1
|
Shuai T, Liu J, Dong M, Wu P, Zhang L, Feng Z, Li W, Liu J. The safety and efficacy of non-typeable Haemophilus influenzae and Moraxella catarrhalis vaccine in chronic obstructive pulmonary disease: a systematic review and meta-analysis of randomized controlled trials. Front Med (Lausanne) 2025; 12:1572726. [PMID: 40255595 PMCID: PMC12006075 DOI: 10.3389/fmed.2025.1572726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/20/2025] [Indexed: 04/22/2025] Open
Abstract
Background Non-typeable Haemophilus influenzae (NTHi) and Moraxella catarrhalis (Mcat) are major pathogens implicated in bacterial exacerbations of chronic obstructive pulmonary disease (COPD). Their involvement contributes to antibiotic resistance and poses significant immune challenges, underscoring the need for targeted vaccine strategies. This systematic review and meta-analysis assessed the safety and efficacy of NTHi-Mcat/NTHi vaccines in COPD patients. Research design and methods Randomized controlled trials (RCTs) assessing the safety and efficacy of NTHi-Mcat/NTHi vaccines for COPD were systematically searched across four databases (PubMed, CENTRAL, Embase, and Medline) from inception to October 2024. Meta-analyses were conducted using random-effects or fixed-effects models, with subgroup analyses to investigate possible sources of heterogeneity. Results This analysis included eight RCTs involving 1,574 participants, primarily conducted in Europe (n = 3) and Australia (n = 2), with interventions administered orally or intramuscularly at varying frequencies (twice or three times). The Meta-analyses revealed that the NTHi-Mcat/NTHi vaccine did not affect the incidence of acute exacerbations of COPD (relative risk (RR): 1.02, 95% confidence interval (CI): 0.76 to 1.36), all-cause mortality (RR: 0.91, 95% CI: 0.38 to 2.21), and hospitalization rate (RR: 0.50, 95% CI: 0.09 to 2.77). Regarding safety, the NTHi-Mcat/NTHi vaccine did not significantly increase the risk of serious adverse events (RR: 1.00, 95% CI: 0.84 to 1.19) or grade 3 serious events (RR: 1.20, 95% CI: 0.93 to 1.53). However, it was associated with a higher risk of local and systemic reactions, including pain (RR: 5.33, 95% CI: 1.98 to 14.33), swelling (RR: 12.15, 95% CI: 4.67 to 31.67), redness (first dose: RR: 12.74, 95% CI: 3.48 to 46.59; second dose: RR: 11.55, 95% CI: 3.90 to 34.22), headaches (RR: 1.20, 95% CI: 1.00 to 1.43), erythema (RR: 15.38, 95% CI: 5.64 to 41.92), and fever (after the second dose: RR: 2.33, 95% CI: 1.24 to 4.38). Conclusion Although the NTHi-Mcat/NTHi vaccines were well-tolerated in COPD patients, they did not significantly reduce the risk of exacerbations or mortality. These findings suggest that further research is needed to validate these results and identify potential subgroups that may derive clinical benefit. Systematic review registration The study was registered in PROSPERO (ID: CRD42023381488).
Collapse
Affiliation(s)
- Tiankui Shuai
- Department of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Jing Liu
- Outpatient Department, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shanxi, China
| | - Meijun Dong
- Department of Internal Medicine, Wenxian First People’s Hospital, Longnan, Lanzhou, Gansu, China
| | - Peng Wu
- KeyMed Biosciences Inc., Chengdu, Sichuan, China
| | - Lu Zhang
- Department of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zhouzhou Feng
- Department of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Wenqiang Li
- Department of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Jian Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Gansu Provincial Central Hospital, Gansu Provincial Maternity and Child-Care Hospital, Lanzhou, Gansu, China
| |
Collapse
|
2
|
Azoicai A, Lupu A, Alexoae MM, Starcea IM, Mocanu A, Lupu VV, Mitrofan EC, Nedelcu AH, Tepordei RT, Munteanu D, Mitrofan C, Salaru DL, Ioniuc I. Lung microbiome: new insights into bronchiectasis' outcome. Front Cell Infect Microbiol 2024; 14:1405399. [PMID: 38895737 PMCID: PMC11183332 DOI: 10.3389/fcimb.2024.1405399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
The present treatments for bronchiectasis, which is defined by pathological dilatation of the airways, are confined to symptom relief and minimizing exacerbations. The condition is becoming more common worldwide. Since the disease's pathophysiology is not entirely well understood, developing novel treatments is critically important. The interplay of chronic infection, inflammation, and compromised mucociliary clearance, which results in structural alterations and the emergence of new infection, is most likely responsible for the progression of bronchiectasis. Other than treating bronchiectasis caused by cystic fibrosis, there are no approved treatments. Understanding the involvement of the microbiome in this disease is crucial, the microbiome is defined as the collective genetic material of all bacteria in an environment. In clinical practice, bacteria in the lungs have been studied using cultures; however, in recent years, researchers use next-generation sequencing methods, such as 16S rRNA sequencing. Although the microbiome in bronchiectasis has not been entirely investigated, what is known about it suggests that Haemophilus, Pseudomonas and Streptococcus dominate the lung bacterial ecosystems, they present significant intraindividual stability and interindividual heterogeneity. Pseudomonas and Haemophilus-dominated microbiomes have been linked to more severe diseases and frequent exacerbations, however additional research is required to fully comprehend the role of microbiome in the evolution of bronchiectasis. This review discusses recent findings on the lung microbiota and its association with bronchiectasis.
Collapse
Affiliation(s)
- Alice Azoicai
- Mother and Child Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Ancuta Lupu
- Mother and Child Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Monica Mihaela Alexoae
- Mother and Child Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Iuliana Magdalena Starcea
- Mother and Child Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Adriana Mocanu
- Mother and Child Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Vasile Valeriu Lupu
- Mother and Child Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | | | - Alin Horatiu Nedelcu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Razvan Tudor Tepordei
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Dragos Munteanu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Costica Mitrofan
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Delia Lidia Salaru
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Ileana Ioniuc
- Mother and Child Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| |
Collapse
|
3
|
Pang X, Liu X. Immune Dysregulation in Chronic Obstructive Pulmonary Disease. Immunol Invest 2024; 53:652-694. [PMID: 38573590 DOI: 10.1080/08820139.2024.2334296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease whose incidence increase with age and is characterised by chronic inflammation and significant immune dysregulation. Inhalation of toxic substances cause oxidative stress in the lung tissue as well as airway inflammation, under the recruitment of chemokines, immune cells gathered and are activated to play a defensive role. However, persistent inflammation damages the immune system and leads to immune dysregulation, which is mainly manifested in the reduction of the body's immune response to antigens, and immune cells function are impaired, further destroy the respiratory defensive system, leading to recurrent lower respiratory infections and progressive exacerbation of the disease, thus immune dysregulation play an important role in the pathogenesis of COPD. This review summarizes the changes of innate and adaptive immune-related cells during the pathogenesis of COPD, aiming to control COPD airway inflammation and improve lung tissue remodelling by regulating immune dysregulation, for further reducing the risk of COPD progression and opening new avenues of therapeutic intervention in COPD.
