1
|
Higham A, Beech A, Singh D. The relevance of eosinophils in chronic obstructive pulmonary disease: inflammation, microbiome, and clinical outcomes. J Leukoc Biol 2024; 116:927-946. [PMID: 38941350 DOI: 10.1093/jleuko/qiae153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 05/31/2024] [Accepted: 06/27/2024] [Indexed: 06/30/2024] Open
Abstract
Chronic obstructive pulmonary disease is caused by the inhalation of noxious particles such as cigarette smoke. The pathophysiological features include airway inflammation, alveolar destruction, and poorly reversible airflow obstruction. A subgroup of patients with chronic obstructive pulmonary disease has higher blood eosinophil counts, associated with an increased response to inhaled corticosteroids and increased biomarkers of pulmonary type 2 inflammation. Emerging evidence shows that patients with chronic obstructive pulmonary disease with increased pulmonary eosinophil counts have an altered airway microbiome. Higher blood eosinophil counts are also associated with increased lung function decline, implicating type 2 inflammation in progressive pathophysiology in chronic obstructive pulmonary disease. We provide a narrative review of the role of eosinophils and type 2 inflammation in the pathophysiology of chronic obstructive pulmonary disease, encompassing the lung microbiome, pharmacological targeting of type 2 pathways in chronic obstructive pulmonary disease, and the clinical use of blood eosinophil count as a chronic obstructive pulmonary disease biomarker.
Collapse
Affiliation(s)
- Andrew Higham
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester and Manchester University NHS Foundation Trust, Manchester, M23 9LT, United Kingdom
| | - Augusta Beech
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester and Manchester University NHS Foundation Trust, Manchester, M23 9LT, United Kingdom
| | - Dave Singh
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester and Manchester University NHS Foundation Trust, Manchester, M23 9LT, United Kingdom
- Medicines Evaluation Unit, The Langley Building, Southmoor Road, Manchester, M23 9QZ, United Kingdom
| |
Collapse
|
2
|
Wechsler ME, Wells JM. What every clinician should know about inflammation in COPD. ERJ Open Res 2024; 10:00177-2024. [PMID: 39319045 PMCID: PMC11417604 DOI: 10.1183/23120541.00177-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/22/2024] [Indexed: 09/26/2024] Open
Abstract
Inflammation drives COPD pathogenesis and exacerbations. Although the conceptual framework and major players in the inflammatory milieu of COPD have been long established, the nuances of cellular interactions and the etiological differences that create heterogeneity in inflammatory profiles and treatment response continue to be revealed. This wealth of data and understanding is not only a boon to the researcher but also provides guidance to the clinician, moving the field closer to precision medicine. It is through this lens that this review seeks to describe the inflammatory processes at play in COPD, relating inflammation to pathological and functional changes, identifying patient-specific and disease-related factors that may influence clinical observations, and providing current insights on existing and emerging anti-inflammatory treatments and treatment targets, including biological therapies and phosphodiesterase (PDE) inhibitors.
Collapse
Affiliation(s)
- Michael E. Wechsler
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - J. Michael Wells
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
3
|
Xu J, Zeng Q, Li S, Su Q, Fan H. Inflammation mechanism and research progress of COPD. Front Immunol 2024; 15:1404615. [PMID: 39185405 PMCID: PMC11341368 DOI: 10.3389/fimmu.2024.1404615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common respiratory disease characterized by irreversible progressive airflow limitation, often manifested by persistent cough, sputum production and other respiratory symptoms that pose a serious threat to human health and affect the quality of life of patients. The disease is associated with chronic inflammation, which is associated with the onset and progression of COPD, but anti-inflammatory therapy is not first-line treatment. Inflammation has multiple manifestations and phenotypes, and this heterogeneity reveals different patterns of inflammation, making treatment difficult. This paper aims to explore the direction of more effective anti-inflammatory treatment by analyzing the nature of inflammation and the molecular mechanism of disease occurrence and development in COPD patients, and to provide new ideas for the treatment of COPD patients.
Collapse
Affiliation(s)
- Jiao Xu
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qingyue Zeng
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shuangqing Li
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiaoli Su
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Fan
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Abi Abdallah G, Diop S, Jamme M, Legriel S, Ferré A. Respiratory Infection Triggering Severe Acute Exacerbations of Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2024; 19:555-565. [PMID: 38440747 PMCID: PMC10909653 DOI: 10.2147/copd.s447162] [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: 11/15/2023] [Accepted: 02/15/2024] [Indexed: 03/06/2024] Open
Abstract
Background Data are scarce on respiratory infections during severe acute exacerbation of chronic obstructive pulmonary disease (COPD). This study aimed to investigate respiratory infection patterns in the intensive care unit (ICU) and identify variables associated with infection type and patient outcome. Methods A retrospective, single-centre cohort study. All patients admitted (2015-2021) to our ICU for severe acute exacerbation of COPD were included. Logistic multivariable regression analysis was performed to predict factors associated with infection and assess the association between infection and outcome. Results We included 473 patients: 288 (60.9%) had respiratory infection and 139 (29.4%) required invasive mechanical ventilation. Eighty-nine (30.9%) had viral, 81 (28.1%) bacterial, 34 (11.8%) mixed, and 84 (29.2%) undocumented infections. Forty-seven (9.9%) patients died in the ICU and 67 (14.2%) in hospital. Factors associated with respiratory infection were temperature (odds ratio [+1°C]=1.43, P=0.008) and blood neutrophils (1.07, P=0.002). Male sex (2.21, P=0.02) and blood neutrophils were associated with bacterial infection (1.06, P=0.04). In a multivariable analysis, pneumonia (cause-specific hazard=1.75, P=0.005), respiratory rate (1.17, P=0.04), arterial partial pressure of carbon-dioxide (1.08, P=0.04), and lactate (1.14, P=0.02) were associated with the need for invasive MV. Age (1.03, P=0.03), immunodeficiency (1.96, P=0.02), and altered performance status (1.78, P=0.002) were associated with hospital mortality. Conclusions Respiratory infections, 39.9% of which were bacterial, were the main cause of severe acute exacerbation of COPD. Body temperature and blood neutrophils were single markers of infection. Pneumonia was associated with the need for invasive mechanical ventilation but not with hospital mortality, as opposed to age, immunodeficiency, and altered performance status.
Collapse
Affiliation(s)
| | - Sylvain Diop
- Cardiothoracic Intensive Care Unit, Department of Anesthesiology, Marie Lannelongue Hospital, Le Plessis Robinson, France
| | - Matthieu Jamme
- Service de Réanimation Polyvalente, Hôpital Privé de l’Ouest Parisien, Ramsay-Générale de Santé, Trappes, France
- CESP, INSERM U1018, Equipe Epidémiologie Clinique, Villejuif, France
| | - Stéphane Legriel
- Intensive Care Unit, Versailles Hospital, Le Chesnay, France
- University Paris-Saclay, UVSQ, INSERM, CESP, Team ”PsyDev”, Villejuif, France
| | - Alexis Ferré
- Intensive Care Unit, Versailles Hospital, Le Chesnay, France
| |
Collapse
|
5
|
Pragman AA, Hodgson SW, Wu T, Zank A, Reilly CS, Wendt CH. Sputum microbiome α-diversity is a key feature of the COPD frequent exacerbator phenotype. ERJ Open Res 2024; 10:00595-2023. [PMID: 38333651 PMCID: PMC10851948 DOI: 10.1183/23120541.00595-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/07/2023] [Indexed: 02/10/2024] Open
Abstract
Background The lung microbiome is an inflammatory stimulus whose role in COPD pathogenesis is incompletely understood. We hypothesised that the frequent exacerbator phenotype is associated with decreased α-diversity and increased lung inflammation. Our objective was to assess correlations between the frequent exacerbator phenotype, the microbiome and inflammation longitudinally during exacerbation-free periods. Methods We conducted a case-control longitudinal observational study of the frequent exacerbator phenotype and characteristics of the airway microbiome. 81 subjects (41 frequent and 40 infrequent exacerbators) provided nasal, oral and sputum microbiome samples at two visits over 2-4 months. Exacerbation phenotype, relevant clinical factors and sputum cytokine values were associated with microbiome findings. Results The frequent exacerbator phenotype was associated with lower sputum microbiome α-diversity (p=0.0031). This decrease in α-diversity among frequent exacerbators was enhanced when the sputum bacterial culture was positive (p<0.001). Older age was associated with decreased sputum microbiome α-diversity (p=0.0030). Between-visit β-diversity was increased among frequent exacerbators and those who experienced a COPD exacerbation between visits (p=0.025 and p=0.014, respectively). Sputum cytokine values did not differ based on exacerbation phenotype or other clinical characteristics. Interleukin (IL)-17A was negatively associated with α-diversity, while IL-6 and IL-8 were positively associated with α-diversity (p=0.012, p=0.012 and p=0.0496, respectively). IL-22, IL-17A and IL-5 levels were positively associated with Moraxella abundance (p=0.027, p=0.0014 and p=0.0020, respectively). Conclusions Even during exacerbation-free intervals, the COPD frequent exacerbator phenotype is associated with decreased sputum microbiome α-diversity and increased β-diversity. Decreased sputum microbiome α-diversity and Moraxella abundance are associated with lung inflammation.
Collapse
Affiliation(s)
- Alexa A. Pragman
- Department of Medicine, Minneapolis VA Health Care System and Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Shane W. Hodgson
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - Tianhua Wu
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Allison Zank
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - Cavan S. Reilly
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Chris H. Wendt
- Department of Medicine, Minneapolis VA Health Care System and Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
6
|
Shin D, Kim J, Lee JH, Kim JI, Oh YM. Profiling of Microbial Landscape in Lung of Chronic Obstructive Pulmonary Disease Patients Using RNA Sequencing. Int J Chron Obstruct Pulmon Dis 2023; 18:2531-2542. [PMID: 38022823 PMCID: PMC10644840 DOI: 10.2147/copd.s426260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose The aim of the study was to use RNA sequencing (RNA-seq) data of lung from chronic obstructive pulmonary disease (COPD) patients to identify the bacteria that are most commonly detected. Additionally, the study sought to investigate the differences in these infections between normal lung tissues and those affected by COPD. Patients and Methods We re-analyzed RNA-seq data of lung from 99 COPD patients and 93 non-COPD smokers to determine the extent to which the metagenomes differed between the two groups and to assess the reliability of the metagenomes. We used unmapped reads in the RNA-seq data that were not aligned to the human reference genome to identify more common infections in COPD patients. Results We identified 18 bacteria that exhibited significant differences between the COPD and non-COPD smoker groups. Among these, Yersinia enterocolitica was found to be more than 30% more abundant in COPD. Additionally, we observed difference in detection rate based on smoking history. To ensure the accuracy of our findings and distinguish them from false positives, we double-check the metagenomic profile using Basic Local Alignment Search Tool (BLAST). We were able to identify and remove specific species that might have been misclassified as other species in Kraken2 but were actually Staphylococcus aureus, as identified by BLAST analysis. Conclusion This study highlighted the method of using unmapped reads, which were not typically used in sequencing data, to identify microorganisms present in patients with lung diseases such as COPD. This method expanded our understanding of the microbial landscape in COPD and provided insights into the potential role of microorganisms in disease development and progression.
