID: 32061412 DOI: 10.1016/j.hrtlng.2020.01.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Cardiovascular diseases are the most common and important comorbidities in patients with chronic obstructive pulmonary disease (COPD). Literature indicates that there may be a relationship between diagonal earlobe crease (DELC) and coronary artery disease (CAD). Accordingly, the present study aimed to assess the relationship with DELC and cardiac comorbidities in patients with COPD during routine physical examination. MATERIALS AND METHODS In this prospective cohort study, we evaluated the demographic data, pulmonary function test (PFT) results, lipid profile, oxygen saturation, and the presence of DELC in patients with COPD and control subjects. RESULTS DELC was diagnosed in 155 (62%) of COPD patients and these patients had a higher prevalence of CAD (p = 0.044). Moreover, DELC was diagnosed in 135 men (68.5%) and 20 (37.7%) women in the COPD group (p<0.001) and in 39 (48.8%) men and 14 (56.0%) women in the control group (p = 0.527). On the other hand, CAD was diagnosed in 18% of patients with early-stage COPD (n = 104) and in 30.8% of patients with late-stage COPD (n = 146) (p = 0.041). The sensitivity and specificity of DELC positivity in predicting CAD were 80.65% and 44.15% in COPD patients, respectively. CONCLUSION The presence of cardiac comorbidities in COPD patients may play a vital role in the severity of the disease, exacerbations, and may also reduce the treatment response. Accordingly, an earlobe examination of patients with COPD may be useful in predicting the presence of cardiac comorbidities with high sensitivity.
Collapse
Affiliation(s)
- Nalan Ogan
- Ufuk University Faculty of Medicine, Department of Chest Diseases, Ankara, Turkey.
| | - Ersin Gunay
- Afyon Kocatepe Faculty of Medicine, Department of Chest Diseases, Afyonkarahisar, Turkey
| | - Berkay Ekici
- Ufuk University Faculty of Medicine, Department of Cardiology, Ankara, Turkey
| | - Ayse Baha
- Kyrenia Akcicek National Hospital, Department of Chest Diseases, Kyrenia, Cyprus
| | - Esen Sayin Gulensoy
- Ufuk University Faculty of Medicine, Department of Chest Diseases, Ankara, Turkey
| | - E Eylem Akpinar
- Ufuk University Faculty of Medicine, Department of Chest Diseases, Ankara, Turkey
| | - Aycan Yuksel
- Ufuk University Faculty of Medicine, Department of Chest Diseases, Ankara, Turkey
| |
Collapse
|
21
|
Rodríguez-Arce I, Al-Jubair T, Euba B, Fernández-Calvet A, Gil-Campillo C, Martí S, Törnroth-Horsefield S, Riesbeck K, Garmendia J. Moonlighting of Haemophilus influenzae heme acquisition systems contributes to the host airway-pathogen interplay in a coordinated manner. Virulence 2019; 10:315-333. [PMID: 30973092 PMCID: PMC6550540 DOI: 10.1080/21505594.2019.1596506] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/18/2019] [Accepted: 03/12/2019] [Indexed: 01/30/2023] Open
Abstract
Nutrient iron sequestration is the most significant form of nutritional immunity and causes bacterial pathogens to evolve strategies of host iron scavenging. Cigarette smoking contains iron particulates altering lung and systemic iron homeostasis, which may enhance colonization in the lungs of patients suffering chronic obstructive pulmonary disease (COPD) by opportunistic pathogens such as nontypeable. NTHi is a heme auxotroph, and the NTHi genome contains multiple heme acquisition systems whose role in pulmonary infection requires a global understanding. In this study, we determined the relative contribution to NTHi airway infection of the four heme-acquisition systems HxuCBA, PE, SapABCDFZ, and HbpA-DppBCDF that are located at the bacterial outer membrane or the periplasm. Our computational studies provided plausible 3D models for HbpA, SapA, PE, and HxuA interactions with heme. Generation and characterization of single mutants in the hxuCBA, hpe, sapA, and hbpA genes provided evidence for participation in heme binding-storage and inter-bacterial donation. The hxuA, sapA, hbpA, and hpe genes showed differential expression and responded to heme. Moreover, HxuCBA, PE, SapABCDFZ, and HbpA-DppBCDF presented moonlighting properties related to resistance to antimicrobial peptides or glutathione import, together likely contributing to the NTHi-host airway interplay, as observed upon cultured airway epithelia and in vivo lung infection. The observed multi-functionality was shown to be system-specific, thus limiting redundancy. Together, we provide evidence for heme uptake systems as bacterial factors that act in a coordinated and multi-functional manner to subvert nutritional- and other sources of host innate immunity during NTHi airway infection.
Collapse
Affiliation(s)
| | - Tamim Al-Jubair
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Begoña Euba
- Instituto de Agrobiotecnología, CSIC-Gobierno, Navarra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | | | | | - Sara Martí
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Departamento Microbiología, Hospital Universitari Bellvitge, University of Barcelona, IDIBELL, Barcelona, Spain
| | - Susanna Törnroth-Horsefield
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, Lund, Sweden
| | - Kristian Riesbeck
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Junkal Garmendia
- Instituto de Agrobiotecnología, CSIC-Gobierno, Navarra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|
22
|
Moghoofei M, Azimzadeh Jamalkandi S, Moein M, Salimian J, Ahmadi A. Bacterial infections in acute exacerbation of chronic obstructive pulmonary disease: a systematic review and meta-analysis. Infection 2019; 48:19-35. [PMID: 31482316 DOI: 10.1007/s15010-019-01350-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 08/16/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Due to the importance of Chronic obstructive pulmonary disease (COPD) as the fourth cause of mortality worldwide and the lack of studies evaluating the prevalence of bacterial infections in disease exacerbation, this systematic review and meta-analysis was performed to determine the prevalence rate of bacterial infections in COPD patients. METHODS PubMed, ISI Web of Science, and Scopus databases were systematically searched for population-based prevalence studies (1980-2018). MeSH terms for "Bacterial infections" and "AECOPD" were used as search keywords. The selected studies were filtered according to the inclusion and exclusion criteria. Fixed and random-effects models were used for estimation of summary effect sizes. Between-study heterogeneity, as well as publication bias, were calculated. RESULTS Finally, 118 out of 31,440 studies were selected. The overall estimation of the prevalence of bacterial infection was 49.59% [95% confidence interval (CI) 0.4418-0.55]. The heterogeneity in estimating the pooled prevalence of bacterial infections was shown in the studies (Cochran Q test: 6615, P < 0.0001, I2 = 98.23%). In addition, S. pneumoniae, H. influenzae, M. catarrhalis, A. baumannii, P. aeruginosa, and S. aureus were the most prevalent reported bacteria. CONCLUSIONS Our results as the first meta-analysis for the issue demonstrated that bacterial infections are an important risk factor for AECOPD. Further studies must be performed for understanding the exact role of bacterial agents in AECOPD and help physicians for more applicable preventive and therapeutic measurements.
Collapse
Affiliation(s)
- Mohsen Moghoofei
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sadegh Azimzadeh Jamalkandi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Masood Moein
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Salimian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Ahmadi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Nishimoto Y, Yasuda H, Masuko K, Usui Y, Ueda K, Kimura G, Ito K, Kizawa Y. [The Involvement of Src in Airway Inflammation Induced by Repeated Exposure to Lipopolysaccharide in Mice]. YAKUGAKU ZASSHI 2019; 139:1211-1217. [PMID: 31189750 DOI: 10.1248/yakushi.19-00086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Corticosteroid insensitive airway inflammation is one of major barrier to effective managements of chronic airway diseases, such as chronic obstructive pulmonary disease (COPD) and severe asthma. The role of nonreceptor tyrosine kinase Src is important in airway inflammation in mice models of atopic asthma and COPD. Thus, in this study, we determined the effects of Src inhibitor, dasatinib, on airway inflammation induced by repeated intranasal exposure to lipopolysaccharide (LPS). Male mice (A/J strain, 5 weeks old) were intranasally exposed to LPS twice daily for 3 d, and dasatinib was intranasally treated 2 h prior to each LPS exposure. A day after the last stimulation, lungs and bronchoalveolar lavage fluid (BALF) were collected. Dasatinib attenuated the accumulation of inflammatory cells in lungs, and the increase in the numbers of inflammatory cells and the accumulation of cytokines/chemokines in BALF in a dose dependent manner. Therefore, this study suggested that targeting the Src can provide a new therapeutic approach for corticosteroid insensitive pulmonary diseases.
Collapse
Affiliation(s)
- Yuki Nishimoto
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University
| | - Hironobu Yasuda
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University
| | - Keita Masuko
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University
| | - Yoshito Usui
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University
| | - Keitaro Ueda
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University
| | - Genki Kimura
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University
| | - Kazuhiro Ito
- Airway Disease Section, National Heart and Lung Institute, Imperial College London
| | - Yasuo Kizawa
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University
| |
Collapse
|
24
|
Nishimoto Y, Iwamoto I, Suzuki A, Ueda K, Kimura G, Ito K, Kizawa Y. [TNF-α Decreased Corticosteroid Responsiveness in Mice Models of Airway Inflammation Induced by Double Strand RNA and/or Tobacco Smoke Exposure]. YAKUGAKU ZASSHI 2019; 139:955-961. [PMID: 30944262 DOI: 10.1248/yakushi.18-00230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Reduction of corticosteroid responsiveness is one of the important clinical problems in chronic obstructive pulmonary disease (COPD). In this study, we determined the effects of neutralization of tumor necrosis factor-α (TNF-α) on corticosteroid insensitivity in mice models of airway inflammation induced by poly(I:C) and tobacco smoke (TS) exposure. Mice (male A/J strain, 5 weeks old) were exposed to TS for 10 d, or TS for 11 d and poly(I:C) for 3 d. Anti-TNF-α antibody was intranasally treated once every other day 2 h before the TS exposure, and dexamethasone 21-phosphate (DEX) was treated 30 min before the TS or poly(I:C) exposure. On the next day of the last stimulation, mice were sacrificed. The combination treatment of DEX and TNF-α neutralization was significantly attenuated the increase of the numbers of inflammatory cells in BALF and the TNF-α mRNA expression levels induced by TS and poly(I:C) exposure, even though TNF-α neutralization alone had little effect. These data indicated that neutralization of TNF-α restores corticosteroid responsiveness. Therefore, our study suggests that targeting TNF-α signaling pathway provides a new therapeutic approach to corticosteroid refractory airway diseases.