Collapse
Affiliation(s)
- Xichen Pang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoju Liu
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
4
|
Feldman C, Theron AJ, Cholo MC, Anderson R. Cigarette Smoking as a Risk Factor for Tuberculosis in Adults: Epidemiology and Aspects of Disease Pathogenesis. Pathogens 2024; 13:151. [PMID: 38392889 PMCID: PMC10892798 DOI: 10.3390/pathogens13020151] [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: 11/22/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
It has been noted by the World Health Organisation that cases of tuberculosis in 2022 globally numbered 10.6 million, resulting in 1.3 million deaths, such that TB is one of the infectious diseases causing the greatest morbidity and mortality worldwide. Since as early as 1918, there has been an ongoing debate as to the relationship between cigarette smoking and TB. However, numerous epidemiological studies, as well as meta-analyses, have indicated that both active and passive smoking are independent risk factors for TB infection, development of reactivation TB, progression of primary TB, increased severity of cavitary disease, and death from TB, among several other considerations. With this considerable body of evidence confirming the association between smoking and TB, it is not surprising that TB control programmes represent a key potential preventative intervention. In addition to coverage of the epidemiology of TB and its compelling causative link with smoking, the current review is also focused on evidence derived from clinical- and laboratory-based studies of disease pathogenesis, most prominently the protective anti-mycobacterial mechanisms of the alveolar macrophage, the primary intracellular refuge of M. tuberculosis. This section of the review is followed by an overview of the major strategies utilised by the pathogen to subvert these antimicrobial mechanisms in the airway, which are intensified by the suppressive effects of smoke inhalation on alveolar macrophage function. Finally, consideration is given to a somewhat under-explored, pro-infective activity of cigarette smoking, namely augmentation of antibiotic resistance due to direct effects of smoke per se on the pathogen. These include biofilm formation, induction of cellular efflux pumps, which eliminate both smoke-derived toxicants and antibiotics, as well as gene modifications that underpin antibiotic resistance.
Collapse
Affiliation(s)
- Charles Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, York Road, Parktown, Johannesburg 2193, South Africa;
| | - Annette J. Theron
- Department of Immunology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Bophelo Road, Prinshof, Pretoria 0083, South Africa; (A.J.T.); (M.C.C.)
| | - Moloko C. Cholo
- Department of Immunology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Bophelo Road, Prinshof, Pretoria 0083, South Africa; (A.J.T.); (M.C.C.)
| | - Ronald Anderson
- Department of Immunology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Bophelo Road, Prinshof, Pretoria 0083, South Africa; (A.J.T.); (M.C.C.)
| |
Collapse
|
5
|
Souza VGP, Forder A, Pewarchuk ME, Telkar N, de Araujo RP, Stewart GL, Vieira J, Reis PP, Lam WL. The Complex Role of the Microbiome in Non-Small Cell Lung Cancer Development and Progression. Cells 2023; 12:2801. [PMID: 38132121 PMCID: PMC10741843 DOI: 10.3390/cells12242801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
In recent years, there has been a growing interest in the relationship between microorganisms in the surrounding environment and cancer cells. While the tumor microenvironment predominantly comprises cancer cells, stromal cells, and immune cells, emerging research highlights the significant contributions of microbial cells to tumor development and progression. Although the impact of the gut microbiome on treatment response in lung cancer is well established, recent investigations indicate complex roles of lung microbiota in lung cancer. This article focuses on recent findings on the human lung microbiome and its impacts in cancer development and progression. We delve into the characteristics of the lung microbiome and its influence on lung cancer development. Additionally, we explore the characteristics of the intratumoral microbiome, the metabolic interactions between lung tumor cells, and how microorganism-produced metabolites can contribute to cancer progression. Furthermore, we provide a comprehensive review of the current literature on the lung microbiome and its implications for the metastatic potential of tumor cells. Additionally, this review discusses the potential for therapeutic modulation of the microbiome to establish lung cancer prevention strategies and optimize lung cancer treatment.
Collapse
Affiliation(s)
- Vanessa G. P. Souza
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- Molecular Oncology Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil (P.P.R.)
| | - Aisling Forder
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | | | - Nikita Telkar
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Rachel Paes de Araujo
- Molecular Oncology Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil (P.P.R.)
| | - Greg L. Stewart
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Juliana Vieira
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Patricia P. Reis
- Molecular Oncology Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil (P.P.R.)
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Wan L. Lam
- British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| |
Collapse
|
6
|
Bedford R, Smith G, Rothwell E, Martin S, Medhane R, Casentieri D, Daunt A, Freiberg G, Hollings M. A multi-organ, lung-derived inflammatory response following in vitro airway exposure to cigarette smoke and next-generation nicotine delivery products. Toxicol Lett 2023; 387:35-49. [PMID: 37774809 DOI: 10.1016/j.toxlet.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Despite increasing use of in vitro models that closely resemble in vivo human biology, their application in understanding downstream effects of airway toxicity, such as inflammation, are at an early stage. In this study, we used various assays to examine the inflammatory response induced in MucilAir™ tissues and A549 cells exposed to three products known to induce toxicity. Reduced barrier integrity was observed in tissues following exposure to each product, with reduced viability and increased cytotoxicity also shown. Similar changes in viability were also observed in A549 cells. Furthermore, whole cigarette smoke (CS) induced downstream phenotypic THP-1 changes and endothelial cell adhesion, an early marker of atherosclerosis. In contrast, exposure to next-generation delivery product (NGP) aerosol did not induce this response. Cytokine, histological and RNA analysis highlighted increased biomarkers linked to inflammatory pathways and immune cell differentiation following exposure to whole cigarette smoke, including GM-CSF, IL-1β, cleaved caspase-3 and cytochrome P450 enzymes. As a result of similar observations in human airway inflammation, we propose that our exposure platform could act as a representative model for studying such events in vitro. Furthermore, this model could be used to test the inflammatory or anti-inflammatory impact posed by inhaled compounds delivered to the lung.
Collapse
Affiliation(s)
- R Bedford
- Labcorp Early Development Laboratories Limited, Harrogate, UK.
| | - G Smith
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - E Rothwell
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - S Martin
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - R Medhane
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - D Casentieri
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - A Daunt
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - G Freiberg
- Labcorp Early Development Laboratories Limited, Eye, UK
| | - M Hollings
- Labcorp Early Development Laboratories Limited, Harrogate, UK.
| |
Collapse
|
7
|
Kotlyarov S. The Role of Smoking in the Mechanisms of Development of Chronic Obstructive Pulmonary Disease and Atherosclerosis. Int J Mol Sci 2023; 24:8725. [PMID: 37240069 PMCID: PMC10217854 DOI: 10.3390/ijms24108725] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Tobacco smoking is a major cause of chronic obstructive pulmonary disease (COPD) and atherosclerotic cardiovascular disease (ASCVD). These diseases share common pathogenesis and significantly influence each other's clinical presentation and prognosis. There is increasing evidence that the mechanisms underlying the comorbidity of COPD and ASCVD are complex and multifactorial. Smoking-induced systemic inflammation, impaired endothelial function and oxidative stress may contribute to the development and progression of both diseases. The components present in tobacco smoke can have adverse effects on various cellular functions, including macrophages and endothelial cells. Smoking may also affect the innate immune system, impair apoptosis, and promote oxidative stress in the respiratory and vascular systems. The purpose of this review is to discuss the importance of smoking in the mechanisms underlying the comorbid course of COPD and ASCVD.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
8
|
Fleischmann M, Jarnicki AG, Brown AS, Yang C, Anderson GP, Garbi N, Hartland EL, van Driel IR, Ng GZ. Cigarette smoke depletes alveolar macrophages and delays clearance of Legionella pneumophila. Am J Physiol Lung Cell Mol Physiol 2023; 324:L373-L384. [PMID: 36719079 PMCID: PMC10026984 DOI: 10.1152/ajplung.00268.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/23/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
Legionella pneumophila is the main etiological agent of Legionnaires' disease, a severe bacterial pneumonia. L. pneumophila is initially engulfed by alveolar macrophages (AMs) and subvert normal cellular functions to establish a replicative vacuole. Cigarette smokers are particularly susceptible to developing Legionnaires' disease and other pulmonary infections; however, little is known about the cellular mechanisms underlying this susceptibility. To investigate this, we used a mouse model of acute cigarette smoke exposure to examine the immune response to cigarette smoke and subsequent L. pneumophila infection. Contrary to previous reports, we show that cigarette smoke exposure alone causes a significant depletion of AMs using enzymatic digestion to extract cells, or via imaging intact lung lobes by light-sheet microscopy. Furthermore, treatment of mice deficient in specific types of cell death with smoke suggests that NLRP3-driven pyroptosis is a contributor to smoke-induced death of AMs. After infection, smoke-exposed mice displayed increased pulmonary L. pneumophila loads and developed more severe disease compared with air-exposed controls. We tested if depletion of AMs was related to this phenotype by directly depleting them with clodronate liposomes and found that this also resulted in increased L. pneumophila loads. In summary, our results showed that cigarette smoke depleted AMs from the lung and that this likely contributed to more severe Legionnaires' disease. Furthermore, the role of AMs in L. pneumophila infection is more nuanced than simply providing a replicative niche, and our studies suggest they play a major role in bacterial clearance.