Collapse
Affiliation(s)
- Dongjin Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Juhyun Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jang Ho Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jong-Il Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Genomic Medicine Institute, Seoul National University, Seoul, Republic of Korea
- Seoul National University Cancer Research Institute, Seoul, Republic of Korea
| | - Yeon-Mok Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
7
|
Rodgers AM, Lindsay J, Monahan A, Dubois AV, Faniyi AA, Plant BJ, Mall MA, Ekkelenkamp MB, Elborn S, Ingram RJ. Biologically Relevant Murine Models of Chronic Pseudomonas aeruginosa Respiratory Infection. Pathogens 2023; 12:1053. [PMID: 37624013 PMCID: PMC10458525 DOI: 10.3390/pathogens12081053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is an opportunistic pathogen and the leading cause of infection in patients with cystic fibrosis (CF). The ability of P. aeruginosa to evade host responses and develop into chronic infection causes significant morbidity and mortality. Several mouse models have been developed to study chronic respiratory infections induced by P. aeruginosa, with the bead agar model being the most widely used. However, this model has several limitations, including the requirement for surgical procedures and high mortality rates. Herein, we describe novel and adapted biologically relevant models of chronic lung infection caused by P. aeruginosa. Three methods are described: a clinical isolate infection model, utilising isolates obtained from patients with CF; an incomplete antibiotic clearance model, leading to bacterial bounce-back; and the establishment of chronic infection; and an adapted water bottle chronic infection model. These models circumvent the requirement for a surgical procedure and, importantly, can be induced with clinical isolates of P. aeruginosa and in wild-type mice. We also demonstrate successful induction of chronic infection in the transgenic βENaC murine model of CF. We envisage that the models described will facilitate the investigations of host and microbial factors, and the efficacy of novel antimicrobials, during chronic P. aeruginosa respiratory infections.
Collapse
Affiliation(s)
- Aoife M. Rodgers
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| | - Jaime Lindsay
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| | - Avril Monahan
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| | - Alice V. Dubois
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| | - Aduragbemi A. Faniyi
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| | - Barry J. Plant
- Cork Centre for Cystic Fibrosis (3CF), Cork University Hospital, University College Cork, T12 E8YV Cork, Ireland
- The HRB Funded Clinical Research Facility, University College Cork, T12 E8YV Cork, Ireland
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—University of Medicine Berlin, 10117 Berlin, Germany
- German Center for Lung Research (DZL), 10117 Berlin, Germany
- Berlin Institute of Health at Charité—University of Medicine Berlin, 10117 Berlin, Germany
| | - Miquel B. Ekkelenkamp
- Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Stuart Elborn
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| | - Rebecca J. Ingram
- Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK (S.E.)
| |
Collapse
|
8
|
Rozaliyani A, Antariksa B, Nurwidya F, Zaini J, Setianingrum F, Hasan F, Nugrahapraja H, Yusva H, Wibowo H, Bowolaksono A, Kosmidis C. The Fungal and Bacterial Interface in the Respiratory Mycobiome with a Focus on Aspergillus spp. Life (Basel) 2023; 13:life13041017. [PMID: 37109545 PMCID: PMC10142979 DOI: 10.3390/life13041017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The heterogeneity of the lung microbiome and its alteration are prevalently seen among chronic lung diseases patients. However, studies to date have primarily focused on the bacterial microbiome in the lung rather than fungal composition, which might play an essential role in the mechanisms of several chronic lung diseases. It is now well established that Aspergillus spp. colonies may induce various unfavorable inflammatory responses. Furthermore, bacterial microbiomes such as Pseudomonas aeruginosa provide several mechanisms that inhibit or stimulate Aspergillus spp. life cycles. In this review, we highlighted fungal and bacterial microbiome interactions in the respiratory tract, with a focus on Aspergillus spp.
Collapse
Affiliation(s)
- Anna Rozaliyani
- Department of Parasitology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Indonesia Pulmonary Mycoses Centre, Jakarta 10430, Indonesia
| | - Budhi Antariksa
- Department of Pulmonoloy and Respiratory Medicine, Faculty of Medicinie, Universitas Indonesia, Persahabatan National Respiratory Referral Hospital, Jakarta 13230, Indonesia
| | - Fariz Nurwidya
- Department of Pulmonoloy and Respiratory Medicine, Faculty of Medicinie, Universitas Indonesia, Persahabatan National Respiratory Referral Hospital, Jakarta 13230, Indonesia
| | - Jamal Zaini
- Department of Pulmonoloy and Respiratory Medicine, Faculty of Medicinie, Universitas Indonesia, Persahabatan National Respiratory Referral Hospital, Jakarta 13230, Indonesia
| | - Findra Setianingrum
- Department of Parasitology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Indonesia Pulmonary Mycoses Centre, Jakarta 10430, Indonesia
| | - Firman Hasan
- Indonesia Pulmonary Mycoses Centre, Jakarta 10430, Indonesia
| | - Husna Nugrahapraja
- Life Science and Biotechnology, Bandung Institute of Technology, Bandung 40312, Indonesia
| | - Humaira Yusva
- Magister Program of Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Heri Wibowo
- Department of Parasitology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Anom Bowolaksono
- Department of Biology, Faculty of Mathematics and Natural Sciences (FMIPA), Universitas Indonesia, Depok 16424, Indonesia
| | - Chris Kosmidis
- Manchester Academic Health Science Centre, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M23 9LT, UK
| |
Collapse
|
9
|
Baker JM, Baba-Dikwa A, Shah R, Lea S, Singh D. Gallium protoporphyrin as an antimicrobial for non-typeable Haemophilus influenzae in COPD patients. Life Sci 2022; 305:120794. [PMID: 35835251 DOI: 10.1016/j.lfs.2022.120794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/06/2022] [Accepted: 07/06/2022] [Indexed: 11/30/2022]
Abstract
AIMS Colonisation with non-typeable Haemophilus influenzae (NTHi) is common in COPD. Iron is required by bacteria for nutrition. Gallium is imported into bacteria using iron import proteins. Gallium cannot fulfill key metabolic functions, causing bactericidal effects. We tested the efficacy of gallium compounds as antimicrobials against NTHi in hemin rich conditions, and their ability to reduce NTHi induced pro-inflammatory responses in macrophages. MAIN METHODS NTHi was cultured with the free iron analogue gallium nitrate (GaN) and heme iron analogue gallium protoporphyrin (GaPP) (0.5-4 μM; 24 h). Growth of NTHi reference strain (NCTC 12699) and 6 clinical isolates from COPD patients (including antibiotic resistant isolates) was assessed by optical density, and viability by Miles Misra. Monocyte derived macrophages (MDMs) were treated with GaPP before/after NTHi exposure. Viable intracellular NTHi was assessed by gentamicin protection assay. GaN or GaPP was added to NTHi cultures prior to culture with MDMs. Cytokine gene expression (qPCR) and protein secretion (ELISA) were measured. KEY FINDINGS NTHi growth and viability were reduced by GaPP but not GaN. GaPP inhibited growth of COPD isolates (4 μM: 87 % reduction). GaPP reduced intracellular viability of NTHi in macrophage infection models. MDM cytokine gene expression and protein secretion (TNF-α, IL-6 and CXCL8) in response to NTHi was reduced (82, 66 and 86 % for gene expression) when cultured with GaPP 4 μM. SIGNIFICANCE GaPP is an effective antimicrobial for NTHi while GaN showed no effect on growth or viability. Culture of NTHi with GaPP also reduced the pro-inflammatory cytokine response in MDMs.
Collapse
Affiliation(s)
- James M Baker
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.
| | - Aisha Baba-Dikwa
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Rajesh Shah
- Department of Thoracic Surgery, Manchester University Hospital NHS Foundation Trust, Manchester, UK
| | - Simon Lea
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Dave Singh
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK; Medicines Evaluation Unit, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
10
|
Lea S, Beech A, Baker J, Gaskell R, Pindolia D, Dikwa AB, Shah R, Singh D. Differential responses of COPD macrophages to respiratory bacterial pathogens. ERJ Open Res 2022; 8:00044-2022. [PMID: 35923420 PMCID: PMC9339767 DOI: 10.1183/23120541.00044-2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/04/2022] [Indexed: 11/08/2022] Open
Abstract
COPD patients have increased susceptibility to airway bacterial colonisation. Haemophilus influenzae, Moraxella catarrhalis and Streptococcus pneumoniae are three of the most common respiratory bacterial species in COPD. H. influenzae colonisation, but not other bacteria, in COPD patients is associated with higher sputum neutrophil counts. Alveolar macrophages are key in clearance of bacteria as well as releasing mediators to recruit and activate other immune cells in response to infection. The aim was to characterise differences in COPD macrophage responses to H. influenzae, M. catarrhalisand S. pneumoniae, focusing on release of inflammatory and chemotactic mediators, and apoptosis regulation. Lung macrophages and monocyte-derived macrophages from COPD patients and control subjects were exposed to H. influenzae, M. catarrhalisor S. pneumoniae. Cytokine secretion (tumour necrosis factor-α, interleukin (IL)-6, CXCL8, CCL5 and IL-1β) were measured by ELISA and quantitative reverse transcriptase PCR (RT-qPCR), and apoptosis genes MCL-1, BCL-2, BAX and BAK1 by RT-qPCR. Apoptosis and reactive oxygen species (ROS) release were also measured. Macrophages responded differentially to the bacterial species, with increased, prolonged production of the neutrophil chemoattractant CXCL8 in response to H. influenzae and M. catarrhalis but not S. pneumoniae. S. pneumoniae initiated macrophage apoptosis and ROS release, H. influenzae and M. catarrhalis did not and increased anti-apoptosis gene expression (BCL-2 5.5-fold and MCL-1 2.4-fold, respectively). Differential cytokine responses of macrophages to these bacterial species can explain neutrophilic airway inflammation associated with H. influenzae, but not S. pneumoniae in COPD. Furthermore, delayed macrophage apoptosis is a potential mechanism contributing to inability to clear H. influenzae. Differential cytokine responses of macrophages to Haemophilus influenzae, Moraxella catarrhalis and Streptococcus pneumoniae can explain neutrophilic airway inflammation associated with H. influenzae but not S. pneumoniae in COPDhttps://bit.ly/3950HVZ
Collapse
|
11
|
Wedzicha JA. Vaccines for COPD exacerbation prevention: do they work? THE LANCET. RESPIRATORY MEDICINE 2022; 10:422-423. [PMID: 35026178 DOI: 10.1016/s2213-2600(22)00007-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 06/14/2023]
Affiliation(s)
- Jadwiga A Wedzicha
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK.
| |
Collapse
|
12
|
Love ME, Proud D. Respiratory Viral and Bacterial Exacerbations of COPD—The Role of the Airway Epithelium. Cells 2022; 11:cells11091416. [PMID: 35563722 PMCID: PMC9099594 DOI: 10.3390/cells11091416] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 12/14/2022] Open
Abstract
COPD is a leading cause of death worldwide, with acute exacerbations being a major contributor to disease morbidity and mortality. Indeed, exacerbations are associated with loss of lung function, and exacerbation frequency predicts poor prognosis. Respiratory infections are important triggers of acute exacerbations of COPD. This review examines the role of bacterial and viral infections, along with co-infections, in the pathogenesis of COPD exacerbations. Because the airway epithelium is the initial site of exposure both to cigarette smoke (or other pollutants) and to inhaled pathogens, we will focus on the role of airway epithelial cell responses in regulating the pathophysiology of exacerbations of COPD. This will include an examination of the interactions of cigarette smoke alone, and in combination with viral and bacterial exposures in modulating epithelial function and inflammatory and host defense pathways in the airways during COPD. Finally, we will briefly examine current and potential medication approaches to treat acute exacerbations of COPD triggered by respiratory infections.