Collapse
Affiliation(s)
- Yuki Nishimoto
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University
| | - Ippei Iwamoto
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University
| | - Ayaka Suzuki
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University
| | - Keitaro Ueda
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University
| | - Genki Kimura
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University
| | - Kazuhiro Ito
- Airway Disease Section, National Heart and Lung Institute, Imperial College London
| | - Yasuo Kizawa
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University
| |
Collapse
|
25
|
Moon JY, Leitao Filho FS, Shahangian K, Takiguchi H, Sin DD. Blood and sputum protein biomarkers for chronic obstructive pulmonary disease (COPD). Expert Rev Proteomics 2018; 15:923-935. [PMID: 30362838 DOI: 10.1080/14789450.2018.1539670] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) is a heterogeneous set of disorders, characterized by airflow limitation, and reduced lung function. Despite increasing knowledge regarding its pathophysiology, there has been limited advancement in therapeutics and the current treatment strategy is symptom management and prevention of exacerbations. Areas covered: Biomarkers represent important tools for the implementation of precision medicine. As fundamental molecules of all living processes, proteins could provide crucial information about how genes interact with the environment. Proteomics studies could act as important tools in identifying reliable biomarkers to enable a more precise therapeutic approach. In this review, we will explore the most promising blood and sputum protein biomarkers in COPD that have been consistently reported in the literature. Expert commentary: Given the complexity of COPD, no single protein biomarker has been able to improve the outcomes of COPD patients. According to preliminary studies, precision medicine in COPD will likely require a combination of different proteins in a biomarker panel for clinical translation. With advancements in current mass spectrometry techniques, an enhancement in the identification of new biomarkers will be observed, and improvements in sequence database search can fill in potential gaps between biomarker discovery and patient care.
Collapse
Affiliation(s)
- Ji-Yong Moon
- a Centre for Heart and Lung Innovation , St. Paul's Hospital & University of British Columbia , Vancouver , Canada.,b Department of Internal Medicine , Hanyang University College of Medicine , Seoul , Korea
| | - Fernando Sergio Leitao Filho
- a Centre for Heart and Lung Innovation , St. Paul's Hospital & University of British Columbia , Vancouver , Canada.,c Division of Pulmonary Medicine, Department of Medicine , Tokai University School of Medicine , Kanagawa , Japan
| | - Kimeya Shahangian
- a Centre for Heart and Lung Innovation , St. Paul's Hospital & University of British Columbia , Vancouver , Canada
| | - Hiroto Takiguchi
- a Centre for Heart and Lung Innovation , St. Paul's Hospital & University of British Columbia , Vancouver , Canada.,d Division of Respiratory Medicine (Department of Medicine) , University of British Columbia , Vancouver , Canada
| | - Don D Sin
- a Centre for Heart and Lung Innovation , St. Paul's Hospital & University of British Columbia , Vancouver , Canada.,d Division of Respiratory Medicine (Department of Medicine) , University of British Columbia , Vancouver , Canada
| |
Collapse
|
26
|
Williams NP, Ostridge K, Devaster JM, Kim V, Coombs NA, Bourne S, Clarke SC, Harden S, Abbas A, Aris E, Lambert C, Tuck A, Williams A, Wootton S, Staples KJ, Wilkinson TMA. Impact of radiologically stratified exacerbations: insights into pneumonia aetiology in COPD. Respir Res 2018; 19:143. [PMID: 30055608 PMCID: PMC6064093 DOI: 10.1186/s12931-018-0842-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/10/2018] [Indexed: 02/03/2023] Open
Abstract
Background COPD patients have increased risk of developing pneumonia, which is associated with poor outcomes. It can be symptomatically indistinguishable from exacerbations, making diagnosis challenging. Studies of pneumonia in COPD have focused on hospitalised patients and are not representative of the ambulant COPD population. Therefore, we sought to determine the incidence and aetiology of acute exacerbation events with evidence of pneumonic radiographic infiltrates in an outpatient COPD cohort. Methods One hundred twenty-seven patients with moderate to very severe COPD aged 42–85 years underwent blood and sputum sampling over one year, at monthly stable visits and within 72 h of exacerbation symptom onset. 343 exacerbations with chest radiographs were included. Results 20.1% of exacerbations had pneumonic infiltrates. Presence of infiltrate was highly seasonal (Winter vs summer OR 3.056, p = 0.027). In paired analyses these exacerbation events had greater increases in systemic inflammation. Bacterial detection rate was higher in the pneumonic group, with Haemophilus influenzae the most common bacteria in both radiological groups. Viral detection and sputum microbiota did not differ with chest radiograph appearance. Conclusions In an outpatient COPD cohort, pneumonic infiltrates at exacerbation were common, and associated with more intense inflammation. Bacterial pathogen detection and lung microbiota were not distinct, suggesting that exacerbations and pneumonia in COPD share common infectious triggers and represent a continuum of severity rather than distinct aetiological events. Trial registration Trial registration Number: NCT01360398. Electronic supplementary material The online version of this article (10.1186/s12931-018-0842-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nicholas P Williams
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK. .,Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Southampton, UK.
| | - Kristoffer Ostridge
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK.,Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Southampton, UK
| | | | - Viktoriya Kim
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK.,Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Southampton, UK
| | - Ngaire A Coombs
- Primary Care and Population Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Simon Bourne
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK.,Present address: Portsmouth Hospitals NHS Trust, Queen Alexandra Hospital, Portsmouth, UK
| | - Stuart C Clarke
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK.,Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | - Stephen Harden
- Department of Radiology, University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Tremona Road, Southampton, UK
| | - Ausami Abbas
- Department of Radiology, University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Tremona Road, Southampton, UK
| | | | | | - Andrew Tuck
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Anthony Williams
- Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | - Stephen Wootton
- Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Karl J Staples
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK.,Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | - Tom M A Wilkinson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK.,Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Southampton, UK.,Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | | |
Collapse
|
27
|
Yang M, Kohler M, Heyder T, Forsslund H, Garberg HK, Karimi R, Grunewald J, Berven FS, Nyrén S, Magnus Sköld C, Wheelock ÅM. Proteomic profiling of lung immune cells reveals dysregulation of phagocytotic pathways in female-dominated molecular COPD phenotype. Respir Res 2018. [PMID: 29514663 PMCID: PMC5842633 DOI: 10.1186/s12931-017-0699-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Smoking is the main risk factor for chronic obstructive pulmonary disease (COPD). Women with COPD who smoke experienced a higher risk of hospitalization and worse decline of lung function. Yet the mechanisms of these gender-related differences in clinical presentations in COPD remain unknown. The aim of our study is to identify proteins and molecular pathways associated with COPD pathogenesis, with emphasis on elucidating molecular gender difference. Method We employed shotgun isobaric tags for relative and absolute quantitation (iTRAQ) proteome analyses of bronchoalveolar lavage (BAL) cells from smokers with normal lung function (n = 25) and early stage COPD patients (n = 18). Multivariate modeling, pathway enrichment analysis, and correlation with clinical characteristics were performed to identify specific proteins and pathways of interest. Results More pronounced alterations both at the protein- and pathway- levels were observed in female COPD patients, involving dysregulation of the FcγR-mediated phagocytosis-lysosomal axis and increase in oxidative stress. Alterations in pathways of the phagocytosis-lysosomal axis associated with a female-dominated COPD phenotype correlated well with specific clinical features: FcγR-mediated phagocytosis correlated with FEV1/FVC, the lysosomal pathway correlated with CT < −950 Hounsfield Units (HU), and regulation of actin cytoskeleton correlated with FEV1 and FEV1/FVC in female COPD patients. Alterations observed in the corresponding male cohort were minor. Conclusion The identified molecular pathways suggest dysregulation of several phagocytosis-related pathways in BAL cells in female COPD patients, with correlation to both the level of obstruction (FEV1/FVC) and disease severity (FEV1) as well as emphysema (CT < −950 HU) in women. Trial registration No.: NCT02627872, retrospectively registered on December 9, 2015. Electronic supplementary material The online version of this article (10.1186/s12931-017-0699-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mingxing Yang
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden.
| | - Maxie Kohler
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden
| | - Tina Heyder
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden
| | - Helena Forsslund
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden
| | - Hilde K Garberg
- Proteomics Unit (PROBE), Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Reza Karimi
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden
| | - Johan Grunewald
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden
| | - Frode S Berven
- Proteomics Unit (PROBE), Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Sven Nyrén
- Department of Molecular Medicine and Surgery, Division of Radiology, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - C Magnus Sköld
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden
| | - Åsa M Wheelock
- Respiratory Medicine Unit, Department of Medicine Solna & Center for Molecular Medicine, Karolinska Institutet, Lung Research Lab L4:01, SE-171 76, Stockholm, Sweden.
| |
Collapse
|
28
|
Devaraj A, Buzzo J, Rocco CJ, Bakaletz LO, Goodman SD. The DNABII family of proteins is comprised of the only nucleoid associated proteins required for nontypeable Haemophilus influenzae biofilm structure. Microbiologyopen 2017; 7:e00563. [PMID: 29230970 PMCID: PMC6011942 DOI: 10.1002/mbo3.563] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/23/2017] [Accepted: 10/30/2017] [Indexed: 11/20/2022] Open
Abstract
Biofilms play a central role in the pathobiology of otitis media (OM), bronchitis, sinusitis, conjunctivitis, and pneumonia caused by nontypeable Haemophilus influenzae (NTHI). Our previous studies show that extracellular DNA (eDNA) and DNABII proteins are essential components of biofilms formed by NTHI. The DNABII protein family includes integration host factor (IHF) and the histone‐like protein HU and plays a central role in NTHI biofilm structural integrity. We demonstrated that immunological targeting of these proteins during NTHI‐induced experimental OM in a chinchilla model caused rapid clearance of biofilms from the middle ear. Given the essential role of DNABII proteins in maintaining the structure of an NTHI biofilm, we investigated whether any of the other nucleoid associated proteins (NAPs) expressed by NTHI might play a similar role, thereby serving as additional target(s) for intervention. We demonstrated that although several NAPs including H‐NS, CbpA, HfQ and Dps are present within the biofilm extracellular matrix, only the DNABII family of proteins is critical for the structural integrity of the biofilms formed by NTHI. We have also demonstrated that IHF and HU are located at distinct regions within the extracellular matrix of NTHI biofilms formed in vitro, indicative of independent functions of these two proteins.