Collapse
Affiliation(s)
- Markus Fleischmann
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
- Institute for Experimental Immunology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Andrew G Jarnicki
- Lung Health Research Centre, Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia
| | - Andrew S Brown
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Chao Yang
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Gary P Anderson
- Lung Health Research Centre, Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria, Australia
| | - Natalio Garbi
- Institute for Experimental Immunology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Elizabeth L Hartland
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Ian R van Driel
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Garrett Z Ng
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
9
|
Sun S, Shen Y, Feng J. Association of toll-like receptors polymorphisms with COPD risk in Chinese population. Front Genet 2022; 13:955810. [PMID: 36386838 PMCID: PMC9643488 DOI: 10.3389/fgene.2022.955810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 10/14/2022] [Indexed: 09/19/2023] Open
Abstract
Background: Previous studies have reported that the Toll-like receptors (TLRs) are related with the progress of chronic obstructive pulmonary disease (COPD). We aimed to explore the association of TLRs single nucleotide polymorphisms (SNPs) and COPD risk. Methods: 170 COPD patients and 181 healthy controls were enrolled in this case-control study. MassARRAY platform was used for genotyping seven tagging SNPs (TLR2: rs3804100, rs4696480, rs3804099; TLR3: rs3775290, rs3775291, rs5743305; TLR9: rs352140) of TLRs. The correlations between the SNPs and COPD risk were determined using logistic regression. Results: We found that the rs3775291 of TLR3 significant decreased the risk of COPD (TT versus CC: non-adjusted OR = 0.329, 95% CI = 0.123-0.879, p = 0.027). In the genetic models analysis, the rs3775291 was associated with a decreased effect of COPD based on the recessive model (TT versus CC/CT: non-adjusted OR = 0.377, 95% CI = 0.144-0.988 p = 0.047). The rs4696480 of TLR2 gene was associated with a decreased risk of COPD after adjustment by age and gender (TA versus AA: adjusted OR = 0.606, 95% CI = 0.376-0.975, p = 0.039). Conclusion: Our study showed that genetic variants in TLRs were associated with risk of COPD. The rs3775291 and rs4696480 may act as a potential biomarker for predicting the risk of COPD in Chinese population.
Collapse
Affiliation(s)
- Shulei Sun
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuehao Shen
- Department of Intensive Care Unit, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Feng
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
10
|
Quan DH, Kwong AJ, Hansbro PM, Britton WJ. No smoke without fire: the impact of cigarette smoking on the immune control of tuberculosis. Eur Respir Rev 2022; 31:210252. [PMID: 35675921 PMCID: PMC9488690 DOI: 10.1183/16000617.0252-2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/20/2022] [Indexed: 12/12/2022] Open
Abstract
Cigarette smoke (CS) exposure is a key risk factor for both active and latent tuberculosis (TB). It is associated with delayed diagnosis, more severe disease progression, unfavourable treatment outcomes and relapse after treatment. Critically, CS exposure is common in heavily populated areas with a high burden of TB, such as China, India and the Russian Federation. It is therefore prudent to evaluate interventions for TB while taking into account the immunological impacts of CS exposure. This review is a mechanistic examination of how CS exposure impairs innate barrier defences, as well as alveolar macrophage, neutrophil, dendritic cell and T-cell functions, in the context of TB infection and disease.
Collapse
Affiliation(s)
- Diana H Quan
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Sydney, Australia
- D.H. Quan and W.J. Britton contributed equally to this article as lead authors and supervised the work
| | | | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Sydney, Australia
- Dept of Clinical Immunology, Royal Prince Alfred Hospital, Sydney, Australia
- D.H. Quan and W.J. Britton contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
11
|
Martín-Medina A, Cerón-Pisa N, Martinez-Font E, Shafiek H, Obrador-Hevia A, Sauleda J, Iglesias A. TLR/WNT: A Novel Relationship in Immunomodulation of Lung Cancer. Int J Mol Sci 2022; 23:6539. [PMID: 35742983 PMCID: PMC9224119 DOI: 10.3390/ijms23126539] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 02/07/2023] Open
Abstract
The most frequent cause of death by cancer worldwide is lung cancer, and the 5-year survival rate is still very poor for patients with advanced stage. Understanding the crosstalk between the signaling pathways that are involved in disease, especially in metastasis, is crucial to developing new targeted therapies. Toll-like receptors (TLRs) are master regulators of the immune responses, and their dysregulation in lung cancer is linked to immune escape and promotes tumor malignancy by facilitating angiogenesis and proliferation. On the other hand, over-activation of the WNT signaling pathway has been reported in lung cancer and is also associated with tumor metastasis via induction of Epithelial-to-mesenchymal-transition (EMT)-like processes. An interaction between both TLRs and the WNT pathway was discovered recently as it was found that the TLR pathway can be activated by WNT ligands in the tumor microenvironment; however, the implications of such interactions in the context of lung cancer have not been discussed yet. Here, we offer an overview of the interaction of TLR-WNT in the lung and its potential implications and role in the oncogenic process.
Collapse
Affiliation(s)
- Aina Martín-Medina
- Instituto de Investigación Sanitaria de les Illes Balears (IdISBa), 07120 Palma, Spain
| | - Noemi Cerón-Pisa
- Instituto de Investigación Sanitaria de les Illes Balears (IdISBa), 07120 Palma, Spain
| | - Esther Martinez-Font
- Instituto de Investigación Sanitaria de les Illes Balears (IdISBa), 07120 Palma, Spain
- Medical Oncology Department, Hospital Universitario Son Espases, 07120 Palma, Spain
| | - Hanaa Shafiek
- Chest Diseases Department, Faculty of Medicine, Alexandria University, Alexandria 21526, Egypt
| | - Antònia Obrador-Hevia
- Instituto de Investigación Sanitaria de les Illes Balears (IdISBa), 07120 Palma, Spain
- Molecular Diagnosis Unit, Hospital Universitario Son Espases, 07120 Palma, Spain
| | - Jaume Sauleda
- Instituto de Investigación Sanitaria de les Illes Balears (IdISBa), 07120 Palma, Spain
- Department of Respiratory Medicine, Hospital Universitario Son Espases, 07120 Palma, Spain
- Centro de Investigación Biomédica en Red in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| | - Amanda Iglesias
- Instituto de Investigación Sanitaria de les Illes Balears (IdISBa), 07120 Palma, Spain
- Centro de Investigación Biomédica en Red in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
12
|
Simpson S, Mclellan R, Wellmeyer E, Matalon F, George O. Drugs and Bugs: The Gut-Brain Axis and Substance Use Disorders. J Neuroimmune Pharmacol 2022; 17:33-61. [PMID: 34694571 PMCID: PMC9074906 DOI: 10.1007/s11481-021-10022-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/06/2021] [Indexed: 02/07/2023]
Abstract
Substance use disorders (SUDs) represent a significant public health crisis. Worldwide, 5.4% of the global disease burden is attributed to SUDs and alcohol use, and many more use psychoactive substances recreationally. Often associated with comorbidities, SUDs result in changes to both brain function and physiological responses. Mounting evidence calls for a precision approach for the treatment and diagnosis of SUDs, and the gut microbiome is emerging as a contributor to such disorders. Over the last few centuries, modern lifestyles, diets, and medical care have altered the health of the microbes that live in and on our bodies; as we develop, our diets and lifestyle dictate which microbes flourish and which microbes vanish. An increase in antibiotic treatments, with many antibiotic interventions occurring early in life during the microbiome's normal development, transforms developing microbial communities. Links have been made between the microbiome and SUDs, and the microbiome and conditions that are often comorbid with SUDs such as anxiety, depression, pain, and stress. A better understanding of the mechanisms influencing behavioral changes and drug use is critical in developing novel treatments for SUDSs. Targeting the microbiome as a therapeutic and diagnostic tool is a promising avenue of exploration. This review will provide an overview of the role of the gut-brain axis in a wide range of SUDs, discuss host and microbe pathways that mediate changes in the brain's response to drugs, and the microbes and related metabolites that impact behavior and health within the gut-brain axis.