Collapse
|
13
|
Unravelling the molecular mechanisms underlying chronic respiratory diseases for the development of novel therapeutics via in vitro experimental models. Eur J Pharmacol 2022; 919:174821. [DOI: 10.1016/j.ejphar.2022.174821] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/01/2022] [Accepted: 02/09/2022] [Indexed: 12/11/2022]
|
14
|
Pereira M, Oh JK, Kang DK, Engstrand L, Valeriano VD. Hacking Commensal Bacteria to Consolidate the Adaptive Mucosal Immune Response in the Gut-Lung Axis: Future Possibilities for SARS-CoV-2 Protection. BIOTECH 2022; 11:3. [PMID: 35822811 PMCID: PMC9245903 DOI: 10.3390/biotech11010003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/04/2022] [Accepted: 01/26/2022] [Indexed: 11/17/2022] Open
Abstract
Infectious diseases caused by mucosal pathogens significantly increase mortality and morbidity. Thus, the possibility to target these pathogens at their primary entry points can consolidate protective immunity. Regarding SARS-CoV-2 infection, it has been observed that the upper respiratory mucosa is highly affected and that dysregulation of resident microbiota in the gut-lung axis plays a crucial role in determining symptom severity. Thus, understanding the possibility of eliciting various mucosal and adaptive immune responses allows us to effectively design bacterial mucosal vaccine vectors. Such design requires rationally selecting resident bacterial candidates as potential host carriers, evaluating effective carrier proteins for stimulating an immune response, and combining these two to improve antigenic display and immunogenicity. This review investigated mucosal vaccine vectors from 2015 to present, where a few have started to utilize Salmonella and lactic acid bacteria (LAB) to display SARS-CoV-2 Spike S proteins or fragments. Although current literature is still lacking for its studies beyond in vitro or in vivo efficiency, decades of research into these vectors show promising results. Here, we discuss the mucosal immune systems focusing on the gut-lung axis microbiome and offer new insight into the potential use of alpha streptococci in the upper respiratory tract as a vaccine carrier.
Collapse
Affiliation(s)
- Marcela Pereira
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165 Stockholm, Sweden; (M.P.); (J.K.O.); (L.E.)
| | - Ju Kyoung Oh
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165 Stockholm, Sweden; (M.P.); (J.K.O.); (L.E.)
| | - Dae-Kyung Kang
- Department of Animal Resources Science, Dankook University, Cheonan 31116, Korea;
| | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165 Stockholm, Sweden; (M.P.); (J.K.O.); (L.E.)
| | - Valerie Diane Valeriano
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17165 Stockholm, Sweden; (M.P.); (J.K.O.); (L.E.)
| |
Collapse
|
15
|
Armitage MN, Spittle DA, Turner AM. A Systematic Review and Meta-Analysis of the Prevalence and Impact of Pulmonary Bacterial Colonisation in Stable State Chronic Obstructive Pulmonary Disease (COPD). Biomedicines 2021; 10:biomedicines10010081. [PMID: 35052762 PMCID: PMC8773377 DOI: 10.3390/biomedicines10010081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Half of acute exacerbations of COPD are due to bacterial infection, and the other half are likely influenced by microbial colonisation. The same organisms commonly cultured during acute exacerbations are often found in the sputum of patients during stability. A robust assessment of the prevalence of potentially pathogenic microorganisms (PPMs) in the sputum of stable COPD patients may help to inform the targeted prevention of exacerbation by these organisms. Methods: A systematic review and meta-analysis was carried out to determine the prevalence of PPMs in patients with COPD in the stable state. Meta-analysis of prevalence was carried out using the Freeman–Tukey double arcsine transformation random effects model, and sub-group analysis was performed for sputum modality. Prevalence of total and individual PPMs was calculated from patient-level data from individual studies. Results: Pooled prevalence of PPMs identified by sputum culture was found to be 41% (95% CI 36–47%). Significant heterogeneity was found across all studies, which can likely be attributed to inconsistent measuring and reporting of PPMs. The most commonly reported organisms were H. influenzae, M catarrhalis, S. pneumoniae, S. aureus, and P. aeruginosa. Declining lung function was weakly correlated with prevalence of PPMs. Conclusion: The airways of patients with COPD are colonised with PPMs during the stable state in almost half of patients. A complex relationship likely exists between the microbiome in the stable state and the phenotype of COPD patients. Targeted microbial therapy for preventing exacerbations of COPD should carefully consider the stable microbiome as well as the exacerbated.
Collapse
Affiliation(s)
- Michael N. Armitage
- Medical Education, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Rd., Coventry CV2 2DX, UK;
| | - Daniella A. Spittle
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK;
| | - Alice M. Turner
- University Hospitals Birmingham NHS Foundation Trust, Institute of Applied Health Research, University of Birmingham, Birmingham B15 2TT, UK
- Correspondence:
| |
Collapse
|
16
|
D’Anna SE, Maniscalco M, Cappello F, Carone M, Motta A, Balbi B, Ricciardolo FLM, Caramori G, Di Stefano A. Bacterial and viral infections and related inflammatory responses in chronic obstructive pulmonary disease. Ann Med 2021; 53:135-150. [PMID: 32997525 PMCID: PMC7877965 DOI: 10.1080/07853890.2020.1831050] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/25/2020] [Indexed: 12/24/2022] Open
Abstract
In chronic obstructive pulmonary disease (COPD) patients, bacterial and viral infections play a relevant role in worsening lung function and, therefore, favour disease progression. The inflammatory response to lung infections may become a specific indication of the bacterial and viral infections. We here review data on the bacterial-viral infections and related airways and lung parenchyma inflammation in stable and exacerbated COPD, focussing our attention on the prevalent molecular pathways in these different clinical conditions. The roles of macrophages, autophagy and NETosis are also briefly discussed in the context of lung infections in COPD. Controlling their combined response may restore a balanced lung homeostasis, reducing the risk of lung function decline. KEY MESSAGE Bacteria and viruses can influence the responses of the innate and adaptive immune system in the lung of chronic obstructive pulmonary disease (COPD) patients. The relationship between viruses and bacterial colonization, and the consequences of the imbalance of these components can modulate the inflammatory state of the COPD lung. The complex actions involving immune trigger cells, which activate innate and cell-mediated inflammatory responses, could be responsible for the clinical consequences of irreversible airflow limitation, lung remodelling and emphysema in COPD patients.
Collapse
Affiliation(s)
| | - Mauro Maniscalco
- Divisione di Pneumologia, Istituti Clinici Scientifici Maugeri, IRCCS, Telese, Italy
| | - Francesco Cappello
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica avanzata (BIND), Istituto di Anatomia Umana e Istologia Università degli Studi di Palermo, Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | - Mauro Carone
- UOC Pulmonology and Pulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri, IRCCS di Bari, Bari, Italy
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council, Pozzuoli, Italy
| | - Bruno Balbi
- Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell’Apparato Cardio Respiratorio, Istituti Clinici Scientifici Maugeri, IRCCS, Veruno, Italy
| | - Fabio L. M. Ricciardolo
- Dipartimento di Scienze Cliniche e Biologiche, Università di Torino, AOU San Luigi Gonzaga, Torino, Italy
| | - Gaetano Caramori
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini morfologiche e funzionali (BIOMORF), Università degli studi di Messina, Italy
| | - Antonino Di Stefano
- Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell’Apparato Cardio Respiratorio, Istituti Clinici Scientifici Maugeri, IRCCS, Veruno, Italy
| |
Collapse
|
17
|
Lea S, Gaskell R, Hall S, Maschera B, Hessel E, Singh D. Assessment of bacterial exposure on phagocytic capability and surface marker expression of sputum macrophages and neutrophils in COPD patients. Clin Exp Immunol 2021; 206:99-109. [PMID: 34143447 PMCID: PMC8446400 DOI: 10.1111/cei.13638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 11/28/2022] Open
Abstract
Defective phagocytosis has been shown in chronic obstructive pulmonary disease (COPD) bronchoalveolar lavage and blood monocyte-derived macrophages. Phagocytic capabilities of sputum macrophages and neutrophils in COPD are unknown. We investigated phagocytosis in these cells from COPD patients and controls. Phagocytosis of Streptococcus pneumoniae or fluorescently labelled non-typeable Haemophilus influenzae (NTHi) by sputum macrophages and neutrophils was determined by gentamycin protection assay (COPD; n = 5) or flow cytometry in 14 COPD patients, 8 healthy smokers (HS) and 9 healthy never-smokers (HNS). Sputum macrophages and neutrophils were differentiated by adherence for the gentamycin protection assay or receptor expression (CD206 and CD66b, respectively), by flow cytometry. The effects of NTHi on macrophage expression of CD206 and CD14 and neutrophil expression of CD16 were determined by flow cytometry. There was greater uptake of S. pneumoniae [~10-fold more colony-forming units (CFU)/ml] by sputum neutrophils compared to macrophages in COPD patients. Flow cytometry showed greater NTHi uptake by neutrophils compared to macrophages in COPD (67 versus 38%, respectively) and HS (61 versus 31%, respectively). NTHi uptake by macrophages was lower in HS (31%, p = 0.019) and COPD patients (38%, p = 0.069) compared to HNS (57%). NTHi uptake by neutrophils was similar between groups. NTHi exposure reduced CD206 and CD14 expression on macrophages and CD16 expression on neutrophils. Sputum neutrophils showed more phagocytic activity than macrophages. There was some evidence that bacterial phagocytosis was impaired in HS sputum macrophages, but no impairment of neutrophils was observed in HS or COPD patients. These results highlight the relative contributions of neutrophils and macrophages to bacterial clearance in COPD.
Collapse
Affiliation(s)
- Simon Lea
- Division of Infection, Immunity and Respiratory MedicineSchool of Biological SciencesFaculty of Biology, Medicine and HealthManchester Academic Health Science CentreThe University of ManchesterManchesterUK
| | - Rosemary Gaskell
- Division of Infection, Immunity and Respiratory MedicineSchool of Biological SciencesFaculty of Biology, Medicine and HealthManchester Academic Health Science CentreThe University of ManchesterManchesterUK
| | - Simon Hall
- Adaptive Immunity Research Unit (AI RU)GlaxoSmithKlineStevenageUK
| | - Barbara Maschera
- Adaptive Immunity Research Unit (AI RU)GlaxoSmithKlineStevenageUK
| | - Edith Hessel
- Refractory Respiratory Inflammation‐DPU (RRI DPU)GlaxoSmithKlineStevenageUK
| | - Dave Singh
- Division of Infection, Immunity and Respiratory MedicineSchool of Biological SciencesFaculty of Biology, Medicine and HealthManchester Academic Health Science CentreThe University of ManchesterManchesterUK
- Medicines Evaluation UnitManchester University NHS Foundation TrustManchesterUK
| |
Collapse
|
18
|
Beech A, Lea S, Li J, Jackson N, Mulvanny A, Singh D. Airway Bacteria Quantification Using Polymerase Chain Reaction Combined with Neutrophil and Eosinophil Counts Identifies Distinct COPD Endotypes. Biomedicines 2021; 9:1337. [PMID: 34680454 PMCID: PMC8533560 DOI: 10.3390/biomedicines9101337] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) inflammatory endotypes are associated with different airway microbiomes. We used quantitative polymerase chain reaction (qPCR) analysis of sputum samples to establish the bacterial load upper limit in healthy controls; these values determined the bacterial colonisation prevalence in a longitudinal COPD cohort. Bacteriology combined with sputum inflammatory cells counts were used to investigate COPD endotypes. METHODS Sixty COPD patients and 15 healthy non-smoking controls were recruited. Sputum was analysed by qPCR (for Haemophilus influenzae, Moraxella catarrhalis, Streptococcus pneumoniae and Psuedomonas aeruginosa) and sputum differential cell counts at baseline and 6 months. RESULTS At baseline and 6 months, 23.1% and 25.6% of COPD patients were colonised with H. influenzae, while colonisation with other bacterial species was less common, e.g., S. pneumoniae-1.9% and 5.1%, respectively. H. influenzae + ve patients had higher neutrophil counts at baseline (90.1% vs. 67.3%, p < 0.01), with similar results at 6 months. COPD patients with sputum eosinophil counts ≥3% at ≥1 visit rarely showed bacterial colonisation. CONCLUSIONS The prevalence of H. influenzae colonisation was approximately 25%, with low colonisation for other bacterial species. H. influenzae colonisation was associated with sputum neutrophilia, while eosinophilic inflammation and H. influenzae colonisation rarely coexisted.