Collapse
Affiliation(s)
- Aishwarya Devaraj
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| | - John Buzzo
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| | - Christopher J Rocco
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| | - Lauren O Bakaletz
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| | - Steven D Goodman
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
29
|
Tan DB, Armitage J, Teo TH, Ong NE, Shin H, Moodley YP. Elevated levels of circulating exosome in COPD patients are associated with systemic inflammation. Respir Med 2017; 132:261-264. [DOI: 10.1016/j.rmed.2017.04.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/06/2017] [Accepted: 04/25/2017] [Indexed: 10/19/2022]
|
30
|
Kim VL, Coombs NA, Staples KJ, Ostridge KK, Williams NP, Wootton SA, Devaster JM, Aris E, Clarke SC, Tuck AC, Bourne SC, Wilkinson TMA. Impact and associations of eosinophilic inflammation in COPD: analysis of the AERIS cohort. Eur Respir J 2017; 50:50/4/1700853. [PMID: 29025891 DOI: 10.1183/13993003.00853-2017] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/16/2017] [Indexed: 01/21/2023]
Abstract
Eosinophilic inflammation in chronic obstructive pulmonary disease (COPD) predicts response to treatment, especially corticosteroids. We studied the nature of eosinophilic inflammation in COPD prospectively to examine the stability of this phenotype and its dynamics across exacerbations, and its associations with clinical phenotype, exacerbations and infection.127 patients aged 40-85 years with moderate to very severe COPD underwent repeated blood and sputum sampling at stable visits and within 72 h of exacerbation for 1 year.Blood eosinophils ≥2% was prevalent at baseline, and predicted both predominantly raised stable-state eosinophils across the year (area under the curve 0.841, 95% CI 0.755-0.928) and increased risk of eosinophilic inflammation at exacerbation (OR 9.16; p<0.001). Eosinophils ≥2% at exacerbation and eosinophil predominance at stable visits were associated with a lower risk of bacterial presence at exacerbation (OR 0.49; p=0.049 and OR 0.25; p=0.065, respectively). Bacterial infection at exacerbation was highly seasonal (winter versus summer OR 4.74; p=0.011) in predominantly eosinophilic patients.Eosinophilic inflammation is a common and stable phenotype in COPD. Blood eosinophil counts in the stable state can predict the nature of inflammation at future exacerbations, which when combined with an understanding of seasonal variation provides the basis for the development of new treatment paradigms for this important condition.
Collapse
Affiliation(s)
- Viktoriya L Kim
- NIHR Southampton Respiratory Biomedical Research Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Ngaire A Coombs
- Primary Care and Population Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Karl J Staples
- NIHR Southampton Respiratory Biomedical Research Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | - Kristoffer K Ostridge
- NIHR Southampton Respiratory Biomedical Research Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Nicholas P Williams
- NIHR Southampton Respiratory Biomedical Research Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Stephen A Wootton
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | | | - Stuart C Clarke
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | - Andrew C Tuck
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Simon C Bourne
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Portsmouth Hospitals NHS Trust, Queen Alexandra Hospital, Portsmouth, UK
| | - Tom M A Wilkinson
- NIHR Southampton Respiratory Biomedical Research Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK .,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | | |
Collapse
|
31
|
Blakeway LV, Tan A, Peak IRA, Seib KL. Virulence determinants of Moraxella catarrhalis: distribution and considerations for vaccine development. MICROBIOLOGY-SGM 2017; 163:1371-1384. [PMID: 28893369 DOI: 10.1099/mic.0.000523] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Moraxella catarrhalis is a human-restricted opportunistic bacterial pathogen of the respiratory mucosa. It frequently colonizes the nasopharynx asymptomatically, but is also an important causative agent of otitis media (OM) in children, and plays a significant role in acute exacerbations of chronic obstructive pulmonary disease (COPD) in adults. As the current treatment options for M. catarrhalis infection in OM and exacerbations of COPD are often ineffective, the development of an efficacious vaccine is warranted. However, no vaccine candidates for M. catarrhalis have progressed to clinical trials, and information regarding the distribution of M. catarrhalis virulence factors and vaccine candidates is inconsistent in the literature. It is largely unknown if virulence is associated with particular strains or subpopulations of M. catarrhalis, or if differences in clinical manifestation can be attributed to the heterogeneous expression of specific M. catarrhalis virulence factors in the circulating population. Further investigation of the distribution of M. catarrhalis virulence factors in the context of carriage and disease is required so that vaccine development may be targeted at relevant antigens that are conserved among disease-causing strains. The challenge of determining which of the proposed M. catarrhalis virulence factors are relevant to human disease is amplified by the lack of a standardized M. catarrhalis typing system to facilitate direct comparisons of worldwide isolates. Here we summarize and evaluate proposed relationships between M. catarrhalis subpopulations and specific virulence factors in the context of colonization and disease, as well as the current methods used to infer these associations.
Collapse
Affiliation(s)
- Luke V Blakeway
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Aimee Tan
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Ian R A Peak
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia.,School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - Kate L Seib
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
32
|
Mantero M, Rogliani P, Di Pasquale M, Polverino E, Crisafulli E, Guerrero M, Gramegna A, Cazzola M, Blasi F. Acute exacerbations of COPD: risk factors for failure and relapse. Int J Chron Obstruct Pulmon Dis 2017; 12:2687-2693. [PMID: 28932112 PMCID: PMC5598966 DOI: 10.2147/copd.s145253] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Acute exacerbations are a leading cause of worsening COPD in terms of lung function decline, quality of life, and survival. They also have a relevant economic burden on the health care system. Determining the risk factors for acute exacerbation and early relapse could be a crucial element for a better management of COPD patients. This review analyzes the current knowledge and underlines the main risk factors for recurrent acute exacerbations. Comprehensive evaluation of COPD patients during stable phase and exacerbation could contribute to prevent treatment failure and relapses.
Collapse
Affiliation(s)
- Marco Mantero
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano.,Internal Medicine Department, Respiratory Unit and Regional Adult Cystic Fibrosis Center, IRCCS Fondazione Cà Granda Ospedale Maggiore Policlinico, Milan
| | - Paola Rogliani
- Respiratory Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Marta Di Pasquale
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano.,Internal Medicine Department, Respiratory Unit and Regional Adult Cystic Fibrosis Center, IRCCS Fondazione Cà Granda Ospedale Maggiore Policlinico, Milan
| | - Eva Polverino
- Respiratory Disease Department, Servei de Pneumologia, Hospital Universitari Vall d'Hebron (HUVH), Institut de Recerca Vall d'Hebron (VHIR), Barcelona, Spain
| | - Ernesto Crisafulli
- Department of Medicine and Surgery, Respiratory Disease and Lung Function Unit, University of Parma, Parma, Italy
| | - Monica Guerrero
- Hospital d'Igualada, Consorci Socisanitari de l'Anoia, Barcelona, Spain
| | - Andrea Gramegna
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano.,Internal Medicine Department, Respiratory Unit and Regional Adult Cystic Fibrosis Center, IRCCS Fondazione Cà Granda Ospedale Maggiore Policlinico, Milan
| | - Mario Cazzola
- Respiratory Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano.,Internal Medicine Department, Respiratory Unit and Regional Adult Cystic Fibrosis Center, IRCCS Fondazione Cà Granda Ospedale Maggiore Policlinico, Milan
| |
Collapse
|
33
|
Briend E, Ferguson GJ, Mori M, Damera G, Stephenson K, Karp NA, Sethi S, Ward CK, Sleeman MA, Erjefält JS, Finch DK. IL-18 associated with lung lymphoid aggregates drives IFNγ production in severe COPD. Respir Res 2017; 18:159. [PMID: 28830544 PMCID: PMC5568255 DOI: 10.1186/s12931-017-0641-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 08/10/2017] [Indexed: 11/16/2022] Open
Abstract
Background Increased interferon gamma (IFNγ) release occurs in Chronic Obstructive Pulmonary Disease (COPD) lungs. IFNγ supports optimal viral clearance, but if dysregulated could increase lung tissue destruction. Methods The present study investigates which mediators most closely correlate with IFNγ in sputum in stable and exacerbating disease, and seeks to shed light on the spatial requirements for innate production of IFNγ, as reported in mouse lymph nodes, to observe whether such microenvironmental cellular organisation is relevant to IFNγ production in COPD lung. Results We show tertiary follicle formation in severe disease alters the dominant mechanistic drivers of IFNγ production, because cells producing interleukin-18, a key regulator of IFNγ, are highly associated with such structures. Interleukin-1 family cytokines correlated with IFNγ in COPD sputum. We observed that the primary source of IL-18 in COPD lungs was myeloid cells within lymphoid aggregates and IL-18 was increased in severe disease. IL-18 released from infected epithelium or from activated myeloid cells, was more dominant in driving IFNγ when IL-18-producing and responder cells were in close proximity. Conclusions Unlike tight regulation to control infection spread in lymphoid organs, this local interface between IL-18-expressing and responder cell is increasingly supported in lung as disease progresses, increasing its potential to increase tissue damage via IFNγ. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0641-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emmanuel Briend
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,Present address: Agenus Ltd, Cambridge, UK
| | | | - Michiko Mori
- Department of Experimental Medical Science, BMC D12, Lund University, SE-221 84, Lund, Sweden
| | - Gautam Damera
- MedImmune LLC, 1 MedImmune Way, Gaithersburg, MD, USA
| | - Katherine Stephenson
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,Present address: University of Nottingham, Nottingham, UK
| | - Natasha A Karp
- Quantitative Biology IMED, AstraZeneca R&D, Cambridge, UK
| | - Sanjay Sethi
- Department of Medicine, University at Buffalo, 3495 Bailey Avenue, Buffalo, NY, 14215, USA
| | - Christine K Ward
- MedImmune LLC, 1 MedImmune Way, Gaithersburg, MD, USA.,Present address: Bristol-Myers Squibb, Princeton, NJ, USA
| | - Matthew A Sleeman
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,Present address: Regeneron Pharmaceuticals Inc, Tarrytown, NY, USA
| | - Jonas S Erjefält
- Department of Experimental Medical Science, BMC D12, Lund University, SE-221 84, Lund, Sweden.,Department of Respiratory Medicine and Allergology, Lund University Hospital, Lund, Sweden
| | - Donna K Finch
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.