Collapse
Affiliation(s)
- Sierra Simpson
- Department of Psychiatry, University of California San Diego, La Jolla, San Diego, CA, 92093, US.
| | - Rio Mclellan
- Department of Psychiatry, University of California San Diego, La Jolla, San Diego, CA, 92093, US
| | - Emma Wellmeyer
- Department of Psychiatry, University of California San Diego, La Jolla, San Diego, CA, 92093, US
| | - Frederic Matalon
- Department of Psychiatry, University of California San Diego, La Jolla, San Diego, CA, 92093, US
| | - Olivier George
- Department of Psychiatry, University of California San Diego, La Jolla, San Diego, CA, 92093, US
| |
Collapse
|
13
|
Galgani I, Annaratone M, Casula D, Di Maro G, Janssens M, Tasciotti A, Schwarz T, Ferguson M, Arora AK. Safety and immunogenicity of three doses of non-typeable Haemophilus influenzae-Moraxella catarrhalis (NTHi-Mcat) vaccine when administered according to two different schedules: a phase 2, randomised, observer-blind study. Respir Res 2022; 23:114. [PMID: 35509077 PMCID: PMC9069748 DOI: 10.1186/s12931-022-02019-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 04/10/2022] [Indexed: 11/12/2022] Open
Abstract
Background Non-typeable Haemophilus influenzae (NTHi) and Moraxella catarrhalis (Mcat) infections are frequently associated with exacerbations of chronic obstructive pulmonary disease (COPD). Results were reported with a two-dose (0–2 months) schedule of an investigational AS01E-adjuvanted NTHi-Mcat vaccine containing three surface proteins from NTHi and one from Mcat. We evaluated the safety and immunogenicity of three NTHi-Mcat vaccine doses administered in two different schedules to adults with a smoking history (≥ 10 pack-years), immunologically representing the COPD population. Methods In this 18-month, randomised (1:1), observer-blind study with 6-month open follow-up, 200 healthy adults aged 40–80 years received NTHi-Mcat vaccine at 0–2–6 months and placebo at 12 months (0–2–6 group), or vaccine at 0–2–12 months and placebo at 6 months (0–2–12 group). Solicited and unsolicited adverse events (AEs) were recorded for 7 and 30 days, respectively, post-vaccination, and potential immune-mediated diseases (pIMDs) and serious AEs (SAEs) throughout the study. Immune responses were assessed. Results No safety concerns were identified with the third vaccine dose or overall. Most solicited AEs were mild/moderate. Unsolicited AEs were reported in 16%, 16.1% and 14.4% of participants in the 0–2–6 group post-dose 1, 2 and 3, respectively, and 20%, 20.4% and 9.7%, respectively, in the 0–2–12 group. In 24 months, SAEs were reported in 12 participants in the 0–2–6 group and 9 in the 0–2–12 group (18 events in each group). There were three deaths (unknown cause, 0–2–6 group; myocardial infarction, lung cancer in 0–2–12 group). pIMDs were reported in three participants in the 0–2–6 group (non-serious inflammatory bowel disease, gout, psoriasis) and three in the 0–2–12 group (serious ulcerative colitis, two with non-serious gout). The SAEs, deaths and pIMDs were considered not causally related to vaccination. Antigen-specific antibody concentrations were higher at 12 months post-dose 1 with the 0–2–6 schedule than with the 0–2–12 schedule and at 12 months post-dose 3 were similar between schedules, remaining higher than baseline. Conclusions No safety concerns were identified when the investigational NTHi-Mcat vaccine was administered via a 0–2–6 months or 0–2–12 months schedule to older adults with a smoking history. Persistent immune responses were observed after the third vaccine dose. Trial registrationhttps://clinicaltrials.gov/; NCT03443427, registered February 23, 2018. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02019-4.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tino Schwarz
- Institute of Laboratory Medicine and Vaccination Centre, Klinikum Würzburg Mitte, Campus Juliusspital, Würzburg, Germany
| | | | | |
Collapse
|
14
|
Andreas S, Testa M, Boyer L, Brusselle G, Janssens W, Kerwin E, Papi A, Pek B, Puente-Maestu L, Saralaya D, Watz H, Wilkinson TMA, Casula D, Di Maro G, Lattanzi M, Moraschini L, Schoonbroodt S, Tasciotti A, Arora AK, Maltais F, Brusselle G, Corhay JL, Janssens E, Janssens W, Leys M, Ferguson M, Fitzgerald M, Maltais F, Mayers I, McNeil S, Pek B, Bourdin A, Boyer L, Couturaud F, Dussart L, Andreas S, Illies G, Eich A, Ludwig-Sengpiel A, Watz H, Blasi F, Centanni S, Papi A, Pomari C, Echave-Sustaeta JM, Llorca Martínez E, Narejos Pérez S, Pascual-Guardia S, Pérez Vera M, Puente-Maestu L, Terns Riera M, Anderson W, Choudhury G, De-Soyza A, Saralaya D, Wilkinson TMA, Boscia III J, Chinsky K, Dunn L, Erb D, Fogarty C, Downey HJ, Kerwin E, Kunz C, Poling T, Sellman R, Sigal B, Southard J, Spangenthal S, Tannous Z, Testa M, Casula D, Di Maro G, Lattanzi M, Moraschini L, Schoonbroodt S, Tasciotti A, Arora AK. Non-typeable Haemophilus influenzae–Moraxella catarrhalis vaccine for the prevention of exacerbations in chronic obstructive pulmonary disease: a multicentre, randomised, placebo-controlled, observer-blinded, proof-of-concept, phase 2b trial. THE LANCET RESPIRATORY MEDICINE 2022; 10:435-446. [DOI: 10.1016/s2213-2600(21)00502-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/25/2021] [Accepted: 11/02/2021] [Indexed: 12/14/2022]
|
15
|
Skronska-Wasek W, Durlanik S, Le HQ, Schroeder V, Kitt K, Garnett JP, Pflanz S. The antimicrobial peptide S100A8/A9 produced by airway epithelium functions as a potent and direct regulator of macrophage phenotype and function. Eur Respir J 2022; 59:13993003.02732-2020. [PMID: 34561292 PMCID: PMC8989056 DOI: 10.1183/13993003.02732-2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 08/10/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Elevated counts of alveolar macrophages and attenuated phagocytic capacity are associated with chronic obstructive pulmonary disease (COPD). Factors governing macrophage phagocytosis are poorly understood. In this study we aimed to compare the influence of airway epithelial cell secretions from individuals with COPD and without COPD (non-COPD) on macrophage phagocytic activity, and the role of antimicrobial peptides (AMPs). METHODS Supernatants from non-COPD and COPD small airway epithelial cell (SAEC) cultures exposed to non-typeable Haemophilus influenzae (NTHi) were applied to human monocyte-derived macrophages (MDMs) to assess their influence on phagocytosis. SAECs were analysed for changes in AMP expression by quantitative reverse transcription PCR, and the influence of select AMPs on macrophage phenotype and function was assessed by flow cytometry and metabolic activity assay. RESULTS Secretions from the apical and basolateral surface of NTHi-exposed SAECs from non-COPD donors elicited superior phagocytic capacity in MDMs. Moreover, NTHi exposure led to a rapid increase in the expression of a range of AMPs by non-COPD SAECs, but this response was delayed in COPD SAECs. We demonstrate that treatment with AMPs β-defensin 2 and S100 calcium binding protein A8/S100 calcium binding protein A9 (S100A8/A9) improved the phagocytic capacity of MDMs. In-depth analysis of the influence of S100A8/A9 on MDMs revealed a role for this AMP in macrophage phenotype and function. Furthermore, we show that the expression of S100A8 and S100A9 is directly regulated by WNT/β-catenin signalling, a known deregulated pathway in COPD. CONCLUSION In conclusion, for the first time, we demonstrate that airway epithelium from patients with COPD has a reduced capacity to support the phagocytic function of macrophages in response to acute NTHi exposure, and we identify the WNT/β-catenin signalling-modulated and epithelium-derived S100A8/A9 as a potent regulator of macrophage phenotype and function.