Collapse
Affiliation(s)
- Augusta Beech
- Manchester Academic Health Science Centre, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (S.L.); (J.L.); (A.M.); (D.S.)
- Medicines Evaluation Unit, Manchester University NHS Foundation Trust, Manchester M23 9QZ, UK;
| | - Simon Lea
- Manchester Academic Health Science Centre, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (S.L.); (J.L.); (A.M.); (D.S.)
| | - Jian Li
- Manchester Academic Health Science Centre, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (S.L.); (J.L.); (A.M.); (D.S.)
| | - Natalie Jackson
- Medicines Evaluation Unit, Manchester University NHS Foundation Trust, Manchester M23 9QZ, UK;
| | - Alex Mulvanny
- Manchester Academic Health Science Centre, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (S.L.); (J.L.); (A.M.); (D.S.)
- Medicines Evaluation Unit, Manchester University NHS Foundation Trust, Manchester M23 9QZ, UK;
| | - Dave Singh
- Manchester Academic Health Science Centre, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (S.L.); (J.L.); (A.M.); (D.S.)
- Medicines Evaluation Unit, Manchester University NHS Foundation Trust, Manchester M23 9QZ, UK;
| |
Collapse
|
19
|
Morissette M, Godbout K, Côté A, Boulet LP. Asthma COPD overlap: Insights into cellular and molecular mechanisms. Mol Aspects Med 2021; 85:101021. [PMID: 34521557 DOI: 10.1016/j.mam.2021.101021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 11/16/2022]
Abstract
Although there is still no consensus on the definition of Asthma-COPD Overlap (ACO), it is generally accepted that some patients with airway disease have features of both asthma and COPD. Just as its constituents, ACO consists of different phenotypes, possibly depending on the predominance of the underlying asthma or COPD-associated pathophysiological mechanisms. The clinical picture is influenced by the development of airway inflammatory processes either eosinophilic, neutrophilic or mixed, in addition to glandular changes leading to mucus hypersecretion and a variety of other airway structural changes. Although animal models have exposed how smoking-related changes can interact with those observed in asthma, much remains to be known about their interactions in humans and the additional modulating effects of environmental exposures. There is currently no solid evidence to establish the optimal treatment of ACO but it should understandably include an avoidance of environmental triggers such as smoking and relevant allergens. The recognition and targeting of "treatable traits" following phenotyping is a pragmatic approach to select the optimal pharmacological treatment for ACO, although an association of inhaled corticosteroids and bronchodilators is always required in these patients. This association acts both as an anti-inflammatory treatment for the asthma component and as a functional antagonist for the airway remodeling features. Research should be promoted on well phenotyped subgroups of ACO patients to determine their optimal management.
Collapse
Affiliation(s)
- Mathieu Morissette
- Quebec Heart and Lung Institute - Université Laval, Canada; Department of Medicine, Université Laval, Québec, Canada.
| | - Krystelle Godbout
- Quebec Heart and Lung Institute - Université Laval, Canada; Department of Medicine, Université Laval, Québec, Canada
| | - Andréanne Côté
- Quebec Heart and Lung Institute - Université Laval, Canada; Department of Medicine, Université Laval, Québec, Canada
| | - Louis-Philippe Boulet
- Quebec Heart and Lung Institute - Université Laval, Canada; Department of Medicine, Université Laval, Québec, Canada.
| |
Collapse
|
20
|
Norwitz NG, Winwood R, Stubbs BJ, D'Agostino DP, Barnes PJ. Case Report: Ketogenic Diet Is Associated With Improvements in Chronic Obstructive Pulmonary Disease. Front Med (Lausanne) 2021; 8:699427. [PMID: 34395478 PMCID: PMC8358145 DOI: 10.3389/fmed.2021.699427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/21/2021] [Indexed: 11/15/2022] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a debilitating inflammatory respiratory condition that presents with worsening breathing difficulties and it is assumed to be progressive and incurable. As an inflammatory disease, COPD is associated with recruitment of immune cells to lung tissue and increased levels of pro-inflammatory cytokines, including TNF-α, IL-1β, IL-6, IL-8, and GM-CSF. Low-carbohydrate ketogenic diets have anti-inflammatory properties that could, in theory, improve COPD symptoms and progression. Herein, we report on a 54-year-old patient (C.A.) with COPD who adopted a ketogenic diet (70% calories from fat). Subsequently, C.A. experienced a reduction in inflammatory markers in association with a meaningful improvement in lung function. His inflammatory markers decreased into the normal range and his forced expiratory volume increased by 37.5% relative to its pre-ketogenic diet value. Future research should explore nutritional ketosis and ketogenic diets as possible therapeutic options for individuals with COPD.
Collapse
Affiliation(s)
- Nicholas G Norwitz
- Department of Nutrition, Harvard Medical School, Boston, MA, United States
| | - Russell Winwood
- Respiratory Network, Ministry of Health Agency for Clinical Innovation, St Leonards, NSW, Australia
| | | | - Dominic P D'Agostino
- Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Institute for Human and Machine Cognition, Pensacola, FL, United States
| | - Peter J Barnes
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| |
Collapse
|
21
|
Wang Z, Locantore N, Haldar K, Ramsheh MY, Beech AS, Ma W, Brown JR, Tal-Singer R, Barer MR, Bafadhel M, Donaldson GC, Wedzicha JA, Singh D, Wilkinson TMA, Miller BE, Brightling CE. Inflammatory Endotype-associated Airway Microbiome in Chronic Obstructive Pulmonary Disease Clinical Stability and Exacerbations: A Multicohort Longitudinal Analysis. Am J Respir Crit Care Med 2021; 203:1488-1502. [PMID: 33332995 PMCID: PMC8483235 DOI: 10.1164/rccm.202009-3448oc] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/17/2020] [Indexed: 12/18/2022] Open
Abstract
Rationale: Understanding the role of the airway microbiome in chronic obstructive pulmonary disease (COPD) inflammatory endotypes may help to develop microbiome-based diagnostic and therapeutic approaches. Objectives: To understand the association of the airway microbiome with neutrophilic and eosinophilic COPD at stability and during exacerbations. Methods: An integrative analysis was performed on 1,706 sputum samples collected longitudinally from 510 patients with COPD recruited at four UK sites of the BEAT-COPD (Biomarkers to Target Antibiotic and Systemic COPD), COPDMAP (Chronic Obstructive Pulmonary Disease Medical Research Council/Association of the British Pharmaceutical Industry), and AERIS (Acute Exacerbation and Respiratory Infections in COPD) cohorts. The microbiome was analyzed using COPDMAP and AERIS as a discovery data set and BEAT-COPD as a validation data set. Measurements and Main Results: The airway microbiome in neutrophilic COPD was heterogeneous, with two primary community types differentiated by the predominance of Haemophilus. The Haemophilus-predominant subgroup had elevated sputum IL-1β and TNFα (tumor necrosis factor α) and was relatively stable over time. The other neutrophilic subgroup with a balanced microbiome profile had elevated sputum and serum IL-17A and was temporally dynamic. Patients in this state at stability were susceptible to the greatest microbiome shifts during exacerbations. This subgroup can temporally switch to both neutrophilic Haemophilus-predominant and eosinophilic states that were otherwise mutually exclusive. Time-series analysis on the microbiome showed that the temporal trajectories of Campylobacter and Granulicatella were indicative of intrapatient switches from neutrophilic to eosinophilic inflammation, in track with patient sputum eosinophilia over time. Network analysis revealed distinct host-microbiome interaction patterns among neutrophilic Haemophilus-predominant, neutrophilic balanced microbiome, and eosinophilic subgroups. Conclusions: The airway microbiome can stratify neutrophilic COPD into subgroups that justify different therapies. Neutrophilic and eosinophilic COPD are interchangeable in some patients. Monitoring temporal variability of the airway microbiome may track patient inflammatory status over time.
Collapse
Affiliation(s)
- Zhang Wang
- Institute of Ecological Sciences, School of Life Sciences, South China Normal University, Guangzhou, China
| | | | - Koirobi Haldar
- Human Genetics, Research and Development, GlaxoSmithKline, Collegeville, Pennsylvania
| | | | - Augusta S. Beech
- Department of Respiratory Sciences, Institute for Lung Health, Leicester National Institute for Health Research Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| | - Wei Ma
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - James R. Brown
- Institute of Statistics and Big Data, Renmin University of China, Beijing, China
| | - Ruth Tal-Singer
- Chronic Obstructive Pulmonary Disease Foundation, Research Department, Washington, District of Columbia
| | - Michael R. Barer
- Human Genetics, Research and Development, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Mona Bafadhel
- Respiratory Medicine Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Gavin C. Donaldson
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Jadwiga A. Wedzicha
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Dave Singh
- Department of Respiratory Sciences, Institute for Lung Health, Leicester National Institute for Health Research Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| | - Tom M. A. Wilkinson
- National Institute for Health Research Southampton Respiratory Biomedical Research Unit, University Hospital Southampton National Health Service Foundation Trust, Southampton, United Kingdom
| | | | | |
Collapse
|
22
|
Variations in fecal microbial profiles of acute exacerbations and stable chronic obstructive pulmonary disease. Life Sci 2020; 265:118738. [PMID: 33181175 DOI: 10.1016/j.lfs.2020.118738] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/28/2020] [Accepted: 11/05/2020] [Indexed: 02/05/2023]
Abstract
AIM Alterations in the respiratory and digestive tract microbiomes influence the occurrence and progression of chronic obstructive pulmonary disease (COPD). Here, we aimed to identify fecal microbiome profiles during COPD development. METHODS Fecal samples were collected from 29 COPD patients with acute exacerbation (AECOPD), 29 stable COPD patients, and 22 normal subjects (NS). The fecal microbial profiles were obtained using 16S rRNA gene sequencing. KEY FINDINGS The diversity and richness were lower and fewer variations in the taxonomic composition of fecal microbiota were observed in AECOPD patients than in stable COPD and NS. The relative abundances of Firmicutes and Actinobacteria were decreased, while those of Bacteroidetes and Proteobacteria were increased in AECOPD compared to COPD and NS. Among the top ten genera, the proportions of Lachnoclostridium and Parabacteroides significantly increased in AECOPD, whereas those of other genera decreased. Discriminative bacteria, such as p_Bacteroidetes, c_Bacteroidia, o_Bacteroidales, Lactobacillales, and Proteobacteria, were identified in AECOPD compared to stable COPD and NS. The weighted gene co-expression networks showed that Firmicutes and Actinobacteria were the main hub bacterial taxa related with lung function (FEV1% and FEV1/FVC%) and inflammatory indices (TNF-α, IL-6, IL-8, PCT, and CRP). SIGNIFICANCE These findings emphasized the changes in the abundance and composition of the fecal microbiome in stable COPD and AECOPD. Variations in fecal microbiota may be associated with COPD progression.