| |
Collapse
|
34
|
Calverley PMA, Sethi S, Dawson M, Ward CK, Finch DK, Penney M, Newbold P, van der Merwe R. A randomised, placebo-controlled trial of anti-interleukin-1 receptor 1 monoclonal antibody MEDI8968 in chronic obstructive pulmonary disease. Respir Res 2017; 18:153. [PMID: 28793896 PMCID: PMC5551010 DOI: 10.1186/s12931-017-0633-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/31/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Interleukin-1 receptor 1 (IL-1R1) inhibition is a potential strategy for treating patients with chronic obstructive pulmonary disease (COPD). MEDI8968, a fully human monoclonal antibody, binds selectively to IL-1R1, inhibiting activation by IL-1α and IL-1β. We studied the efficacy and safety/tolerability of MEDI8968 in adults with symptomatic, moderate-to-very severe COPD. METHODS This was a phase II, randomised, double-blind, placebo-controlled, multicentre, parallel-group study. Subjects aged 45-75 years and receiving standard maintenance therapy with ≥2 exacerbations in the past year were randomised 1:1 to receive placebo or MEDI8968 300 mg (600 mg intravenous loading dose) subcutaneously every 4 weeks, for 52 weeks. The primary endpoint was the moderate/severe acute exacerbations of COPD (AECOPD) rate (week 56 post-randomisation). Secondary endpoints were severe AECOPD rate and St George's Respiratory Questionnaire-COPD (SGRQ-C) score (week 56 post-randomisation). RESULTS Of subjects randomised to placebo (n = 164) and MEDI8968 (n = 160), 79.3% and 75.0%, respectively, completed the study. There were neither statistically significant differences between treatment groups in moderate/severe AECOPD rate ([90% confidence interval]: 0.78 [0.63, 0.96], placebo; 0.71 [0.57, 0.90], MEDI8968), nor in severe AECOPD rate or SGRQ-C scores. Post-hoc analysis of subject subgroups (by baseline neutrophil count or tertiles of circulating neutrophil counts) did not alter the study outcome. The incidence of treatment-emergent adverse events (TEAEs) with placebo and MEDI8968 treatment was similar. The most common TEAE was worsening of COPD. CONCLUSIONS In this phase II study, MEDI8968 did not produce statistically significant improvements in AECOPD rate, lung function or quality of life. TRIAL REGISTRATION ClinicalTrials.gov, NCT01448850 , date of registration: 06 October 2011.
Collapse
Affiliation(s)
- Peter M. A. Calverley
- School of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Clinical Science Centre, University Hospital Aintree, Longmoor Lane, Liverpool, L9 7AL UK
| | - Sanjay Sethi
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Buffalo, State University of New York, Buffalo, NY USA
| | | | - Christine K. Ward
- MedImmmune, Gaithersburg, MD USA
- Present address: Bristol-Myers Squibb, Princeton, NJ USA
| | | | - Mark Penney
- MedImmune, Cambridge, UK
- Present address: UCB Pharma, Slough, UK
| | | | | |
Collapse
|
35
|
Hinks TSC, Wallington JC, Williams AP, Djukanović R, Staples KJ, Wilkinson TMA. Steroid-induced Deficiency of Mucosal-associated Invariant T Cells in the Chronic Obstructive Pulmonary Disease Lung. Implications for Nontypeable Haemophilus influenzae Infection. Am J Respir Crit Care Med 2017; 194:1208-1218. [PMID: 27115408 DOI: 10.1164/rccm.201601-0002oc] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
RATIONALE Mucosal-associated invariant T (MAIT) cells are a recently described abundant, proinflammatory T-cell subset with unknown roles in pulmonary immunity. Nontypeable Haemophilus influenzae (NTHi) is the leading bacterial pathogen during chronic obstructive pulmonary disease (COPD) exacerbations and is a plausible target for MAIT cells. OBJECTIVES To investigate whether MAIT cells respond to NTHi and the effects of inhaled corticosteroids (ICS) on their frequency and function in COPD. METHODS Eleven subjects with COPD receiving ICS, 8 steroid-naive subjects with COPD, and 21 healthy control subjects underwent phlebotomy, sputum induction, bronchoalveolar lavage, and endobronchial biopsy. Pulmonary and monocyte-derived macrophages were cultured in vitro with NTHi. MEASUREMENTS AND MAIN RESULTS Frequencies of Vα7.2+CD161+ MAIT cells, surface expression of the major histocompatibility complex-related protein 1 (MR1), and intracellular IFN-γ expression were measured by flow cytometry. MAIT-cell frequencies were reduced in peripheral blood of ICS-treated subjects with COPD (median 0.38%; interquartile range [IQR], 0.25-0.96) compared with healthy control subjects (1.8%; IQR, 1.4-2.5; P = 0.001) or steroid-naive patients with COPD (1.8%; IQR, 1.2-2.3; P = 0.04). MAIT cells were reduced in bronchial biopsies from subjects with COPD treated with steroids (0.73%; IQR, 0.46-1.3) compared with healthy control subjects (4.0%; IQR, 1.6-5.0; P = 0.02). Coculture of live NTHi increased macrophage surface expression of MR1 and induced IFN-γ from CD4 cells and CD8 cells, but most potently from MAIT cells (median IFN-γ-positive frequencies, 2.9, 8.6, and 27.6%, respectively). In vitro fluticasone and budesonide reduced MR1 surface expression twofold and decreased NTHi-induced IFN-γ secretion eightfold. CONCLUSIONS MAIT cells are deficient in blood and bronchial tissue in steroid-treated, but not steroid-naive, COPD. NTHi constitutes a target for pulmonary MAIT-cell immune responses, which are significantly impaired by corticosteroids.
Collapse
Affiliation(s)
- Timothy S C Hinks
- 1 Clinical & Experimental Sciences and.,2 Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Southampton, United Kingdom.,3 Department for Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia; and
| | | | - Anthony P Williams
- 4 Cancer Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Southampton, United Kingdom.,5 Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, United Kingdom
| | - Ratko Djukanović
- 1 Clinical & Experimental Sciences and.,2 Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Southampton, United Kingdom
| | - Karl J Staples
- 1 Clinical & Experimental Sciences and.,5 Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, United Kingdom
| | - Tom M A Wilkinson
- 1 Clinical & Experimental Sciences and.,2 Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Southampton, United Kingdom.,5 Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
36
|
Chronic lung inflammation primes humoral immunity and augments antipneumococcal resistance. Sci Rep 2017; 7:4972. [PMID: 28694492 PMCID: PMC5504016 DOI: 10.1038/s41598-017-05212-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 05/25/2017] [Indexed: 11/16/2022] Open
Abstract
Airway epithelial cells (AECs) display remarkable plasticity in response to infectious stimuli and their functional adaptations are critical for antimicrobial immunity. However, the roles of AECs and humoral mediators to host defense in non-communicable lung inflammation remain elusive. We dissected pulmonary defense against Streptococcus pneumoniae in hosts with pre-existing inflammatory conditions (SPC-HAxTCR-HA mice). Lung tissue transcriptomics and bronchoalveolar lavage fluid (BALF) proteomics revealed an induction of humoral defense mechanisms in inflamed lungs. Accordingly, besides antibacterial proteins and complement components being overrepresented in inflamed lungs, elevated polymeric immunoglobulin receptor (pIgR)-expression in AECs correlated with increased secretory immunoglobulin (SIg) transport. Consequently, opsonization assays revealed augmented pneumococcal coverage by SIgs present in the BALF of SPC-HAxTCR-HA mice, which was associated with enhanced antipneumococcal resistance. These findings emphasize the immunologic potential of AECs as well as their central role in providing antibacterial protection and put forward pIgR as potential target for therapeutic manipulation in infection-prone individuals.
Collapse
|
37
|
Ahearn CP, Gallo MC, Murphy TF. Insights on persistent airway infection by non-typeable Haemophilus influenzae in chronic obstructive pulmonary disease. Pathog Dis 2017; 75:3753446. [PMID: 28449098 PMCID: PMC5437125 DOI: 10.1093/femspd/ftx042] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/28/2017] [Indexed: 12/21/2022] Open
Abstract
Non-typeable Haemophilus influenzae (NTHi) is the most common bacterial cause of infection of the lower airways in adults with chronic obstructive pulmonary disease (COPD). Infection of the COPD airways causes acute exacerbations, resulting in substantial morbidity and mortality. NTHi has evolved multiple mechanisms to establish infection in the hostile environment of the COPD airways, allowing the pathogen to persist in the airways for months to years. Persistent infection of the COPD airways contributes to chronic airway inflammation that increases symptoms and accelerates the progressive loss of pulmonary function, which is a hallmark of the disease. Persistence mechanisms of NTHi include the expression of multiple redundant adhesins that mediate binding to host cellular and extracellular matrix components. NTHi evades host immune recognition and clearance by invading host epithelial cells, forming biofilms, altering gene expression and displaying surface antigenic variation. NTHi also binds host serum factors that confer serum resistance. Here we discuss the burden of COPD and the role of NTHi infections in the course of the disease. We provide an overview of NTHi mechanisms of persistence that allow the pathogen to establish a niche in the hostile COPD airways.