Collapse
Affiliation(s)
- Wioletta Skronska-Wasek
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH and Co KG, Biberach, Germany,Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH and Co KG, Biberach, Germany,Corresponding author: Wioletta Skronska-Wasek ()
| | - Sibel Durlanik
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH and Co KG, Biberach, Germany
| | - Huy Quang Le
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH and Co KG, Biberach, Germany
| | - Victoria Schroeder
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH and Co KG, Biberach, Germany
| | - Kerstin Kitt
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH and Co KG, Biberach, Germany
| | - James Peter Garnett
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH and Co KG, Biberach, Germany
| | - Stefan Pflanz
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH and Co KG, Biberach, Germany
| |
Collapse
|
16
|
Finicelli M, Digilio FA, Galderisi U, Peluso G. The Emerging Role of Macrophages in Chronic Obstructive Pulmonary Disease: The Potential Impact of Oxidative Stress and Extracellular Vesicle on Macrophage Polarization and Function. Antioxidants (Basel) 2022; 11:464. [PMID: 35326114 PMCID: PMC8944669 DOI: 10.3390/antiox11030464] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/20/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most common airway diseases, and it is considered a major global health problem. Macrophages are the most representative immune cells in the respiratory tract, given their role in surveying airways, removing cellular debris, immune surveillance, and resolving inflammation. Macrophages exert their functions by adopting phenotypical changes based on the stimuli they receive from the surrounding tissue. This plasticity is described as M1/M2 macrophage polarization, which consists of a strictly coordinated process leading to a difference in the expression of surface markers, the production of specific factors, and the execution of biological activities. This review focuses on the role played by macrophages in COPD and their implication in inflammatory and oxidative stress processes. Particular attention is on macrophage polarization, given macrophage plasticity is a key feature in COPD. We also discuss the regulatory influence of extracellular vesicles (EVs) in cell-to-cell communications. EV composition and cargo may influence many COPD-related aspects, including inflammation, tissue remodeling, and macrophage dysfunctions. These findings could be useful for better addressing the role of macrophages in the complex pathogenesis and outcomes of COPD.
Collapse
Affiliation(s)
- Mauro Finicelli
- Research Institute on Terrestrial Ecosystems (IRET), National Research Council of Italy (CNR), Via Pietro Castellino 111, 80131 Naples, Italy;
| | - Filomena Anna Digilio
- Research Institute on Terrestrial Ecosystems (IRET), National Research Council of Italy (CNR), Via Pietro Castellino 111, 80131 Naples, Italy;
| | - Umberto Galderisi
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via Santa Maria di Costantinopoli 16, 80138 Naples, Italy;
| | - Gianfranco Peluso
- Research Institute on Terrestrial Ecosystems (IRET), National Research Council of Italy (CNR), Via Pietro Castellino 111, 80131 Naples, Italy;
- Faculty of Medicine and Surgery, Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
| |
Collapse
|
17
|
Wang S, Chen Y, Hong W, Li B, Zhou Y, Ran P. Chronic exposure to biomass ambient particulate matter triggers alveolar macrophage polarization and activation in the rat lung. J Cell Mol Med 2022; 26:1156-1168. [PMID: 34994052 PMCID: PMC8831949 DOI: 10.1111/jcmm.17169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022] Open
Abstract
The role of alveolar macrophages (AMs) in chronic obstructive pulmonary disease is unclear. We characterized the function of AMs in rats chronically exposed to biomass fuel smoke (BMF) and studied the signal pathways that regulate AMs polarization. One hundred and eighty male Sprague‐Dawley rats were divided into BMF group and clean air control (CON) group. After BMF smoke exposure for 4 days, 1 month and 6 months, the cytokine secretion and function of AMs were determined by flow cytometry, quantitative polymerase chain reaction, Western blotting and immunofluorescence. Bone marrow‐derived macrophages were cultured and exposed to particulate matter (PM) from the smoke. Exposure initially promoted pro‐inflammatory factors, but pro‐inflammatory macrophages shared features of anti‐inflammatory macrophages. Consistent with IL‐4 upregulated in bronchoalveolar lavage fluid, p‐Stat6 and peroxisome proliferator‐activated receptor γ (PPARγ) in AMs elevated at 4 days of exposure. After 6 months of exposure, CD206, TGF‐β1 and p‐Smad3 were significantly higher than the control groups. PPARγ reversed the M1 phenotype induced by PM in vitro and drove the macrophages into the M2 phenotype. Altogether, the study demonstrates the dynamic phenotype and functional changes in AMs during exposure to BMF smoke.
Collapse
Affiliation(s)
- Shenlin Wang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Department of Respiratory Medicine, Ningxia Hui Autonomous Region People's Hospital, The First Affiliated Hospital of Northwest University for Nationalities, Yinchuan, Ningxia, China
| | - Yuhua Chen
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Hong
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Bing Li
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yumin Zhou
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Pixin Ran
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
18
|
Uddin MB, Sajib EH, Hoque SF, Hassan MM, Ahmed SSU. Macrophages in respiratory system. RECENT ADVANCEMENTS IN MICROBIAL DIVERSITY 2022:299-333. [DOI: 10.1016/b978-0-12-822368-0.00014-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
19
|
De Smedt P, Leroux-Roels G, Vandermeulen C, Tasciotti A, Di Maro G, Dozot M, Casula D, Annaratone M, Riccucci D, Arora AK. Long-term immunogenicity and safety of a non-typeable Haemophilus influenzae- Moraxella catarrhalis vaccine: 4-year follow-up of a phase 1 multicentre trial. Vaccine X 2021; 9:100124. [PMID: 34820619 PMCID: PMC8600057 DOI: 10.1016/j.jvacx.2021.100124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/09/2021] [Accepted: 10/29/2021] [Indexed: 11/24/2022] Open
Abstract
Older adults with smoking history received two doses of combined NTHi-Mcat vaccine. We evaluated antibody persistence during 4 years of follow-up after vaccination. Immune responses against the NTHi protein antigens persisted up to 4 years. There was no persistent immune response against the Mcat antigen. No safety concerns were identified during the long-term follow-up period.
A multicomponent vaccine has been developed to reduce the frequency of acute exacerbations of COPD associated with non-typeable Haemophilus influenzae (NTHi) and Moraxella catarrhalis (Mcat) infections, containing NTHi (PD and PE-PilA) and Mcat (UspA2) surface proteins. In a randomised, observer-blind, placebo-controlled study with two steps (NCT02547974), the investigational vaccine had good immunogenicity and no safety concerns were identified. In step 2, 90 adults aged 50–71 years with smoking history received two doses 60 days apart of one of two AS01E-adjuvanted formulations containing 10 µg of each antigen (10–10-AS01) or 10 µg NTHi antigens and 3.3 µg UspA2 (10–3-AS01), or placebo. Long-term persistence of antigen-specific humoral antibodies was assessed in 81 participants during 3 years of follow-up after the initial 14-month study (NCT03201211). Antigen-specific antibody concentrations were measured in blood samples taken every 6 months. Safety monitoring evaluated serious adverse events (SAEs) and potential immune-mediated disease (pIMD). Immune responses against NTHi antigens persisted up to 4 years post-vaccination. For PD, PE and PilA, at each follow-up time point, adjusted antibody geometric mean concentrations (GMCs) were higher (non-overlapping 95% confidence intervals [CIs]) in the vaccine groups versus placebo and versus pre-vaccination. Antibody GMC point estimates were higher with 10–3-AS01 than with 10–10-AS01. For UspA2, 95% CIs included 1 for GMC ratios of 10–10-AS01 or 10–3-AS01 to placebo at each time point. During follow-up, SAEs were reported in nine (11.1%) participants, one of which was fatal (lung cancer, 607 days after second 10–10-AS01 dose). One non-serious pIMD, trigeminal neuralgia, was reported 771 days after second 10–3-AS01 dose. The SAEs and pIMD were considered not related to vaccination. Immune responses against NTHi antigens persisted for 4 years after two-dose vaccination with the investigational NTHi-Mcat vaccine. There was no persistent response against the Mcat antigen. No safety concerns were identified during the long-term follow-up.