Collapse
|
23
|
Beech AS, Lea S, Kolsum U, Wang Z, Miller BE, Donaldson GC, Wedzicha JA, Brightling CE, Singh D. Bacteria and sputum inflammatory cell counts; a COPD cohort analysis. Respir Res 2020; 21:289. [PMID: 33131502 PMCID: PMC7603729 DOI: 10.1186/s12931-020-01552-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/20/2020] [Indexed: 01/08/2023] Open
Abstract
Background There is evidence that bacterial colonisation in chronic obstructive pulmonary disease (COPD) is associated with increased neutrophilic airway inflammation. This study tested the hypothesis that different bacterial phyla and species cause different inflammatory profiles in COPD patients. Methods Sputum was analysed by quantitative polymerase chain reaction (qPCR) to quantify bacterial load and 16S rRNA gene sequencing to identify taxonomic composition. Sputum differential cell counts (DCC) and blood DCC were obtained at baseline and 6 months. Patients were categorised into five groups based on bacterial load defined by genome copies/ml of ≥ 1 × 104, no colonisation and colonisation by Haemophilus influenzae (H. influenzae), Moraxella catarrhalis (M. catarrhalis), Streptococcus pneumoniae (S. pneumoniae), or > 1 potentially pathogenic microorganism (PPM). Results We observed an increase in sputum neutrophil (%), blood neutrophil (%) and neutrophil–lymphocyte ratio (NLR) in patients colonised with H. influenzae (82.6, 67.1, and 3.29 respectively) compared to those without PPM colonisation at baseline (69.5, 63.51 and 2.56 respectively) (p < 0.05 for all analyses), with similar findings at 6 months. The bacterial load of H. influenzae and Haemophilus determined by qPCR and 16s rRNA gene sequencing respectively, and sputum neutrophil % were positively correlated between baseline and 6 months visits (p < 0.0001, 0.0150 and 0.0002 with r = 0.53, 0.33 and 0.44 respectively). Conclusions These results demonstrate a subgroup of COPD patients with persistent H. influenzae colonisation that is associated with increased airway and systemic neutrophilic airway inflammation, and less eosinophilic airway inflammation.
Collapse
Affiliation(s)
- Augusta S Beech
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK. .,Medicines Evaluation Unit, University of Manchester, Manchester University NHS Foundation Trust, Southmoor Road, Manchester, M23 9QZ, UK.
| | - Simon Lea
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Umme Kolsum
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Zhang Wang
- Institute of Ecological Sciences, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Bruce E Miller
- Medical Innovation, Value Evidence and Outcomes, GSK R&D, Collegeville, PA, USA
| | - Gavin C Donaldson
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | - Dave Singh
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.,Medicines Evaluation Unit, University of Manchester, Manchester University NHS Foundation Trust, Southmoor Road, Manchester, M23 9QZ, UK
| |
Collapse
|
24
|
Consensus document on the diagnosis and treatment of chronic bronchial infection in chronic obstructive pulmonary disease. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.arbr.2020.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
25
|
de la Rosa Carrillo D, López-Campos JL, Alcázar Navarrete B, Calle Rubio M, Cantón Moreno R, García-Rivero JL, Máiz Carro L, Olveira Fuster C, Martínez-García MÁ. Consensus Document on the Diagnosis and Treatment of Chronic Bronchial Infection in Chronic Obstructive Pulmonary Disease. Arch Bronconeumol 2020; 56:651-664. [PMID: 32540279 DOI: 10.1016/j.arbres.2020.04.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/22/2020] [Accepted: 04/01/2020] [Indexed: 02/07/2023]
Abstract
Although the chronic presence of microorganisms in the airways of patients with stable chronic obstructive pulmonary disease (COPD) confers a poor outcome, no recommendations have been established in disease management guidelines on how to diagnose and treat these cases. In order to guide professionals, the Spanish Society of Pulmonology and Thoracic Surgery (SEPAR) has prepared a document which aims to answer questions on the clinical management of COPD patients in whom microorganisms are occasionally or habitually isolated. Since the available scientific evidence is too heterogeneous to use in the creation of a clinical practice guideline, we have drawn up a document based on existing scientific literature and clinical experience, addressing the definition of different clinical situations and their diagnosis and management. The text was drawn up by consensus and approved by a large group of respiratory medicine experts with extensive clinical and scientific experience in the field, and has been endorsed by the SEPAR Scientific Committee.
Collapse
Affiliation(s)
| | - José Luís López-Campos
- Servicio de Neumología, Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Sevilla, España; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, España
| | - Bernardino Alcázar Navarrete
- Servicio de Neumología, Hospital Regional Universitario de Málaga. Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, España
| | - Myriam Calle Rubio
- Servicio de Neumología, Hospital de Alta Resolución de Loja, Loja, Granada, España
| | - Rafael Cantón Moreno
- Servicio de Neumología, Unidad de Infección Bronquial Crónica, Fibrosis Quística y Bronquiectasias, Hospital Universitario Ramón y Cajal, Madrid, España
| | - Juan Luis García-Rivero
- Servicio de Neumología, Hospital Clínico San Carlos. Departamento de Medicina, Facultad de Medicina, UCM, Madrid, España
| | - Luís Máiz Carro
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal. Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, España
| | | | | |
Collapse
|
26
|
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) is a heterogeneous syndrome and may comprise several different phenotypes that are driven by different molecular mechanisms (endotypes). Several different clinical, genetic, and inflammatory phenotypes of COPD have been recognized and this may lead to more precise effective therapies. AREAS COVERED The different clinical phenotypes, including smoking versus nonsmoking COPD, small airway disease versus emphysema, non-exacerbators versus frequent exacerbators are discussed. Rare genetic endotypes (alpha1-antitrypsin deficiency, telomerase polymorphisms), and inflammatory phenotypes (eosinophilic versus neutrophilic) are also recognized in stable and exacerbating patients and have implications for the choice of therapy. EXPERT OPINION Clinical phenotypes have so far not proved to be very useful in selecting more personalized therapy for COPD. Even with genetic endotypes, this has not led to improved therapy. More promising is the recognition that COPD patients who have increased sputum or blood eosinophils tend to have more frequent exacerbations and inhaled corticosteroids are more effective in preventing exacerbation. Increased blood eosinophils have proved to be a useful biomarker now used to target ICS more effectively. Furthermore, COPD patients with low eosinophils are more likely to get pneumonia with ICS and to have lower airway bacterial colonization.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart and Lung Institute, Imperial College London , London, UK
| |
Collapse
|
27
|
Finney LJ, Belchamber KBR, Fenwick PS, Kemp SV, Edwards MR, Mallia P, Donaldson G, Johnston SL, Donnelly LE, Wedzicha JA. Human Rhinovirus Impairs the Innate Immune Response to Bacteria in Alveolar Macrophages in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2020; 199:1496-1507. [PMID: 30562053 DOI: 10.1164/rccm.201806-1095oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Rationale: Human rhinovirus (HRV) is a common cause of chronic obstructive pulmonary disease (COPD) exacerbations. Secondary bacterial infection is associated with more severe symptoms and delayed recovery. Alveolar macrophages clear bacteria from the lung and maintain lung homeostasis through cytokine secretion. These processes are defective in COPD. The effect of HRV on macrophage function is unknown. Objectives: To investigate the effect of HRV on phagocytosis and cytokine response to bacteria by alveolar macrophages and monocyte-derived macrophages (MDM) in COPD and healthy control subjects. Methods: Alveolar macrophages were obtained by bronchoscopy and MDM by adherence. Macrophages were exposed to HRV16 (multiplicity of infection 5), polyinosinic:polycytidylic acid (poly I:C) 30 μg/ml, IFN-β 10 μg/ml, IFN-γ 10 μg/ml, or medium control for 24 hours. Phagocytosis of fluorescently labeled Haemophilus influenzae or Streptococcus pneumoniae was assessed by fluorimetry. CXCL8 (IL-8), IL-6, TNF-α (tumor necrosis factor-α), and IL-10 release was measured by ELISA. Measurements and Main Results: HRV significantly impaired phagocytosis of H. influenzae by 23% in MDM (n = 37; P = 0.004) and 18% in alveolar macrophages (n = 20; P < 0.0001) in COPD. HRV also significantly reduced phagocytosis of S. pneumoniae by 33% in COPD MDM (n = 20; P = 0.0192). There was no effect in healthy control subjects. Phagocytosis of H. influenzae was also impaired by poly I:C but not IFN-β or IFN-γ in COPD MDM. HRV significantly reduced cytokine responses to H. influenzae. The IL-10 response to H. influenzae was significantly impaired by poly I:C, IFN-β, and IFN-γ in COPD cells. Conclusions: HRV impairs phagocytosis of bacteria in COPD, which may lead to an outgrowth of bacteria. HRV also impairs cytokine responses to bacteria via the TLR3/IFN pathway, which may prevent resolution of inflammation leading to prolonged exacerbations in COPD.
Collapse
Affiliation(s)
- Lydia J Finney
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Kylie B R Belchamber
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Peter S Fenwick
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Samuel V Kemp
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and.,2 Royal Brompton Hospital, London, United Kingdom
| | - Michael R Edwards
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Patrick Mallia
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Gavin Donaldson
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Sebastian L Johnston
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Louise E Donnelly
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Jadwiga A Wedzicha
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| |
Collapse
|
28
|
Sibila O, Laserna E, Shoemark A, Keir HR, Finch S, Rodrigo-Troyano A, Perea L, Lonergan M, Goeminne PC, Chalmers JD. Airway Bacterial Load and Inhaled Antibiotic Response in Bronchiectasis. Am J Respir Crit Care Med 2020; 200:33-41. [PMID: 31109172 DOI: 10.1164/rccm.201809-1651oc] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Rationale: The principal underlying inhaled antibiotic treatment in bronchiectasis is that airway bacterial load drives inflammation, and therefore antibiotic treatment will reduce symptoms. Objectives: To determine the relationship between bacterial load and clinical outcomes, assess the stability of bacterial load over time, and test the hypothesis that response to inhaled antibiotics would be predicted by baseline bacterial load. Methods: We performed three studies. Studies 1 and 2 were prospective studies including adults with bronchiectasis. Study 3 was a post hoc analysis of a randomized trial of inhaled aztreonam. A priori patients were divided into low (<105 cfu/g), moderate (105-106 cfu/g), and high bacterial load (≥107 cfu/g) using quantitative sputum culture. Measurements and Main Results: Bacterial load was a stable trait associated with worse quality of life and more airway inflammation in studies 1, 2, and 3. In study 3, patients with high bacterial load showed an improvement in the primary endpoint (Quality of Life-Bronchiectasis-Respiratory Symptoms Score at Week 4) in favor of aztreonam (mean difference of 9.7 points; 95% confidence interval, 3.4-16.0; P = 0.003). The proportion of patients who achieved an increase above the minimum clinically important difference was higher in the aztreonam group at Week 4 (63% vs. 37%; P = 0.01) and at Week 12 (62% vs. 38%; P = 0.01) only in high bacterial load patients. Conclusions: Improvement of quality of life with inhaled aztreonam was only evident in patients with high bacterial load. Bacterial load may be a useful biomarker of severity of disease and treatment response.