Collapse
Affiliation(s)
- Christian P. Ahearn
- Department of Microbiology and Immunology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
- Clinical and Translational Research Center, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Mary C. Gallo
- Department of Microbiology and Immunology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
- Clinical and Translational Research Center, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Timothy F. Murphy
- Department of Microbiology and Immunology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
- Clinical and Translational Research Center, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
- Division of Infectious Disease, Department of Medicine, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| |
Collapse
|
38
|
Garcia-Nuñez M, Marti S, Puig C, Perez-Brocal V, Millares L, Santos S, Ardanuy C, Moya A, Liñares J, Monsó E. Bronchial microbiome, PA biofilm-forming capacity and exacerbation in severe COPD patients colonized by P. aeruginosa. Future Microbiol 2017; 12:379-392. [PMID: 28339291 DOI: 10.2217/fmb-2016-0127] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 11/14/2016] [Indexed: 02/07/2023] Open
Abstract
AIM The bronchial microbiome of severe chronic obstructive pulmonary disease patients colonized by Pseudomonas aeruginosa was analyzed using 16S rRNA gene sequencing to identify differences related to biofilm-forming capacity. PATIENTS & METHODS Patient sputum samples from 21 patients were studied. RESULTS Statistically significant differences related to biofilm-forming capacity were only found for genera with relative abundances <1%, and Fusobacterium was over-represented when biofilm-forming capacity was high. Genera with relative abundances >50% which increased from baseline were observed in 10/14 exacerbations, but corresponded to Pseudomonas only in three episodes, while other pathogenic genera were identified in seven. CONCLUSION The bronchial microbiome shows differences according with P. aeruginosa biofilm-forming capacity. Pathogenic microorganisms other than P. aeruginosa cause a significant part of the exacerbations in colonized chronic obstructive pulmonary disease patients.
Collapse
Affiliation(s)
- Marian Garcia-Nuñez
- CIBER de Enfermedades Respiratorias (CIBERes), Instituto de Salud Carlos III, Madrid, Spain E-28029
- Department of Respiratory Medicine, Parc Taulí Hospital Universitari. Institut d'Investigació i Innovació Parc Taulí I3PT, Sabadell, Spain E-08208
- Fundació Institut d'Investigació Germans Trias i Pujol, Badalona, Spain E-08916
- Universitat Autonoma de Barcelona, Esfera UAB, Barcelona, Spain E-08193
| | - Sara Marti
- CIBER de Enfermedades Respiratorias (CIBERes), Instituto de Salud Carlos III, Madrid, Spain E-28029
- Microbiology Department, Hospital Universitari Bellvitge, Universitat de Barcelona-IDIBELL, Barcelona, Spain E-08908
| | - Carmen Puig
- CIBER de Enfermedades Respiratorias (CIBERes), Instituto de Salud Carlos III, Madrid, Spain E-28029
- Microbiology Department, Hospital Universitari Bellvitge, Universitat de Barcelona-IDIBELL, Barcelona, Spain E-08908
| | - Vicente Perez-Brocal
- Genomics & Health Area, Centro Superior de Investigación en Salud Pública - Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (CSISP-FISABIO), Valencia, Spain E-46020
- Department of Genetics, Institut Cavanilles de Biodiversitat i Biologia Evolutiva, (ICBiBE) Universitat de València, València, Spain E-46020
- CIBER Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain E-28029
| | - Laura Millares
- CIBER de Enfermedades Respiratorias (CIBERes), Instituto de Salud Carlos III, Madrid, Spain E-28029
- Department of Respiratory Medicine, Parc Taulí Hospital Universitari. Institut d'Investigació i Innovació Parc Taulí I3PT, Sabadell, Spain E-08208
- Fundació Institut d'Investigació Germans Trias i Pujol, Badalona, Spain E-08916
- Universitat Autonoma de Barcelona, Esfera UAB, Barcelona, Spain E-08193
| | - Salud Santos
- CIBER de Enfermedades Respiratorias (CIBERes), Instituto de Salud Carlos III, Madrid, Spain E-28029
- Department of Pulmonary Medicine, Hospital Universitari de Bellvitge, Institut d'Investigacions Biomèdiques de Bellvitge (IDIBELL), Universitat de Barcelona, Barcelona, Spain E-08908
| | - Carmen Ardanuy
- CIBER de Enfermedades Respiratorias (CIBERes), Instituto de Salud Carlos III, Madrid, Spain E-28029
- Microbiology Department, Hospital Universitari Bellvitge, Universitat de Barcelona-IDIBELL, Barcelona, Spain E-08908
| | - Andres Moya
- Genomics & Health Area, Centro Superior de Investigación en Salud Pública - Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (CSISP-FISABIO), Valencia, Spain E-46020
- Department of Genetics, Institut Cavanilles de Biodiversitat i Biologia Evolutiva, (ICBiBE) Universitat de València, València, Spain E-46020
- CIBER Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain E-28029
| | - Josefina Liñares
- CIBER de Enfermedades Respiratorias (CIBERes), Instituto de Salud Carlos III, Madrid, Spain E-28029
- Microbiology Department, Hospital Universitari Bellvitge, Universitat de Barcelona-IDIBELL, Barcelona, Spain E-08908
| | - Eduard Monsó
- CIBER de Enfermedades Respiratorias (CIBERes), Instituto de Salud Carlos III, Madrid, Spain E-28029
- Department of Respiratory Medicine, Parc Taulí Hospital Universitari. Institut d'Investigació i Innovació Parc Taulí I3PT, Sabadell, Spain E-08208
- Fundació Institut d'Investigació Germans Trias i Pujol, Badalona, Spain E-08916
- Universitat Autonoma de Barcelona, Esfera UAB, Barcelona, Spain E-08193
| |
Collapse
|
39
|
Lin W, Jianbo S, Wanzhong L, Yanna L, Weiwei S, Gang W, Chunzhen Z. Protective effect of eucalyptus oil on pulmonary destruction and inflammation in chronic obstructive pulmonary disease (COPD) in rats. ACTA ACUST UNITED AC 2017. [DOI: 10.5897/jmpr2015.5910] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
40
|
Vij N. Nano-based rescue of dysfunctional autophagy in chronic obstructive lung diseases. Expert Opin Drug Deliv 2016; 14:483-489. [PMID: 27561233 DOI: 10.1080/17425247.2016.1223040] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION ΔF508-CFTR (cystic fibrosis transmembrane conductance regulator) is a common CF-mutation that is known to induce oxidative-inflammatory stress through activation of reactive oxygen species (ROS), which induces autophagy-impairment resulting in accumulation of CFTR in aggresome-bodies. Cysteamine, the reduced form of cystamine, is a FDA-approved drug that has anti-oxidant, anti-bacterial, and mucolytic properties. This drug has been shown in a recent clinical trial to decrease lung inflammation and improve lung function in CF patients by potentially restoring autophagy and allowing CFTR to be trafficked to the cell membrane. Areas covered: The delivery of cysteamine to airway epithelia of chronic subjects prerequisite the need for a delivery system to allow rescue of dysfunctional autophagy. Expert opinion: We anticipate based on our ongoing studies that PLGA-PEG- or Dendrimer-mediated cysteamine delivery could allow sustained airway delivery over standard cysteamine tablets or delay release capsules that are currently used for systemic treatment. In addition, proposed nano-based autophagy induction strategy can also allow rescue of cigarette smoke (CS) induced acquired-CFTR dysfunction seen in chronic obstructive pulmonary disease (COPD)-emphysema subjects. The CS induced acquired-CFTR dysfunction involves CFTR-accumulation in aggresome-bodies that can be rescued by an autophagy-inducing antioxidant drug, cysteamine. Moreover, chronic CS-exposure generates ROS that induces overall protein-misfolding and aggregation of ubiquitinated-proteins as aggresome-bodies via autophagy-impairment that can be also be resolved by treatment with autophagy-inducing antioxidant drug, cysteamine.
Collapse
Affiliation(s)
- Neeraj Vij
- a College of Medicine , Central Michigan University , Mount Pleasant , MI , USA.,b Department of Pediatric Respiratory Sciences , The Johns Hopkins School of Medicine , Baltimore , MD , USA
| |
Collapse
|
41
|
Ueda K, Nishimoto Y, Kimura G, Masuko T, Barnes PJ, Ito K, Kizawa Y. Repeated lipopolysaccharide exposure causes corticosteroid insensitive airway inflammation via activation of phosphoinositide-3-kinase δ pathway. Biochem Biophys Rep 2016; 7:367-373. [PMID: 28955927 PMCID: PMC5613638 DOI: 10.1016/j.bbrep.2016.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 12/19/2022] Open
Abstract
Corticosteroid resistance is one of major barriers to effective management of chronic inflammatory respiratory diseases, such as chronic obstructive pulmonary disease (COPD) and severe asthma. These patients often experience exacerbations with viral and/or bacterial infection, which may cause continuous corticosteroid insensitive inflammation. In this study, we observed that repeated exposure of lipopolysaccharide (LPS) intranasally attenuated the anti-inflammatory effects of the corticosteroid fluticasone propionate (FP) on neutrophils and CXCL1 levels in bronchoalveolar lavage (BAL) fluid in an in vivo murine model. Histone deacetylase-2 (HDAC2) and NF-E2 related factor 2 (Nrf2) levels in lungs after LPS administration for 3 consecutive days were significantly decreased to 38.9±6.3% (mean±SEM) and 77.5±2.7% of the levels seen after only one day of LPS exposure, respectively. In addition, 3 days LPS exposure resulted in an increase of Akt phosphorylation, indicating activation of the phosphoinositide-3-kinase (PI3K) pathway by 4-fold in lungs compared with 1 day of exposure. Furthermore, combination treatment with theophylline and FP significantly decreased the neutrophil accumulation and CXCL1 concentrations in BAL fluid from 22.5±1.8×104 cells/mL and 214.6±20.6 pg/mL to 7.9±0.5×104 cells/mL and 61.9±13.3 pg/mL, respectively. Combination treatment with IC87114, a selective PI3Kδ inhibitor, and FP also significantly decreased neutrophils and CXCL1 levels from 16.8±0.7×104 cells/mL and 182.4±4.6 pg/mL to 5.9±0.3×104 cells/mL and 71.4±2.7 pg/mL, respectively. Taken together, repeated exposure of LPS causes corticosteroid-insensitive airway inflammation in vivo, and the corticosteroid-resistance induced by LPS is at least partly mediated through the activation of PI3Kδ, resulting in decreased levels of HDAC2 and Nrf2. These findings provide a potentially new therapeutic approach to COPD and severe asthma. We first time proposed clinically relevant steroid insensitive exacerbation model. LPS caused corticosteroid insensitive airway inflammation via PI3Kδ activation. This murine model are useful to discover new therapies for airway inflammation.