Collapse
Key Words
- AECOPD, acute exacerbations of chronic obstructive pulmonary disease
- ANCOVA, analysis of covariance
- AS01E, Adjuvant System AS01E, containing 3-O-desacyl-4′-monophosphoryl lipid A, QS-21 (Quillaja saponaria Molina, fraction 21) and liposome
- Acute exacerbation
- Antibody persistence
- CI, confidence interval
- COPD
- COPD, chronic obstructive pulmonary disease
- Clinical trial
- ELISA, enzyme-linked immunosorbent assay
- EU, enzyme-linked immunosorbent assay units
- GMC, geometric mean concentration
- GMR, geometric mean ratio
- Haemophilus influenzae
- LLOQ, lower limit of quantification
- MPL, 3-O-desacyl-4′-monophosphoryl lipid A
- Mcat, Moraxella catarrhalis
- Moraxella catarrhalis
- NTHi, non-typeable Haemophilus influenzae
- PD, protein D
- PE, protein E
- PilA, Pilin A
- QS-21, Quillaja saponaria Molina, fraction 21
- SAE, serious adverse event
- UspA2, ubiquitous surface protein A2
- pIMD, potential immune-mediated disease
Collapse
Affiliation(s)
- Philippe De Smedt
- Centre for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Geert Leroux-Roels
- Centre for Vaccinology, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Corinne Vandermeulen
- Leuven University Vaccinology Centre, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Parlak Ak T. Bronchus-Associated Lymphoid Tissue (BALT) Histology and Its Role in Various Pathologies. Vet Med Sci 2021. [DOI: 10.5772/intechopen.99366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The lower respiratory tract is in direct communication with the external environment for gas exchange to occur. Therefore, it is constantly exposed to allergens, antigens, bacteria, viruses, and a wide variety of airborne foreign bodies. Bronchus-associated lymphoid tissue (BALT), which develops in response to these exposures and is one of the most prominent representatives of mucosa-associated lymphoid tissue (MALT), is important for generating rapid and specific bronchopulmonary adaptive immune responses. Therefore, this chapter focuses on the lymphoid architecture of BALT, which was first discovered in the bronchial wall of rabbits, its inducible form called inducible BALT (iBALT), its immunological response mechanisms, and its roles in certain pathologies including infectious and autoimmune diseases as well as in allergic and malignant conditions. In conclusion, it is hypothesized that BALT plays an important role in maintaining health and in the development of lower respiratory tract diseases; thanks to the pulmonary immune system in which it functions as a functional lymphoid tissue.
Collapse
|
21
|
Lugg ST, Scott A, Parekh D, Naidu B, Thickett DR. Cigarette smoke exposure and alveolar macrophages: mechanisms for lung disease. Thorax 2021; 77:94-101. [PMID: 33986144 PMCID: PMC8685655 DOI: 10.1136/thoraxjnl-2020-216296] [Citation(s) in RCA: 198] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 11/17/2022]
Abstract
Cigarette smoking is the leading cause of preventable death worldwide. It causes chronic lung disease and predisposes individuals to acute lung injury and pulmonary infection. Alveolar macrophages are sentinel cells strategically positioned in the interface between the airway lumen and the alveolar spaces. These are the most abundant immune cells and are the first line of defence against inhaled particulates and pathogens. Recently, there has been a better understanding about the ontogeny, phenotype and function of alveolar macrophages and their role, not only in phagocytosis, but also in initiating and resolving immune response. Many of the functions of the alveolar macrophage have been shown to be dysregulated following exposure to cigarette smoke. While the mechanisms for these changes remain poorly understood, they are important in the understanding of cigarette smoking-induced lung disease. We review the mechanisms by which smoking influences alveolar macrophage: (1) recruitment, (2) phenotype, (3) immune function (bacterial killing, phagocytosis, proteinase/anti-proteinase release and reactive oxygen species production) and (4) homeostasis (surfactant/lipid processing, iron homeostasis and efferocytosis). Further understanding of the mechanisms of cigarette smoking on alveolar macrophages and other lung monocyte/macrophage populations may allow novel ways of restoring cellular function in those patients who have stopped smoking in order to reduce the risk of subsequent infection or further lung injury.
Collapse
Affiliation(s)
- Sebastian T Lugg
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Aaron Scott
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Dhruv Parekh
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Babu Naidu
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - David R Thickett
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
22
|
Bhat TA, Kalathil SG, Bogner PN, Lehmann PV, Thatcher TH, Sime PJ, Thanavala Y. AT-RvD1 Mitigates Secondhand Smoke-Exacerbated Pulmonary Inflammation and Restores Secondhand Smoke-Suppressed Antibacterial Immunity. THE JOURNAL OF IMMUNOLOGY 2021; 206:1348-1360. [PMID: 33558371 DOI: 10.4049/jimmunol.2001228] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/09/2021] [Indexed: 02/07/2023]
Abstract
Cigarette smoke is a potent proinflammatory trigger contributing to acute lung injury and the development of chronic lung diseases via mechanisms that include the impairment of inflammation resolution. We have previously demonstrated that secondhand smoke (SHS) exposure exacerbates bacterial infection-induced pulmonary inflammation and suppresses immune responses. It is now recognized that resolution of inflammation is a bioactive process mediated by lipid-derived specialized proresolving mediators that counterregulate proinflammatory signaling and promote resolution pathways. We therefore hypothesized that proresolving mediators could reduce the burden of inflammation due to chronic lung infection following SHS exposure and restore normal immune responses to respiratory pathogens. To address this question, we exposed mice to SHS followed by chronic infection with nontypeable Haemophilus influenzae (NTHI). Some groups of mice were treated with aspirin-triggered resolvin D1 (AT-RvD1) during the latter half of the smoke exposure period or during a period of smoking cessation and before infection. Treatment with AT-RvD1 markedly reduced the recruitment of neutrophils, macrophages, and T cells in lung tissue and bronchoalveolar lavage and levels of proinflammatory cytokines in the bronchoalveolar lavage. Additionally, treatment with AT-RvD1 improved Ab titers against the NTHI outer membrane lipoprotein Ag P6 following infection. Furthermore, treatment with AT-RvD1 prior to classically adjuvanted immunization with P6 increased Ag-specific Ab titers, resulting in rapid clearance of NTHI from the lungs after acute challenge. Collectively, we have demonstrated that AT-RvD1 potently reverses the detrimental effects of SHS on pulmonary inflammation and immunity and thus could be beneficial in reducing lung injury associated with smoke exposure and infection.