Collapse
Affiliation(s)
- Oriol Sibila
- 1 Respiratory Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,2 Biomedical Research Institute Sant Pau, Barcelona, Spain
| | - Elena Laserna
- 3 Hospital Comarcal de Mollet, Mollet del Vallés, Spain
| | - Amelia Shoemark
- 4 Scottish Centre for Respiratory Medicine, University of Dundee, Dundee, United Kingdom
| | - Holly R Keir
- 4 Scottish Centre for Respiratory Medicine, University of Dundee, Dundee, United Kingdom
| | - Simon Finch
- 4 Scottish Centre for Respiratory Medicine, University of Dundee, Dundee, United Kingdom
| | - Ana Rodrigo-Troyano
- 1 Respiratory Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,2 Biomedical Research Institute Sant Pau, Barcelona, Spain
| | - Lidia Perea
- 2 Biomedical Research Institute Sant Pau, Barcelona, Spain
| | - Mike Lonergan
- 4 Scottish Centre for Respiratory Medicine, University of Dundee, Dundee, United Kingdom
| | - Pieter C Goeminne
- 5 Department of Respiratory Medicine, AZ Nikolaas, Sint-Niklaas, Belgium; and.,6 Department of Respiratory Medicine, UZ Leuven, Leuven, Belgium
| | - James D Chalmers
- 4 Scottish Centre for Respiratory Medicine, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
29
|
Lung Macrophage Functional Properties in Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2020; 21:ijms21030853. [PMID: 32013028 PMCID: PMC7037150 DOI: 10.3390/ijms21030853] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is caused by the chronic exposure of the lungs to toxic particles and gases. These exposures initiate a persistent innate and adaptive immune inflammatory response in the airways and lung tissues. Lung macrophages (LMs) are key innate immune effector cells that identify, engulf, and destroy pathogens and process inhaled particles, including cigarette smoke and particulate matter (PM), the main environmental triggers for COPD. The number of LMs in lung tissues and airspaces is increased in COPD, suggesting a potential key role for LMs in initiating and perpetuating the chronic inflammatory response that underpins the progressive nature of COPD. The purpose of this brief review is to discuss the origins of LMs, their functional properties (chemotaxis, recruitment, mediator production, phagocytosis and apoptosis) and changes in these properties due to exposure to cigarette smoke, ambient particulate and pathogens, as well as their persistent altered functional properties in subjects with established COPD. We also explore the potential to therapeutically modulate and restore LMs functional properties, to improve impaired immune system, prevent the progression of lung tissue destruction, and improve both morbidity and mortality related to COPD.
Collapse
|
30
|
Bu T, Wang LF, Yin YQ. How Do Innate Immune Cells Contribute to Airway Remodeling in COPD Progression? Int J Chron Obstruct Pulmon Dis 2020; 15:107-116. [PMID: 32021149 PMCID: PMC6966950 DOI: 10.2147/copd.s235054] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
Recently, the therapeutic potential of immune-modulation during the progression of chronic obstructive pulmonary disease (COPD) has been attracting increasing interest. However, chronic inflammatory response has been over-simplified in descriptions of the mechanism of COPD progression. As a form of first-line airway defense, epithelial cells exhibit phenotypic alteration, and participate in epithelial layer disorganization, mucus hypersecretion, and extracellular matrix deposition. Dendritic cells (DCs) exhibit attenuated antigen-presenting capacity in patients with advanced COPD. Immature DCs migrate into small airways, where they promote a pro-inflammatory microenvironment and bacterial colonization. In response to damage-associated molecular patterns (DAMPs) in lung tissue affected by COPD, neutrophils are excessively recruited and activated, where they promote a proteolytic microenvironment and fibrotic repair in small airways. Macrophages exhibit decreased phagocytosis in the large airways, while they demonstrate high pro-inflammatory potential in the small airways, and mediate alveolar destruction and chronic airway inflammation. Natural killer T (NKT) cells, eosinophils, and mast cells also play supplementary roles in COPD progression; however, their cellular activities are not yet entirely clear. Overall, during COPD progression, “exhausted” innate immune responses can be observed in the large airways. On the other hand, the innate immune response is enhanced in the small airways. Approaches that inhibit the inflammatory cascade, chemotaxis, or the activation of inflammatory cells could possibly delay the progression of airway remodeling in COPD, and may thus have potential clinical significance.
Collapse
Affiliation(s)
- Tegeleqi Bu
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Li Fang Wang
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Yi Qing Yin
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, People's Republic of China
| |
Collapse
|
31
|
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: 36] [Impact Index Per Article: 6.0] [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
|
32
|
Wang JX, Li HQ, Zhang F, Ning W. Systemic inflammation and the effects of short-term antibiotic treatment for PPM positive patients with stable COPD. Int J Chron Obstruct Pulmon Dis 2019; 14:1923-1932. [PMID: 31692553 PMCID: PMC6711567 DOI: 10.2147/copd.s217971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 08/12/2019] [Indexed: 11/23/2022] Open
Abstract
Objective To evaluate patients with stable COPD for the presence of potentially pathogenic microorganisms (PPM), systemic inflammation and the effects of short-term antibiotic therapy in PPM positive patients. Methods From January 2016 to June 2017, we enrolled 96 stable COPD patients. Bacterial cultures from sputum collections were quantitated, along with markers for systemic inflammation including serum C-reactive protein (CRP), interleukin-8 (IL-8) and plasma fibrinogen (FIB) in all patients. All enrolled patients were followed for 12 months. Forty patients were identified as PPM positive and were randomly divided into an antibiotic group and a control group. The antibiotic group was treated with moxifloxacin orally for 6 days. Lung function and markers for systemic inflammation were repeatedly measured at 30 days and 6 months in PPM positive subjects. Results Binary logistic regression analysis showed that risk factors for PPM positive are bronchiectasis (OR 4.18, 95% CI 1.20-14.59; P=0.025), COPD assessment test (CAT) ≥20 (OR 17.55, 95% CI 2.82-109.18; P=0.002), spontaneous sputum (OR 15.09, 95% CI 1.36-168.02; P=0.027) and sputum purulence (OR 38.43, 95% CI 5.39-274.21; P=0.000). CRP and IL-8 were higher in PPM positive group than those in PPM negative group (P=0.001, P=0.007, respectively), but there were no differences of FIB between the two groups (P=0.086). Compared to the PPM negative group, the rate of acute exacerbation of COPD was higher (P=0.029) and time to next acute exacerbation was shorter (P=0.030) in PPM positive group. There were no differences in lung function and systemic inflammatory markers either in the control group or the antibiotic group at different time points of follow-up. Conclusion PPM exists in stable COPD patients and can cause systemic inflammation and is associated with acute exacerbation of COPD. Short-term antibiotic therapy had no effect on systemic inflammation nor on acute exacerbation of COPD.China Clinical Trials Registry: ChiCTR-IOR-15006769.
Collapse
Affiliation(s)
- Jin-Xiang Wang
- Department of Pulmonary and Critical Care Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hui-Qiao Li
- Department of Pulmonary and Critical Care Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Fang Zhang
- Department of Clinical Laboratory, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wei Ning
- Pulmonary Function Test Room, Department of Pulmonary and Critical Care Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
33
|
The lung microbiome dynamics between stability and exacerbation in chronic obstructive pulmonary disease (COPD): Current perspectives. Respir Med 2019; 157:1-6. [PMID: 31450162 DOI: 10.1016/j.rmed.2019.08.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 12/13/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disorder with a course that is not uniform for all COPD patients. Although smoking is considered as the major cause of the disease, persistent or recurrent infections seem to play a particular role in the disease establishment and progression. COPD is characterized by dysregulated immunity that has been associated with the bacterial colonization and infections. The establishment of culture-independent techniques has shed new light on the relationships between bacterial ecology and health status and expanded our knowledge on the lung microbiome. Interactions between the host and lung microbiome result in inflammation and activation of resident cells. The lung microbiome contains populations of symbionts and pathobionts in balance which lose their equilibrium and disturb the balance of T-helper and regulatory T-cells (Treg) upon infection, or lung disease. In COPD factors such as disease severity, exacerbations, degree of inflammation, and type of treatment used (e.g inhaled or systemic steroids and antibiotics) affect the composition of lung microbiota. Recent data indicate that the presence of specific bacterial taxa in the airways has the potential to influence the host immune response and possibly to interfere with disease phenotype. Although, there is a growing body of evidence for the role of microbiome in COPD several unanswered questions still exist for its clinical relevance.
Collapse
|
34
|
Sapey E, Bafadhel M, Bolton CE, Wilkinson T, Hurst JR, Quint JK. Building toolkits for COPD exacerbations: lessons from the past and present. Thorax 2019; 74:898-905. [PMID: 31273049 PMCID: PMC6824608 DOI: 10.1136/thoraxjnl-2018-213035] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/03/2019] [Accepted: 05/05/2019] [Indexed: 02/06/2023]
Abstract
In the nineteenth century, it was recognised that acute attacks of chronic bronchitis were harmful. 140 years later, it is clearer than ever that exacerbations of chronic obstructive pulmonary disease (ECOPD) are important events. They are associated with significant mortality, morbidity, a reduced quality of life and an increasing reliance on social care. ECOPD are common and are increasing in prevalence. Exacerbations beget exacerbations, with up to a quarter of in-patient episodes ending with readmission to hospital within 30 days. The healthcare costs are immense. Yet despite this, the tools available to diagnose and treat ECOPD are essentially unchanged, with the last new intervention (non-invasive ventilation) introduced over 25 years ago.An ECOPD is 'an acute worsening of respiratory symptoms that results in additional therapy'. This symptom and healthcare utility-based definition does not describe pathology and is unable to differentiate from other causes of an acute deterioration in breathlessness with or without a cough and sputum. There is limited understanding of the host immune response during an acute event and no reliable and readily available means to identify aetiology or direct treatment at the point of care (POC). Corticosteroids, short acting bronchodilators with or without antibiotics have been the mainstay of treatment for over 30 years. This is in stark contrast to many other acute presentations of chronic illness, where specific biomarkers and mechanistic understanding has revolutionised care pathways. So why has progress been so slow in ECOPD? This review examines the history of diagnosing and treating ECOPD. It suggests that to move forward, there needs to be an acceptance that not all exacerbations are alike (just as not all COPD is alike) and that clinical presentation alone cannot identify aetiology or stratify treatment.
Collapse
Affiliation(s)
- Elizabeth Sapey
- Birmingham Acute Care Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Mona Bafadhel
- Respiratory Medicine Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Charlotte Emma Bolton
- Respiratory Medicine, Nottingham Respiratory BRU, University of Nottingham, Nottingham, UK
| | - Thomas Wilkinson
- Clinical and Experimental Medicine, University of Southampton, Southampton, UK
| | - John R Hurst
- Academic Unit of Respiratory Medicine, UCL Medical School, London, UK
| | - Jennifer K Quint
- Respiratory Epidemiology, Occupational Medicine and Public Health, Imperial College London, London, UK
| |
Collapse
|
35
|
Abstract
Chronic obstructive pulmonary disease (COPD) is a major global health problem that is poorly treated by current therapies as it has proved difficult to treat the underlying inflammation, which is largely corticosteroid-resistant in most patients. Although rare genetic endotypes of COPD have been recognized, despite the clinical heterogeneity of COPD, it has proved difficult to identify distinct inflammatory endotypes. Most patients have increased neutrophils and macrophages in sputum, reflecting the increased secretion of neutrophil and monocyte chemotactic mediators in the lungs. However, some patients also have increased eosinophils in sputum and this may be reflected by increased blood eosinophils. Increased blood and sputum eosinophils are associated with more frequent exacerbations and predict a good response to corticosteroids in reducing and treating acute exacerbations. Eosinophilic COPD may represent an overlap with asthma but the mechanism of eosinophilia is uncertain as, although an increase in sputum IL-5 has been detected, anti-IL-5 therapies are not effective in preventing exacerbations. More research is needed to link inflammatory endotypes to clinical manifestations and outcomes in COPD and in particular to predict response to precision medicines.