Collapse
Affiliation(s)
- Keitaro Ueda
- Department of Physiology and Anatomy, Nihon University School of Pharmacy, Chiba, Japan
| | - Yuki Nishimoto
- Department of Physiology and Anatomy, Nihon University School of Pharmacy, Chiba, Japan.,PMS Business Solutions Division, CMIC-PMS Co., Ltd., Tokyo, Japan
| | - Genki Kimura
- Department of Physiology and Anatomy, Nihon University School of Pharmacy, Chiba, Japan
| | - Takashi Masuko
- Department of Physiology and Anatomy, Nihon University School of Pharmacy, Chiba, Japan
| | - Peter J Barnes
- Airway Disease Division, NHLI, Imperial College, London, United Kingdom
| | - Kazuhiro Ito
- Airway Disease Division, NHLI, Imperial College, London, United Kingdom
| | - Yasuo Kizawa
- Department of Physiology and Anatomy, Nihon University School of Pharmacy, Chiba, Japan
| |
Collapse
|
42
|
van de Geijn GJM, Denker S, Meuleman-van Waning V, Koeleman HGM, Birnie E, Braunstahl GJ, Njo TL. Evaluation of new laboratory tests to discriminate bacterial from nonbacterial chronic obstructive pulmonary disease exacerbations. Int J Lab Hematol 2016; 38:616-628. [PMID: 27459873 DOI: 10.1111/ijlh.12550] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 06/03/2016] [Indexed: 01/30/2023]
Abstract
INTRODUCTIONS Discriminating bacterial from nonbacterial acute exacerbations of chronic obstructive pulmonary disease (AECOPD) is difficult, causing antibiotics overuse and bacterial resistance. Sputum cultures are of limited use because results take time. In our hospital, only leukocyte concentration and CRP are laboratory parameters evaluated in AECOPD. We evaluated additional tests to discriminate bacterial vs. nonbacterial AECOPD: 5-part leukocyte differentiation (hematology analyzer), leukocyte differentiation using flow cytometry (Leukoflow, Cytodiff), Leuko64 kit, and procalcitonin. METHODS Retrospectively, patients were classified as bacterial or nonbacterial AECOPD. ROC analyses tested how the additional tests discriminate these groups. RESULTS Twenty-two AECOPD were classified as bacterial and 23 as nonbacterial. From the additional tests, basophil percentage (Cytodiff) has superior AUC (0.800). At a cutoff resulting in ≥90% sensitivity, neutrophil/lymphocyte ratio (AUC:0.755) and CD4-positive T cells (Leukoflow, AUC:0.747) have the highest specificity (57%). Both neutrophil mean volume and standard deviation (Cell Population Data, DxH800 hematology analyzer) had good combined sensitivity and specificity (AUC:0.846/0.804, 91% sensitivity, 69% specificity). Addition of leukocyte populations and procalcitonin to CRP in regression models (AUC: 0.907/0.876/0.890) increased specificity compared to CRP alone (71% or 73% vs. 39%). CONCLUSION No additional test has sufficient accuracy on its own to predict bacterial AECOPD. Combining CRP with several parameters from the additional tests may improve this.
Collapse
Affiliation(s)
- G-J M van de Geijn
- Department of Clinical Chemistry (KCHL), Franciscus Gasthuis, Rotterdam, The Netherlands
| | - S Denker
- Department of Pulmonology/Internal Medicine, Franciscus Gasthuis, Rotterdam, The Netherlands
| | - V Meuleman-van Waning
- Department of Pulmonology/Internal Medicine, Franciscus Gasthuis, Rotterdam, The Netherlands
| | - H G M Koeleman
- Department of Microbiology, Franciscus Gasthuis, Rotterdam, The Netherlands
| | - E Birnie
- Department of Statistics and Education, Franciscus Gasthuis, Rotterdam, The Netherlands
| | - G-J Braunstahl
- Department of Pulmonology/Internal Medicine, Franciscus Gasthuis, Rotterdam, The Netherlands
| | - T L Njo
- Department of Clinical Chemistry (KCHL), Franciscus Gasthuis, Rotterdam, The Netherlands
| |
Collapse
|
43
|
Rationale and Design of the Genomic Research in Alpha-1 Antitrypsin Deficiency and Sarcoidosis Study. Alpha-1 Protocol. Ann Am Thorac Soc 2016; 12:1551-60. [PMID: 26153726 DOI: 10.1513/annalsats.201503-143oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Severe deficiency of alpha-1 antitrypsin has a highly variable clinical presentation. The Genomic Research in Alpha-1 Antitrypsin Deficiency and Sarcoidosis α1 Study is a prospective, multicenter, cross-sectional study of adults older than age 35 years with PiZZ or PiMZ alpha-1 antitrypsin genotypes. It is designed to better understand if microbial factors influence this heterogeneity. Clinical symptoms, pulmonary function testing, computed chest tomography, exercise capacity, and bronchoalveolar lavage (BAL) will be used to define chronic obstructive pulmonary disease (COPD) phenotypes that can be studied with an integrated systems biology approach that includes plasma proteomics; mouth, BAL, and stool microbiome and virome analysis; and blood microRNA and blood mononuclear cell RNA and DNA profiling. We will rely on global genome, transcriptome, proteome, and metabolome datasets. Matched cohorts of PiZZ participants on or off alpha-1 antitrypsin augmentation therapy, PiMZ participants not on augmentation therapy, and control participants from the Subpopulations and Intermediate Outcome Measures in COPD Study who match on FEV1 and age will be compared. In the primary analysis, we will determine if the PiZZ individuals on augmentation therapy have a difference in lower respiratory tract microbes identified compared with matched PiZZ individuals who are not on augmentation therapy. By characterizing the microbiome in alpha-1 antitrypsin deficiency (AATD), we hope to define new phenotypes of COPD that explain some of the diversity of clinical presentations. As a unique genetic cause of COPD, AATD may inform typical COPD pathogenesis, and better understanding of it may illuminate the complex interplay between environment and genetics. Although the biologic approaches are hypothesis generating, the results may lead to development of novel biomarkers, better understanding of COPD phenotypes, and development of novel diagnostic and therapeutic trials in AATD and COPD. Clinical trial registered with www.clinicaltrials.gov (NCT01832220).
Collapse
|
44
|
Damera G, Pham TH, Zhang J, Ward CK, Newbold P, Ranade K, Sethi S. A Sputum Proteomic Signature That Associates with Increased IL-1β Levels and Bacterial Exacerbations of COPD. Lung 2016; 194:363-9. [PMID: 27083436 DOI: 10.1007/s00408-016-9877-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 04/04/2016] [Indexed: 11/25/2022]
Abstract
PURPOSE Activation of the interleukin-1β (IL-1β) signaling pathway has been implicated in COPD, but the proportion of COPD subjects whose disease is principally driven by activation of this pathway is poorly understood. In this study, we sought to differentiate an IL-1β-associated sputum signature from other inflammation-associated COPD phenotypes. METHODS Luminex-multiplex assays were used to study IL-1β-mediated signature proteins within airway epithelium, smooth muscle, and vascular endothelial cell cultures. The IL-1β-mediated signature was tested in a longitudinal study comprising of 35 paired stable-COPD and acute exacerbation (AECOPD) sputum samples. The presence of respiratory pathogens (H. influenzae, M. catarrhalis, S. pneumoniae, and P. aeruginosa) was evaluated by sputum cultures. RESULTS Five proteins namely TNF-α, GCSF, IL-6, CD-40L, and MIP-1β were found to be IL-1β-regulated across all donors and cell types. All five of these IL-1β-mediated proteins were significantly increased (p < 0.05) in sputum corresponding to AECOPD events showing at least a twofold increase in IL-1β (IL-1β(+) events, 18 of 35 total events), relative to preceding stable-COPD state. Sputum IL-1β levels showed no significant association (p > 0.05, spearman) with known markers of other major COPD inflammation phenotypes. In addition, there was a significant association with bacterial presence in sputum culture with an odds ratio of 9 (95 % CI 1.56, 51.9) in IL-1β(+) events versus IL-1β(-) events. CONCLUSION Our findings provide insights into potential markers of IL-1β-associated AECOPD, and reaffirm association between IL-1β pathway activation and airway bacterial infection in COPD. Taken together, our findings could help identify COPD patient subsets who may benefit from therapies targeting IL-1β pathway.