Collapse
Affiliation(s)
- Tariq A Bhat
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | | | - Paul N Bogner
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | | | - Thomas H Thatcher
- Department of Medicine, University of Rochester, Rochester, NY 14620; and.,Department of Environmental Medicine, University of Rochester, Rochester, NY 14620
| | - Patricia J Sime
- Department of Medicine, University of Rochester, Rochester, NY 14620; and.,Department of Environmental Medicine, University of Rochester, Rochester, NY 14620
| | - Yasmin Thanavala
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263;
| |
Collapse
|
23
|
Zhang J, Zhu C, Gao H, Liang X, Fan X, Zheng Y, Chen S, Wan Y. Identification of biomarkers associated with clinical severity of chronic obstructive pulmonary disease. PeerJ 2020; 8:e10513. [PMID: 33354437 PMCID: PMC7733647 DOI: 10.7717/peerj.10513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 11/17/2020] [Indexed: 11/20/2022] Open
Abstract
We sought to identify the biomarkers related to the clinical severity of stage I to stage IV chronic obstructive pulmonary disease (COPD). Gene expression profiles from the blood samples of COPD patients at each of the four stages were acquired from the Gene Expression Omnibus Database (GEO, accession number: GSE54837). Genes showing expression changes among the different stages were sorted by soft clustering. We performed functional enrichment, protein-protein interaction (PPI), and miRNA regulatory network analyses for the differentially expressed genes. The biomarkers associated with the clinical classification of COPD were selected from logistic regression models and the relationships between TLR2 and inflammatory factors were verified in clinical blood samples by qPCR and ELISA. Gene clusters demonstrating continuously rising or falling changes in expression (clusters 1, 2, and 7 and clusters 5, 6, and 8, respectively) from stage I to IV were defined as upregulated and downregulated genes, respectively, and further analyzed. The upregulated genes were enriched in functions associated with defense, inflammatory, or immune responses. The downregulated genes were associated with lymphocyte activation and cell activation. TLR2, HMOX1, and CD79A were hub proteins in the integrated network of PPI and miRNA regulatory networks. TLR2 and CD79A were significantly correlated with clinical classifications. TLR2 was closely associated with inflammatory responses during COPD progression. Functions associated with inflammatory and immune responses as well as lymphocyte activation may play important roles in the progression of COPD from stage I to IV. TLR2 and CD79A may serve as potential biomarkers for the clinical severity of COPD. TLR2 and CD79A may also serve as independent biomarkers in the clinical classification in COPD. TLR2 may play an important role in the inflammatory responses of COPD.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Changli Zhu
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Hong Gao
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Xun Liang
- College of Nursing and Midwifery, Jiangsu College of Nursing, Huai'an, Jiangsu, China
| | - Xiaoqian Fan
- Department of Emergency Medicine, Suqian First Hospital, Suqian, Jiangsu, China
| | - Yulong Zheng
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Song Chen
- Institute of Medicinal Biotechnology, Jiangsu College of Nursing, Huai'an, Jiangsu, China
| | - Yufeng Wan
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| |
Collapse
|
24
|
Ween MP, Moshensky A, Thredgold L, Bastian NA, Hamon R, Badiei A, Nguyen PT, Herewane K, Jersmann H, Bojanowski CM, Shin J, Reynolds PN, Crotty Alexander LE, Hodge SJ. E-cigarettes and health risks: more to the flavor than just the name. Am J Physiol Lung Cell Mol Physiol 2020; 320:L600-L614. [PMID: 33295836 DOI: 10.1152/ajplung.00370.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The growing interest in regulating flavored E-liquids must incorporate understanding of the "flavoring profile" of each E-liquid-which flavorings (flavoring chemicals) are present and at what concentrations not just focusing on the flavor on the label. We investigated the flavoring profile of 10 different flavored E-liquids. We assessed bronchial epithelial cell viability and apoptosis, phagocytosis of bacteria and apoptotic cells by macrophages after exposure to E-cigarette vapor extract (EVE). We validated our data in normal human bronchial epithelial cells (NHBE) and alveolar macrophages (AM) from healthy donors. We also assessed cytokine release and validated in the saliva from E-cigarette users. Increased necrosis/apoptosis (16.1-64.5% apoptosis) in 16HBE cells was flavor dependent, and NHBEs showed an increased susceptibility to flavors. In THP-1 differentiated macrophages phagocytosis was also flavor dependent, with AM also showing increased susceptibility to flavors. Further, Banana and Chocolate were shown to reduce surface expression of phagocytic target recognition receptors on alveolar macrophages. Banana and Chocolate increased IL-8 secretion by NHBE, whereas all 4 flavors reduced AM IL-1β secretion, which was also reduced in the saliva of E-cigarette users compared with healthy controls. Flavorant profiles of E-liquids varied from simple 2 compound mixtures to complex mixtures containing over a dozen flavorants. E-liquids with high benzene content, complex flavoring profiles, high chemical concentration had the greatest impacts. The Flavorant profile of E-liquids is key to disruption of the airway status quo by increasing bronchial epithelial cell apoptosis, causing alveolar macrophage phagocytic dysfunction, and altering airway cytokines.
Collapse
Affiliation(s)
- M P Ween
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - A Moshensky
- Pulmonary Critical Care Section, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, California
| | - L Thredgold
- Department of Occupational and Environmental Health, School of Public Health, University of Adelaide, Adelaide, South Australia, Australia
| | - N A Bastian
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - R Hamon
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - A Badiei
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - P T Nguyen
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - K Herewane
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - H Jersmann
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - C M Bojanowski
- Pulmonary Critical Care Section, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, California
| | - J Shin
- Pulmonary Critical Care Section, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, California
| | - P N Reynolds
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - L E Crotty Alexander
- Pulmonary Critical Care Section, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, California
| | - S J Hodge
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
25
|
Hilty M, Wüthrich TM, Godel A, Adelfio R, Aebi S, Burgener SS, Illgen-Wilcke B, Benarafa C. Chronic cigarette smoke exposure and pneumococcal infection induce oropharyngeal microbiota dysbiosis and contribute to long-lasting lung damage in mice. Microb Genom 2020; 6:mgen000485. [PMID: 33295863 PMCID: PMC8116676 DOI: 10.1099/mgen.0.000485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022] Open
Abstract
Environmental factors, such as cigarette smoking or lung infections, may influence chronic obstructive pulmonary disease (COPD) progression by modifying the respiratory tract microbiome. However, whether the disease itself induces or maintains dysbiosis remains undefined. In this longitudinal study, we investigated the oropharyngeal microbiota composition and disease progression of mice (in cages of 5-10 mice per cage) before, during and up to 3 months after chronic cigarette smoke exposure or exposure to room air for 6 months. Cigarette smoke exposure induced pulmonary emphysema measurable at the end of exposure for 6 months, as well as 3 months following smoke exposure cessation. Using both classical culture methods and 16S rRNA sequencing, we observed that cigarette smoke exposure altered the relative composition of the oropharyngeal microbiota and reduced its diversity (P <0.001). More than 60 taxa were substantially reduced after 6 months of smoke exposure (P <0.001) However, oropharyngeal microbiota disordering was reversed 3 months after smoke exposure cessation and no significant difference was observed compared to age-matched control mice. The effects of lung infection with Streptococcus pneumoniae on established smoke-induced emphysema and on the oropharyngeal microbiota were also evaluated. Inoculation with S. pneumoniae induced lung damage and altered the microbiota composition for a longer time compared to control groups infected but not previously exposed to smoke (P=0.01). Our data demonstrate effects of cigarette smoke and pneumococcus infection leading to altered microbiota and emphysema development. The reversal of the disordering of the microbiota composition, but not lung damage, following smoke exposure cessation and after clearance of infection suggest that changes in lung structure are not sufficient to sustain a disordered microbiota in mice. Whether changes in the airway microbiota contribute to inducing emphysema requires further investigation.