Collapse
Affiliation(s)
- Peter J. Barnes
- National Heart and Lung Institute Imperial College London UK
| |
Collapse
|
36
|
Stockley RA. Corporate Memory and Rediscovering the Wheel. Am J Respir Crit Care Med 2019; 200:2-4. [PMID: 31109188 PMCID: PMC6603050 DOI: 10.1164/rccm.201901-0066ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Robert A. Stockley
- University Hospitals Birmingham NHS Foundation TrustQueen Elizabeth Hospital BirminghamBirmingham, United Kingdom
| |
Collapse
|
37
|
Sapey E, Stockley RA. Getting stuck or choosing to stay? Neutrophil transit times in the lung in acute inflammation and COPD. Thorax 2019; 74:631-632. [PMID: 31097614 PMCID: PMC6585287 DOI: 10.1136/thoraxjnl-2018-213000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2019] [Indexed: 01/31/2023]
Affiliation(s)
- Elizabeth Sapey
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Robert A Stockley
- Respiratory Medicine, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
38
|
Su YC, Jalalvand F, Thegerström J, Riesbeck K. The Interplay Between Immune Response and Bacterial Infection in COPD: Focus Upon Non-typeable Haemophilus influenzae. Front Immunol 2018; 9:2530. [PMID: 30455693 PMCID: PMC6230626 DOI: 10.3389/fimmu.2018.02530] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a debilitating respiratory disease and one of the leading causes of morbidity and mortality worldwide. It is characterized by persistent respiratory symptoms and airflow limitation due to abnormalities in the lower airway following consistent exposure to noxious particles or gases. Acute exacerbations of COPD (AECOPD) are characterized by increased cough, purulent sputum production, and dyspnea. The AECOPD is mostly associated with infection caused by common cold viruses or bacteria, or co-infections. Chronic and persistent infection by non-typeable Haemophilus influenzae (NTHi), a Gram-negative coccobacillus, contributes to almost half of the infective exacerbations caused by bacteria. This is supported by reports that NTHi is commonly isolated in the sputum from COPD patients during exacerbations. Persistent colonization of NTHi in the lower airway requires a plethora of phenotypic adaptation and virulent mechanisms that are developed over time to cope with changing environmental pressures in the airway such as host immuno-inflammatory response. Chronic inhalation of noxious irritants in COPD causes a changed balance in the lung microbiome, abnormal inflammatory response, and an impaired airway immune system. These conditions significantly provide an opportunistic platform for NTHi colonization and infection resulting in a "vicious circle." Episodes of large inflammation as the consequences of multiple interactions between airway immune cells and NTHi, accumulatively contribute to COPD exacerbations and may result in worsening of the clinical status. In this review, we discuss in detail the interplay and crosstalk between airway immune residents and NTHi, and their effect in AECOPD for better understanding of NTHi pathogenesis in COPD patients.
Collapse
Affiliation(s)
- Yu-Ching Su
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Farshid Jalalvand
- Department of Biology, Centre for Bacterial Stress Response and Persistence, University of Copenhagen, Copenhagen, Denmark
| | - John Thegerström
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Kristian Riesbeck
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
39
|
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
|
40
|
Yang X, Li H, Ma Q, Zhang Q, Wang C. Neutrophilic Asthma Is Associated with Increased Airway Bacterial Burden and Disordered Community Composition. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9230234. [PMID: 30105264 PMCID: PMC6076954 DOI: 10.1155/2018/9230234] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/14/2018] [Indexed: 12/31/2022]
Abstract
Neutrophilic asthma (NA) is an important asthma inflammatory phenotype associated with disease severity, airflow limitation, and steroid resistance, and its mechanism is still uncertain. Evidences suggest a potential role for bacteria in its pathogenesis, but, so far, this remains poorly understood. We sought to investigate airway bacterial burden, community composition, and inflammatory response in NA. Fifty-four stable asthmatics without infection were enrolled and separated into either NA group (n = 20) or non-NA group (n = 34). Subject demographics, Asthma Control Test (ACT) scores, medications, and pulmonary functions were documented. Sputum cytology, airway bacterial burden, microbial community composition, and inflammatory cytokines were assessed. The total airway bacterial burden was significantly increased in subjects with NA versus non-NA and was positively correlated with the sputum neutrophil percentage. Airway neutrophilia was associated with less airway bacterial community richness and diversity, along with a distinct community composition. In patients with NA, bacteria in phylum Proteobacteria, especially Haemophilus spp. and Moraxella spp., showed significant increases in both actual loads and relative abundances, while bacteria in phyla Firmicutes, Actinobacteria, and Saccharibacteria showed decreased relative abundances compared with non-NA. Patients with NA demonstrated higher levels of interleukin-1β (IL-1β), IL-6, IL-8, IL-12, IL-17A, and tumor necrosis factor-α (TNF-α) in sputum samples compared with non-NA. Increased bacterial burden and distinct microbiota composition were the key characters of neutrophilic phenotype in asthma, accompanied by excessive airway inflammation. Understanding the relationship between airway microbiota and neutrophilic inflammation may help in treatment and management of asthma, such as targeting airway microbiota.
Collapse
Affiliation(s)
- Xu Yang
- Institute of Respiratory Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Haining Li
- Institute of Respiratory Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Qianli Ma
- Institute of Respiratory Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Qiao Zhang
- Institute of Respiratory Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Changzheng Wang
- Institute of Respiratory Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| |
Collapse
|
41
|
Yang X, Wang Y, Zhao S, Wang R, Wang C. Long-term exposure to low-dose Haemophilus influenzae during allergic airway disease drives a steroid-resistant neutrophilic inflammation and promotes airway remodeling. Oncotarget 2018; 9:24898-24913. [PMID: 29861841 PMCID: PMC5982741 DOI: 10.18632/oncotarget.24653] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 01/14/2018] [Indexed: 12/19/2022] Open
Abstract
Growing evidences indicate that bacteria are associated with pathogenesis of neutrophilic asthma. However, the long-term effect of airway bacterial colonization remains unclear. We sought to establish a murine model to simulate the airway inflammation of long-term bacterial colonization, and to assess the effects of bacteria on allergic airway disease (AAD). BALB/c mice were sensitized twice and subsequently challenged with ovalbumin (OVA) and exposed to low-dose Haemophilus influenzae for approximately 2 months. Mice in treatment groups inhaled budesonide for consecutively 6 days in the last week. Airway inflammatory phenotype, immune response, phagocytic capacity, mucus production, airway remodeling and steroid sensitivity were assessed. Long-term exposure to low-dose H. influenzae during AAD did not cause serious infection but only a slightly increased airway inflammation, which resembled the colonization. Inflammatory phenotype was converted from a steroid-sensitive T helper (Th) 2-associated eosinophilic inflammation to a steroid-resistant Th17-associated neutrophilic inflammation. The increased neutrophilic inflammation was accompanied by defects in regulatory T cell (Treg)-associated immunosuppression and macrophage phagocytosis, and finally promoted mucus hypersecretion and airway remodeling. These features resembled those of refractory neutrophilic asthma in humans. These findings indicate that in asthmatic patients, airway bacterial colonization may be a potential therapeutic target. Minimizing the pathogen burden in airway, such as Haemophilus influenzae, may be beneficial.
Collapse
Affiliation(s)
- Xu Yang
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Yijie Wang
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China.,Department of Respiratory Medicine, The 305 Hospital of PLA, Beijing, 100017, China
| | - Shengtao Zhao
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China.,Department of Respiratory Medicine, Kunming General Hospital of Chengdu Military Region, Kunming, 650032, China
| | - Ran Wang
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Changzheng Wang
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| |
Collapse
|
42
|
Abstract
Asthma and COPD remain two diseases of the respiratory tract with unmet medical needs. This review considers the current state of play with respect to what is known about the underlying pathogenesis of these two chronic inflammatory diseases of the lung. The review highlights why they are different conditions requiring different approaches to treatment and provides a backdrop for the subsequent chapters in this volume discussing recent advances in the pharmacology and treatment of asthma and COPD.
Collapse
Affiliation(s)
- Clive Page
- Sackler Institute of Pulmonary Pharmacology, King's College London, 150 Stamford Street, London, SE1 9NH, UK.
| | - Blaze O'Shaughnessy
- Sackler Institute of Pulmonary Pharmacology, King's College London, 150 Stamford Street, London, SE1 9NH, UK
| | - Peter Barnes
- Department of Thoracic Medicine, National Heart and Lung Institute, Imperial College London, Dovehouse Street, London, SW3 6LY, UK
| |
Collapse
|
43
|
Cellular and molecular mechanisms of asthma and COPD. Clin Sci (Lond) 2017; 131:1541-1558. [PMID: 28659395 DOI: 10.1042/cs20160487] [Citation(s) in RCA: 303] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 12/19/2022]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) both cause airway obstruction and are associated with chronic inflammation of the airways. However, the nature and sites of the inflammation differ between these diseases, resulting in different pathology, clinical manifestations and response to therapy. In this review, the inflammatory and cellular mechanisms of asthma and COPD are compared and the differences in inflammatory cells and profile of inflammatory mediators are highlighted. These differences account for the differences in clinical manifestations of asthma and COPD and their response to therapy. Although asthma and COPD are usually distinct, there are some patients who show an overlap of features, which may be explained by the coincidence of two common diseases or distinct phenotypes of each disease. It is important to better understand the underlying cellular and molecular mechanisms of asthma and COPD in order to develop new treatments in areas of unmet need, such as severe asthma, curative therapy for asthma and effective anti-inflammatory treatments for COPD.
Collapse
|
44
|
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
|
45
|
Sriram KB, Cox AJ, Clancy RL, Slack MPE, Cripps AW. Nontypeable Haemophilus influenzae and chronic obstructive pulmonary disease: a review for clinicians. Crit Rev Microbiol 2017; 44:125-142. [PMID: 28539074 DOI: 10.1080/1040841x.2017.1329274] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a leading cause of morbidity and mortality worldwide. In the lower airways of COPD patients, bacterial infection is a common phenomenon and Haemophilus influenzae is the most commonly identified bacteria. Haemophilus influenzae is divided into typeable and nontypeable (NTHi) strains based on the presence or absence of a polysaccharide capsule. While NTHi is a common commensal in the human nasopharynx, it is associated with considerable inflammation when it is present in the lower airways of COPD patients, resulting in morbidity due to worsening symptoms and increased frequency of COPD exacerbations. Treatment of lower airway NTHi infection with antibiotics, though successful in the short term, does not offer long-term protection against reinfection, nor does it change the course of the disease. Hence, there has been much interest in the development of an effective NTHi vaccine. This review will summarize the current literature concerning the role of NTHi infections in COPD patients and the consequences of using prophylactic antibiotics in patients with COPD. There is particular focus on the rationale, findings of clinical studies and possible future directions of NTHi vaccines in patients with COPD.