Collapse
Affiliation(s)
- Gautam Damera
- Translational Medicine - Respiratory, Inflammation, Autoimmunity, MedImmune LLC, One Medimmune Way, Gaithersburg, MD, 20878, USA.
| | - Tuyet-Hang Pham
- Translational Medicine - Respiratory, Inflammation, Autoimmunity, MedImmune LLC, One Medimmune Way, Gaithersburg, MD, 20878, USA
| | - Jianchun Zhang
- Nonclinical Statistics, MedImmune LLC, One Medimmune Way, Gaithersburg, MD, 20878, USA
| | - Christine K Ward
- Translational Medicine - Respiratory, Inflammation, Autoimmunity, MedImmune LLC, One Medimmune Way, Gaithersburg, MD, 20878, USA
| | - Paul Newbold
- Translational Medicine - Respiratory, Inflammation, Autoimmunity, MedImmune LLC, One Medimmune Way, Gaithersburg, MD, 20878, USA
| | - Koustubh Ranade
- Translational Medicine - Respiratory, Inflammation, Autoimmunity, MedImmune LLC, One Medimmune Way, Gaithersburg, MD, 20878, USA
| | - Sanjay Sethi
- VA WNY Healthcare System and University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
45
|
Guerrero M, Crisafulli E, Liapikou A, Huerta A, Gabarrús A, Chetta A, Soler N, Torres A. Readmission for Acute Exacerbation within 30 Days of Discharge Is Associated with a Subsequent Progressive Increase in Mortality Risk in COPD Patients: A Long-Term Observational Study. PLoS One 2016; 11:e0150737. [PMID: 26943928 PMCID: PMC4778849 DOI: 10.1371/journal.pone.0150737] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/18/2016] [Indexed: 01/10/2023] Open
Abstract
Background and Objective Twenty per cent of chronic obstructive pulmonary disease (COPD) patients are readmitted for acute exacerbation (AECOPD) within 30 days of discharge. The prognostic significance of early readmission is not fully understood. The objective of our study was to estimate the mortality risk associated with readmission for acute exacerbation within 30 days of discharge in COPD patients. Methods The cohort (n = 378) was divided into patients readmitted (n = 68) and not readmitted (n = 310) within 30 days of discharge. Clinical, laboratory, microbiological, and severity data were evaluated at admission and during hospital stay, and mortality data were recorded at four time points during follow-up: 30 days, 6 months, 1 year and 3 years. Results Patients readmitted within 30 days had poorer lung function, worse dyspnea perception and higher clinical severity. Two or more prior AECOPD (HR, 2.47; 95% CI, 1.51–4.05) was the only variable independently associated with 30-day readmission. The mortality risk during the follow-up period showed a progressive increase in patients readmitted within 30 days in comparison to patients not readmitted; moreover, 30-day readmission was an independent risk factor for mortality at 1 year (HR, 2.48; 95% CI, 1.10–5.59). In patients readmitted within 30 days, the estimated absolute increase in the mortality risk was 4% at 30 days (number needed to harm NNH, 25), 17% at 6-months (NNH, 6), 19% at 1-year (NNH, 6) and 24% at 3 years (NNH, 5). Conclusion In conclusion a readmission for AECOPD within 30 days is associated with a progressive increased long-term risk of death.
Collapse
Affiliation(s)
- Mónica Guerrero
- Respiratory Department, Clinic Institute of Respiratory (ICR), Hospital Clinic of Barcelona - Institut d'Investigacions Biomèdiques Agusti Pi i Sunyer (IDIBAPS) - CIBERES - University of Barcelona (UB), Barcelona, Spain
| | - Ernesto Crisafulli
- Department of Clinical and Experimental Medicine, Respiratory Disease and Lung Function Unit, University of Parma, Parma, Italy
| | | | - Arturo Huerta
- Respiratory Department, Clinic Institute of Respiratory (ICR), Hospital Clinic of Barcelona - Institut d'Investigacions Biomèdiques Agusti Pi i Sunyer (IDIBAPS) - CIBERES - University of Barcelona (UB), Barcelona, Spain
| | - Albert Gabarrús
- Respiratory Department, Clinic Institute of Respiratory (ICR), Hospital Clinic of Barcelona - Institut d'Investigacions Biomèdiques Agusti Pi i Sunyer (IDIBAPS) - CIBERES - University of Barcelona (UB), Barcelona, Spain
| | - Alfredo Chetta
- Department of Clinical and Experimental Medicine, Respiratory Disease and Lung Function Unit, University of Parma, Parma, Italy
| | - Nestor Soler
- Respiratory Department, Clinic Institute of Respiratory (ICR), Hospital Clinic of Barcelona - Institut d'Investigacions Biomèdiques Agusti Pi i Sunyer (IDIBAPS) - CIBERES - University of Barcelona (UB), Barcelona, Spain
| | - Antoni Torres
- Respiratory Department, Clinic Institute of Respiratory (ICR), Hospital Clinic of Barcelona - Institut d'Investigacions Biomèdiques Agusti Pi i Sunyer (IDIBAPS) - CIBERES - University of Barcelona (UB), Barcelona, Spain
- * E-mail:
| |
Collapse
|
46
|
Gupta V, Banyard A, Mullan A, Sriskantharajah S, Southworth T, Singh D. Characterization of the inflammatory response to inhaled lipopolysaccharide in mild to moderate chronic obstructive pulmonary disease. Br J Clin Pharmacol 2016; 79:767-76. [PMID: 25377849 DOI: 10.1111/bcp.12546] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 10/28/2014] [Indexed: 01/10/2023] Open
Abstract
AIMS Lipopolysaccharide (LPS) inhalation causes increased airway and systemic inflammation. We investigated LPS inhalation in patients with chronic obstructive pulmonary disease (COPD) as a model of bacterial exacerbations. We studied safety, changes in sputum and systemic biomarkers. We have also investigated interleukin (IL)-17 concentrations in this model. METHODS Twelve COPD patients inhaled 5 μg LPS. Safety was monitored over 24 h. Sputum was induced at baseline, 6 and 24 h for cells and IL-8, IL-17, neutrophil elastase, monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-1β (MIP-1β) in supernatants. Serum was collected at baseline, 4, 8 and 24 h for IL-6, C-reactive protein (CRP) and Clara cell protein (CC-16) concentrations. Peripheral blood mononuclear cells (PBMCs) were isolated at baseline and 4 h for systemic IL-17 analysis. RESULTS LPS 5 μg was well tolerated. The greatest FEV1 change was 11.7% (mean) at 1 h (95% CI 5.1-18.2%). There was a large range in maximal fall (2.5-37.7%). Total sputum cell count and neutrophil count significantly increased 6 and 24 h post-LPS. There was no change in sputum supernatant mediators. IL-6, CRP and CC-16 increased post-inhalation, with different temporal patterns. CD4+ and CD8+ cell associated IL-17 significantly increased at 4 h. CONCLUSIONS Inhaled LPS in COPD patients safely causes increased airway and systemic inflammation. This may be a model for studying COPD exacerbations.
Collapse
Affiliation(s)
- Vandana Gupta
- Medicines Evaluation Unit, University Hospital of South Manchester Foundation Trust, University of Manchester, Southmoor Rd, Manchester, UK
| | | | | | | | | | | |
Collapse
|
47
|
Martin C, Burgel PR, Lepage P, Andréjak C, de Blic J, Bourdin A, Brouard J, Chanez P, Dalphin JC, Deslée G, Deschildre A, Gosset P, Touqui L, Dusser D. Host-microbe interactions in distal airways: relevance to chronic airway diseases. Eur Respir Rev 2015; 24:78-91. [PMID: 25726559 PMCID: PMC9487770 DOI: 10.1183/09059180.00011614] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
This article is the summary of a workshop, which took place in November 2013, on the roles of microorganisms in chronic respiratory diseases. Until recently, it was assumed that lower airways were sterile in healthy individuals. However, it has long been acknowledged that microorganisms could be identified in distal airway secretions from patients with various respiratory diseases, including cystic fibrosis (CF) and non-CF bronchiectasis, chronic obstructive pulmonary disease, asthma and other chronic airway diseases (e.g. post-transplantation bronchiolitis obliterans). These microorganisms were sometimes considered as infectious agents that triggered host immune responses and contributed to disease onset and/or progression; alternatively, microorganisms were often considered as colonisers, which were considered unlikely to play roles in disease pathophysiology. These concepts were developed at a time when the identification of microorganisms relied on culture-based methods. Importantly, the majority of microorganisms cannot be cultured using conventional methods, and the use of novel culture-independent methods that rely on the identification of microorganism genomes has revealed that healthy distal airways display a complex flora called the airway microbiota. The present article reviews some aspects of current literature on host–microbe (mostly bacteria and viruses) interactions in healthy and diseased airways, with a special focus on distal airways. Understanding host–microbe interactions in distal airways may lead to novel therapies for chronic airway diseaseshttp://ow.ly/HfENz
Collapse
Affiliation(s)
- Clémence Martin
- Hôpital Cochin, AP-HP, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Pierre-Régis Burgel
- Hôpital Cochin, AP-HP, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Patricia Lepage
- UMR1913-Microbiologie de l'Alimentation au Service de la Santé, l'Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - Claire Andréjak
- Respiratory Intensive Care Unit, Centre Hospitalier Universitaire Sud, Amiens, France
| | - Jacques de Blic
- Hôpital Necker-Enfants Malades, AP-HP, Université Paris Descartes, Paris, France
| | - Arnaud Bourdin
- Hôpital Arnaud de Villeneuve, CHU Montpellier, Montpellier, France
| | - Jacques Brouard
- Dept of Pediatrics, CHU de Caen, Research Unit EA 4655 U2RM, Caen, France
| | - Pascal Chanez
- Dépt des Maladies Respiratoires, AP-HM, Laboratoire d'immunologie INSERM CNRS U 1067, UMR 7733, Aix Marseille Université, Marseille, France
| | | | - Gaetan Deslée
- Dept of Pulmonary Medicine, University Hospital of Reims, Reims, France
| | | | - Philippe Gosset
- Unité de défense innée et inflammation, Institut Pasteur, Paris, France INSERM U874, Paris, France
| | - Lhousseine Touqui
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Daniel Dusser
- Hôpital Cochin, AP-HP, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
48
|
Ikeda M, Enomoto N, Hashimoto D, Fujisawa T, Inui N, Nakamura Y, Suda T, Nagata T. Nontypeable Haemophilus influenzae exploits the interaction between protein-E and vitronectin for the adherence and invasion to bronchial epithelial cells. BMC Microbiol 2015; 15:263. [PMID: 26572616 PMCID: PMC4647820 DOI: 10.1186/s12866-015-0600-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 11/06/2015] [Indexed: 12/05/2022] Open
Abstract