Collapse
Affiliation(s)
- Krishna Bajee Sriram
- a Department of Respiratory Medicine , Gold Coast University Hospital, Gold Coast Health , Southport , Australia.,b Griffith University School of Medicine , Southport , Australia
| | - Amanda J Cox
- c Menzies Health Institute , Griffith University School of Medical Science , Gold Coast , Australia
| | - Robert L Clancy
- d Faculty of Health and Medicine , University of Newcastle , Callaghan , Australia
| | - Mary P E Slack
- b Griffith University School of Medicine , Southport , Australia
| | - Allan W Cripps
- b Griffith University School of Medicine , Southport , Australia
| |
Collapse
|
46
|
Kolsum U, Donaldson GC, Singh R, Barker BL, Gupta V, George L, Webb AJ, Thurston S, Brookes AJ, McHugh TD, Wedzicha JA, Brightling CE, Singh D. Blood and sputum eosinophils in COPD; relationship with bacterial load. Respir Res 2017; 18:88. [PMID: 28482840 PMCID: PMC5422866 DOI: 10.1186/s12931-017-0570-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/01/2017] [Indexed: 12/20/2022] Open
Abstract
Background Sputum and blood eosinophil counts predict corticosteroid effects in COPD patients. Bacterial infection causes increased airway neutrophilic inflammation. The relationship of eosinophil counts with airway bacterial load in COPD patients is uncertain. We tested the hypothesis that bacterial load and eosinophil counts are inversely related. Methods COPD patients were seen at stable state and exacerbation onset. Sputum was processed for quantitative polymerase chain reaction detection of the potentially pathogenic microorganisms (PPM) H. influenzae, M. catarrhalis and S. pneumoniae. PPM positive was defined as total load ≥1 × 104copies/ml. Sputum and whole blood were analysed for differential cell counts. Results At baseline, bacterial counts were not related to blood eosinophils, but sputum eosinophil % was significantly lower in patients with PPM positive compared to PPM negative samples (medians: 0.5% vs. 1.25% respectively, p = 0.01). Patients with PPM positive samples during an exacerbation had significantly lower blood eosinophil counts at exacerbation compared to baseline (medians: 0.17 × 109/L vs. 0.23 × 109/L respectively, p = 0.008), while no blood eosinophil change was observed with PPM negative samples. Conclusions These findings indicate an inverse relationship between bacterial infection and eosinophil counts. Bacterial infection may influence corticosteroid responsiveness by altering the profile of neutrophilic and eosinophilic inflammation. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0570-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Umme Kolsum
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and University Hospital of South Manchester NHS Foundation Trust, Manchester, M23 9QZ, UK. .,The Medicines Evaluation Unit, Manchester, UK.
| | - Gavin C Donaldson
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Richa Singh
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Bethan L Barker
- Department of Infection, Immunity, and Inflammation, Institute for Lung Health, NIHR Respiratory Biomedical Research Unit, University of Leicester, Leicester, UK
| | - Vandana Gupta
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and University Hospital of South Manchester NHS Foundation Trust, Manchester, M23 9QZ, UK
| | - Leena George
- Department of Infection, Immunity, and Inflammation, Institute for Lung Health, NIHR Respiratory Biomedical Research Unit, University of Leicester, Leicester, UK
| | - Adam J Webb
- Department of Genetics, University of Leicester, Leicester, UK
| | - Sarah Thurston
- Centre for Clinical Microbiology, University College London, London, UK
| | | | - Timothy D McHugh
- Centre for Clinical Microbiology, University College London, London, UK
| | - Jadwiga A Wedzicha
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Christopher E Brightling
- Department of Infection, Immunity, and Inflammation, Institute for Lung Health, NIHR Respiratory Biomedical Research Unit, University of Leicester, Leicester, UK
| | - Dave Singh
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and University Hospital of South Manchester NHS Foundation Trust, Manchester, M23 9QZ, UK.,The Medicines Evaluation Unit, Manchester, UK
| |
Collapse
|
47
|
Perez AC, Murphy TF. Potential impact of a Moraxella catarrhalis vaccine in COPD. Vaccine 2017; 37:5551-5558. [PMID: 28185742 DOI: 10.1016/j.vaccine.2016.12.066] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 12/28/2016] [Indexed: 12/27/2022]
Abstract
Moraxella catarrhalis is the second most common cause of exacerbations in adults with COPD, resulting in enormous morbidity and mortality in this clinical setting. Vaccine development for M. catarrhalis has lagged behind the other two important causes of exacerbations in COPD, nontypeable Haemophilus influenzae and Streptococcus pneumoniae. While no licensed vaccine is currently available for M. catarrhalis, several promising candidate vaccine antigens have been identified and characterized and are close to entering clinical trials. Key steps that are required to advance vaccines for M. catarrhalis along the translational pipeline include standardization of assay systems to assess candidate antigens, identification of a reliable correlate of protection and expansion of partnerships between industry, academia and government to overcome regulatory hurdles. A vaccine to prevent M. catarrhalis infections in COPD would have a major impact in reducing morbidity, mortality and healthcare costs in COPD.
Collapse
Affiliation(s)
- Antonia C Perez
- Clinical and Translational Research Center, University at Buffalo, The State University of New York, 875 Ellicott Street, Buffalo, NY 14203, USA; Division of Infectious Diseases, Department of Medicine, University at Buffalo, The State University of New York, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - Timothy F Murphy
- Clinical and Translational Research Center, University at Buffalo, The State University of New York, 875 Ellicott Street, Buffalo, NY 14203, USA; Division of Infectious Diseases, Department of Medicine, University at Buffalo, The State University of New York, 875 Ellicott Street, Buffalo, NY 14203, USA; Department of Microbiology, University at Buffalo, The State University of New York, 875 Ellicott Street, Buffalo, NY 14203, USA.
| |
Collapse
|
48
|
Staples KJ, Taylor S, Thomas S, Leung S, Cox K, Pascal TG, Ostridge K, Welch L, Tuck AC, Clarke SC, Gorringe A, Wilkinson TMA. Relationships between Mucosal Antibodies, Non-Typeable Haemophilus influenzae (NTHi) Infection and Airway Inflammation in COPD. PLoS One 2016; 11:e0167250. [PMID: 27898728 PMCID: PMC5127575 DOI: 10.1371/journal.pone.0167250] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/10/2016] [Indexed: 12/31/2022] Open
Abstract
Non-typeable Haemophilus influenzae (NTHi) is a key pathogen in COPD, being associated with airway inflammation and risk of exacerbation. Why some patients are susceptible to colonisation is not understood. We hypothesised that this susceptibility may be due to a deficiency in mucosal humoral immunity. The aim of our study (NCT01701869) was to quantify the amount and specificity of antibodies against NTHi in the lungs and the associated risk of infection and inflammation in health and COPD. Phlebotomy, sputum induction and bronchoscopy were performed on 24 mild-to-moderate COPD patients and 8 age and smoking-matched controls. BAL (Bronchoalveolar lavage) total IgG1, IgG2, IgG3, IgM and IgA concentrations were significantly increased in COPD patients compared to controls. NTHi was detected in the lungs of 7 of the COPD patients (NTHi+ve-29%) and these patients had a higher median number of previous exacerbations than NTHi-ve patients as well as evidence of increased systemic inflammation. When comparing NTHi+ve versus NTHi-ve patients we observed a decrease in the amount of both total IgG1 (p = 0.0068) and NTHi-specific IgG1 (p = 0.0433) in the BAL of NTHi+ve patients, but no differences in total IgA or IgM. We observed no evidence of decreased IgG1 in the serum of NTHi+ve patients, suggesting this phenomenon is restricted to the airway. Furthermore, the NTHi+ve patients had significantly greater levels of IL-1β (p = 0.0003), in BAL than NTHi-ve COPD patients.This study indicates that the presence of NTHi is associated with reduced levels and function of IgG1 in the airway of NTHi-colonised COPD patients. This decrease in total and NTHI-specific IgG1 was associated with greater systemic and airway inflammation and a history of more frequent exacerbations and may explain the susceptibility of some COPD patients to the impacts of NTHi.
Collapse
Affiliation(s)
- Karl J. Staples
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Tremona Road, Southampton, United Kingdom
- Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Tremona Road, Southampton, United Kingdom
- * E-mail:
| | - Stephen Taylor
- Public Health England, Porton Down, Salisbury, United Kingdom
| | - Steve Thomas
- Public Health England, Porton Down, Salisbury, United Kingdom
| | - Stephanie Leung
- Public Health England, Porton Down, Salisbury, United Kingdom
| | - Karen Cox
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Tremona Road, Southampton, United Kingdom
| | | | - Kristoffer Ostridge
- Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Tremona Road, Southampton, United Kingdom
| | - Lindsay Welch
- Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Tremona Road, Southampton, United Kingdom
| | - Andrew C. Tuck
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Tremona Road, Southampton, United Kingdom
| | - Stuart C. Clarke
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Tremona Road, Southampton, United Kingdom
- Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Tremona Road, Southampton, United Kingdom
- Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Tremona Road, Southampton, United Kingdom
| | - Andrew Gorringe
- Public Health England, Porton Down, Salisbury, United Kingdom
| | - Tom M. A. Wilkinson
- Clinical & Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Tremona Road, Southampton, United Kingdom
- Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Tremona Road, Southampton, United Kingdom
- Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Tremona Road, Southampton, United Kingdom
| |
Collapse
|
49
|
Clancy RL, Dunkley ML, Sockler J, McDonald CF. Multi-site placebo-controlled randomised clinical trial to assess protection following oral immunisation with inactivated non-typeableHaemophilus influenzaein chronic obstructive pulmonary disease. Intern Med J 2016; 46:684-93. [DOI: 10.1111/imj.13072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 01/25/2023]
Affiliation(s)
- R. L. Clancy
- Faculty of Health and Medicine; University of Newcastle; Newcastle New South Wales Australia
| | - M. L. Dunkley
- Faculty of Health and Medicine; University of Newcastle; Newcastle New South Wales Australia
- Hunter Medical Research Institute; Newcastle New South Wales Australia
| | - J. Sockler
- Datapharm Australia Pty Ltd; Sydney New South Wales Australia
| | - C. F. McDonald
- Department of Respiratory and Sleep Medicine; Austin Hospital; Melbourne Victoria Australia
- Department of Medicine; The University of Melbourne; Melbourne Victoria Australia
| |
Collapse
|
50
|
Barnes PJ. Inflammatory mechanisms in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol 2016; 138:16-27. [PMID: 27373322 DOI: 10.1016/j.jaci.2016.05.011] [Citation(s) in RCA: 923] [Impact Index Per Article: 102.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 12/15/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is associated with chronic inflammation affecting predominantly the lung parenchyma and peripheral airways that results in largely irreversible and progressive airflow limitation. This inflammation is characterized by increased numbers of alveolar macrophages, neutrophils, T lymphocytes (predominantly TC1, TH1, and TH17 cells), and innate lymphoid cells recruited from the circulation. These cells and structural cells, including epithelial and endothelial cells and fibroblasts, secrete a variety of proinflammatory mediators, including cytokines, chemokines, growth factors, and lipid mediators. Although most patients with COPD have a predominantly neutrophilic inflammation, some have an increase in eosinophil counts, which might be orchestrated by TH2 cells and type 2 innate lymphoid cells though release of IL-33 from epithelial cells. These patients might be more responsive to corticosteroids and bronchodilators. Oxidative stress plays a key role in driving COPD-related inflammation, even in ex-smokers, and might result in activation of the proinflammatory transcription factor nuclear factor κB (NF-κB), impaired antiprotease defenses, DNA damage, cellular senescence, autoantibody generation, and corticosteroid resistance though inactivation of histone deacetylase 2. Systemic inflammation is also found in patients with COPD and can worsen comorbidities, such as cardiovascular diseases, diabetes, and osteoporosis. Accelerated aging in the lungs of patients with COPD can also generate inflammatory protein release from senescent cells in the lung. In the future, it will be important to recognize phenotypes of patients with optimal responses to more specific therapies, and development of biomarkers that identify the therapeutic phenotypes will be important.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart and Lung Institute, Imperial College, London, United Kingdom.
| |
Collapse
|