Background Nontypeable Haemophilus influenzae (NTHi) is one of the most common Gram-negative pathogens in otitis media and exacerbation of chronic obstructive pulmonary disease. NTHi has been reported to invade bronchial epithelial cells. This penetration enables NTHi to evade the host immune system and antibiotics, and it seems to be related to the intractable features of these diseases. However, the precise mechanism of the invasion has been unknown. We hypothesized that protein-E, an outer membrane protein of NTHi, plays a role in this penetration into bronchial epithelial cells. Results We utilized two NTHi strains. NTHi efficiently attached to plate-bound vitronectin (254–309 / field at 1,000× magnification) and this attachment was blocked by pretreatment with protein-E peptide (PE84–108). The blockade of adhesion was dependent on the concentration of PE84–108. NTHi strains invaded bronchial epithelial cells and the intracellular bacteria were localized in early endosomes. Furthermore, intracellular invasion of NTHi was also blocked by PE84–108, but not by Arg-Gly-Asp (RGD) peptide. Pretreatment with PE84–108 significantly prevented cells from being invaded by both NTHi strains, which was confirmed by fluorescent microscope observation. In addition, pretreatment with PE84–108 significantly reduced percentages of CFU after gentamicin treatment of cells per input CFU. Conclusions These results suggest that NTHi does not directly bind to the cell surface, but binds to host vitronectin that is bound to the cell surface, via bacterial protein-E. Bacterial protein-E and host vitronectin play a role in the attachment to bronchial epithelial cells and is also involved in the subsequent intracellular invasion of NTHi. A novel vaccine or treatment strategy targeting the protein-E-vitronectin axis may prevent respiratory intracellular infection of NTHi and may lead to better clinical outcomes. Electronic supplementary material The online version of this article (doi:10.1186/s12866-015-0600-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Masaki Ikeda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Dai Hashimoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Naoki Inui
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Yutaro Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Toshi Nagata
- Department of Health Science, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| |
Collapse
|
49
|
Chillappagari S, Preuss J, Licht S, Müller C, Mahavadi P, Sarode G, Vogelmeier C, Guenther A, Nahrlich L, Rubin BK, Henke MO. Altered protease and antiprotease balance during a COPD exacerbation contributes to mucus obstruction. Respir Res 2015; 16:85. [PMID: 26169056 PMCID: PMC4501272 DOI: 10.1186/s12931-015-0247-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/01/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Proteases have been shown to degrade airway mucin proteins and to damage the epithelium impairing mucociliary clearance. There are increased proteases in the COPD airway but changes in protease-antiprotease balance and mucin degradation have not been investigated during the course of a COPD exacerbation. We hypothesized that increased protease levels would lead to mucin degradation in acute COPD exacerbations. METHODS We measured neutrophil elastase (NE) and alpha 1 protease inhibitor (A1-PI) levels using immunoblotting, and conducted protease inhibitor studies, zymograms, elastin substrate assays and cigarette smoke condensate experiments to evaluate the stability of the gel-forming mucins, MUC5AC and MUC5B, before and 5-6 weeks after an acute pulmonary exacerbation of COPD (n = 9 subjects). RESULTS Unexpectedly, mucin concentration and mucin stability were highest at the start of the exacerbation and restored to baseline after 6 weeks. Consistent with these data, immunoblots and zymograms confirmed decreased NE concentration and activity and increased A1-PI at the start of the exacerbation. After recovery there was an increase in NE activity and a decrease in A1-PI levels. In vitro, protease inhibitor studies demonstrated that serine proteases played a key role in mucin degradation. Mucin stability was further enhanced upon treating with cigarette smoke condensate (CSC). CONCLUSION There appears to be rapid consumption of secreted proteases due to an increase in antiproteases, at the start of a COPD exacerbation. This leads to increased mucin gel stability which may be important in trapping and clearing infectious and inflammatory mediators, but this may also contribute acutely to mucus retention.
Collapse
Affiliation(s)
- Shashi Chillappagari
- Department of Pediatrics, Justus Liebig University Giessen, Feulgenstrasse 12, Giessen, 35392, Germany.,Department of Internal Medicine, Justus-Liebig-University Giessen, Klinikstrasse-36, Giessen, 35392, Germany.,Department of Pulmonary Medicine, Philipps-University Marburg, Baldingerstrasse 1, Marburg, 35043, Germany.,Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Jenni Preuss
- Department of Pulmonary Medicine, Philipps-University Marburg, Baldingerstrasse 1, Marburg, 35043, Germany
| | - Sebastian Licht
- Department of Pulmonary Medicine, Philipps-University Marburg, Baldingerstrasse 1, Marburg, 35043, Germany
| | - Christian Müller
- Department of Pulmonary Medicine, Philipps-University Marburg, Baldingerstrasse 1, Marburg, 35043, Germany
| | - Poornima Mahavadi
- Department of Internal Medicine, Justus-Liebig-University Giessen, Klinikstrasse-36, Giessen, 35392, Germany.,Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Gaurav Sarode
- Asklepios Fachkliniken München-Gauting, Robert-Koch-Allee 2, Gauting, 82131, Germany.,Member of the Comprehensive Pneumology Center (CPC), Helmholtz Zentrum, Munich, Germany
| | - Claus Vogelmeier
- Department of Pulmonary Medicine, Philipps-University Marburg, Baldingerstrasse 1, Marburg, 35043, Germany
| | - Andreas Guenther
- Department of Internal Medicine, Justus-Liebig-University Giessen, Klinikstrasse-36, Giessen, 35392, Germany.,Member of the German Centre for Lung Research (DZL), Giessen, Germany.,Member of the European IPF Registry/Biobank, Giessen, Germany.,Lung Clinic Waldhof-Elgershausen, Greifenstein, Germany
| | - Lutz Nahrlich
- Department of Pediatrics, Justus Liebig University Giessen, Feulgenstrasse 12, Giessen, 35392, Germany.,Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Bruce K Rubin
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, 1001 East Marshall Street, Richmond, 23298, VA, USA
| | - Markus O Henke
- Department of Pulmonary Medicine, Philipps-University Marburg, Baldingerstrasse 1, Marburg, 35043, Germany. .,Asklepios Fachkliniken München-Gauting, Robert-Koch-Allee 2, Gauting, 82131, Germany. .,Member of the Comprehensive Pneumology Center (CPC), Helmholtz Zentrum, Munich, Germany.
| |
Collapse
|
50
|
Gutbier B, Fischer K, Doehn JM, von Lachner C, Herr C, Klaile E, Frischmann U, Singer BB, Riesbeck K, Zimmermann W, Suttorp N, Bachmann S, Bals R, Witzenrath M, Slevogt H. Moraxella catarrhalis induces an immune response in the murine lung that is independent of human CEACAM5 expression and long-term smoke exposure. Am J Physiol Lung Cell Mol Physiol 2015; 309:L250-61. [PMID: 26047639 DOI: 10.1152/ajplung.00265.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 05/29/2015] [Indexed: 01/09/2023] Open
Abstract
In patients with chronic obstructive pulmonary disease (COPD), Moraxella catarrhalis infection of the lower airways is associated with chronic colonization and inflammation during stable disease and acute exacerbations. Chronic smoke exposure induces chronic inflammation and impairs mucociliary clearance, thus contributing to bacterial colonization of the lower airways in COPD patients. The human-specific carcinoembryonic antigen-related cell adhesion molecule (CEACAM) 5, expressed in human airways, has been shown to contribute to epithelial colonization of CEACAM-binding pathogens. To investigate the impact of CEACAM5 expression on pulmonary M. catarrhalis colonization, we infected mice transgenic for human CEACAM5 (hCEACAM5) and wild type mice intratracheally with M. catarrhalis with or without preceding smoke exposure and analyzed bacterial colonization and local and systemic inflammation. Our results show that airway infection with M. catarrhalis accelerated acute local but not systemic inflammation, albeit independent of hCEACAM5 expression. Long-term smoke exposure alone or prior to M. catarrhalis infection did not contribute to increased local or systemic inflammation. No difference was found in pulmonary clearance of M. catarrhalis in hCEACAM5-transgenic mice compared with wild-type mice. Smoke exposure neither altered time nor extent of persistence of M. catarrhalis in the lungs of both genotypes. In conclusion, M. catarrhalis induced a local acute immune response in murine airways. Neither hCEACAM5 expression nor chronic smoke exposure nor a combination of both was sufficient as prerequisites for the establishment of chronic M. catarrhalis colonization. Our results demonstrate the difficulties in mirroring conditions of chronic airways colonization of M. catarrhalis in a murine model.
Collapse
Affiliation(s)
- Birgitt Gutbier
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Katja Fischer
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany; Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Jan-Moritz Doehn
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Christian Herr
- Department of Internal Medicine V-Pulmonology, Allergology, Respiratory Intensive Care Medicine, University of the Saarland, Homburg Saar, Germany
| | - Esther Klaile
- Septomics Research Center, Jena University Hospital, Jena, Germany; Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | | | - Bernhard B Singer
- Institute of Anatomy, Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - Kristian Riesbeck
- Clinical Microbiology, Department of Translational Medicine Malmö, Lund University, Malmö, Sweden
| | - Wolfgang Zimmermann
- Tumor Immunology Laboratory, LIFE-Center, Klinikum Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany; and
| | - Norbert Suttorp
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Bachmann
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Bals
- Department of Internal Medicine V-Pulmonology, Allergology, Respiratory Intensive Care Medicine, University of the Saarland, Homburg Saar, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hortense Slevogt
- Septomics Research Center, Jena University Hospital, Jena, Germany;
| |
Collapse
|