Collapse
Affiliation(s)
- Markus Hilty
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Tsering M. Wüthrich
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Freiestrasse 1, 3012 Bern, Switzerland
| | - Aurélie Godel
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland
| | - Roberto Adelfio
- Department of Infectious Diseases and Pathobiology (DIP), Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Susanne Aebi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Sabrina S. Burgener
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology (DIP), Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | | | - Charaf Benarafa
- Institute of Virology and Immunology, 3147 Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology (DIP), Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
26
|
Stockfelt M, Christenson K, Andersson A, Björkman L, Padra M, Brundin B, Ganguly K, Asgeirsdottir H, Lindén S, Qvarfordt I, Bylund J, Lindén A. Increased CD11b and Decreased CD62L in Blood and Airway Neutrophils from Long-Term Smokers with and without COPD. J Innate Immun 2020; 12:480-489. [PMID: 32829330 PMCID: PMC7734395 DOI: 10.1159/000509715] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/10/2020] [Indexed: 01/23/2023] Open
Abstract
There is incomplete mechanistic understanding of the mobilization of neutrophils in the systemic and local compartment in smokers with chronic obstructive pulmonary disease (COPD). In this pilot study, we characterized how the adhesion molecules CD11b and CD62L, surface markers indicative of priming, are altered as neutrophils extravasate, and whether surface density of CD11b and CD62L differs between long-term tobacco smokers (LTS) with and without COPD compared with healthy never-smokers (HNS). Unstimulated blood neutrophils from LTS with (n = 5) and without (n = 9) COPD displayed lower surface density of CD62L compared with HNS (n = 8). In addition, surface density of CD11b was higher in bronchoalveolar lavage (BAL) neutrophils from LTS without COPD compared with those with COPD and HNS. Moreover, in BAL neutrophils from all study groups, CD62L was lower compared with matched blood neutrophils. In addition, BAL neutrophils responded with a further decrease in CD62L to ex vivo TNF stimulation. Thus, neutrophils in the airway lumen display a higher state of priming than systemic neutrophils and bear the potential to be further primed by local cytokines even with no smoking or the presence of COPD, findings that may represent a universal host defense mechanism against local bacteria. Moreover, systemic neutrophils are primed in LTS regardless of COPD. Further studies in larger materials are warranted to determine whether the priming of neutrophils is protective against COPD or merely preceding it.
Collapse
Affiliation(s)
- Marit Stockfelt
- Section of Respiratory Medicine and Allergology, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden,
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden,
| | - Karin Christenson
- Department of Oral Microbiology & Immunology, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Anders Andersson
- COPD Center, Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, Gothenburg, Sweden
- COPD Center, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Lena Björkman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Médea Padra
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Bettina Brundin
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Koustav Ganguly
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Unit of Integrative Toxicology, Institute of Environmental Medicine (IMM), Karolinska Institutet, Stockholm, Sweden
| | - Helga Asgeirsdottir
- Section of Respiratory Medicine and Allergology, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Sara Lindén
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ingemar Qvarfordt
- Section of Respiratory Medicine and Allergology, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Johan Bylund
- Department of Oral Microbiology & Immunology, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Anders Lindén
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
27
|
Dhamodharan P, Arumugam M. Multiple Gene Expression Dataset Analysis Reveals Toll-Like Receptor Signaling Pathway is Strongly Associated With Chronic Obstructive Pulmonary Disease Pathogenesis. COPD 2020; 17:684-698. [PMID: 32757672 DOI: 10.1080/15412555.2020.1793314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chronic obstructive pulmonary disease is a complex pulmonary disease that causes airflow obstruction in humans. To identify the core genes in COPD pathogenesis, seven diverse microarray datasets (GSE475, GSE1122, GSE1650, GSE3212, GSE8823, GSE37768, and GSE22148) were downloaded from the gene expression omnibus database. All the datasets were analyzed independently with the R/Bioconductor package to screen the differentially expressed genes (DEGs). The gene ontology and pathway enrichment analysis were performed for the acquired DEGs using DAVID (Database for Annotation, Visualization, and Integrated Discovery). Further protein-protein interaction network was constructed for the DEGs and their potential hub genes and sub-networks were identified using Cytoscape software. From the selected seven datasets, 188 overlapped DEGs were perceived eventually based on considering the repetitive genes between at-least one dataset. Gene Ontology analysis reveals that most of the DEGs were significantly enriched in immune response, inflammatory response, extracellular region, lipid binding, cytokine, and chemokine activity. Moreover, genes from the sub-network analysis were again submitted to the DAVID server to validate the results which uncover the Toll-like receptor signaling pathway was significantly enriched and all the genes present in this pathway were likewise detected as hub genes from Cytoscape software. CXCL9, CXCL10, CXCL11, CCL4, TLR7, and SPP1 hub genes in the toll-like receptor signaling pathway were explored in this study as potential biomarker genes associated with COPD pathogenesis.
Collapse
Affiliation(s)
- Pavithra Dhamodharan
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Mohanapriya Arumugam
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
28
|
Singanayagam A, Glanville N, Cuthbertson L, Bartlett NW, Finney LJ, Turek E, Bakhsoliani E, Calderazzo MA, Trujillo-Torralbo MB, Footitt J, James PL, Fenwick P, Kemp SV, Clarke TB, Wedzicha JA, Edwards MR, Moffatt M, Cookson WO, Mallia P, Johnston SL. Inhaled corticosteroid suppression of cathelicidin drives dysbiosis and bacterial infection in chronic obstructive pulmonary disease. Sci Transl Med 2020; 11:11/507/eaav3879. [PMID: 31462509 DOI: 10.1126/scitranslmed.aav3879] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 02/12/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022]
Abstract
Bacterial infection commonly complicates inflammatory airway diseases such as chronic obstructive pulmonary disease (COPD). The mechanisms of increased infection susceptibility and how use of the commonly prescribed therapy inhaled corticosteroids (ICS) accentuates pneumonia risk in COPD are poorly understood. Here, using analysis of samples from patients with COPD, we show that ICS use is associated with lung microbiota disruption leading to proliferation of streptococcal genera, an effect that could be recapitulated in ICS-treated mice. To study mechanisms underlying this effect, we used cellular and mouse models of streptococcal expansion with Streptococcus pneumoniae, an important pathogen in COPD, to demonstrate that ICS impairs pulmonary clearance of bacteria through suppression of the antimicrobial peptide cathelicidin. ICS impairment of pulmonary immunity was dependent on suppression of cathelicidin because ICS had no effect on bacterial loads in mice lacking cathelicidin (Camp -/-) and exogenous cathelicidin prevented ICS-mediated expansion of streptococci within the microbiota and improved bacterial clearance. Suppression of pulmonary immunity by ICS was mediated by augmentation of the protease cathepsin D. Collectively, these data suggest a central role for cathepsin D/cathelicidin in the suppression of antibacterial host defense by ICS in COPD. Therapeutic restoration of cathelicidin to boost antibacterial immunity and beneficially modulate the lung microbiota might be an effective strategy in COPD.
Collapse
Affiliation(s)
- Aran Singanayagam
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK.
| | - Nicholas Glanville
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK
| | - Leah Cuthbertson
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Nathan W Bartlett
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK.,Faculty of Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle, Newcastle, NSW 2305, Australia
| | - Lydia J Finney
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Elena Turek
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Eteri Bakhsoliani
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK
| | | | | | - Joseph Footitt
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK
| | - Phillip L James
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Peter Fenwick
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Samuel V Kemp
- Royal Brompton Hospital, Fulham Road, London SW2 6NP, UK
| | - Thomas B Clarke
- MRC Centre for Molecular Bacteriology and Infection, Department of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Jadwiga A Wedzicha
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Michael R Edwards
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK
| | - Miriam Moffatt
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - William O Cookson
- National Heart and Lung Institute, Brompton Campus, Imperial College London, London SW3 6LY, UK
| | - Patrick Mallia
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK
| | - Sebastian L Johnston
- National Heart and Lung Institute, St Mary's Campus, Imperial College London, London W2 1PG, UK.
| |
Collapse
|
29
|
Gómez AC, Rodríguez-Fernández P, Villar-Hernández R, Gibert I, Muriel-Moreno B, Lacoma A, Prat-Aymerich C, Domínguez J. E-cigarettes: Effects in phagocytosis and cytokines response against Mycobacterium tuberculosis. PLoS One 2020; 15:e0228919. [PMID: 32040536 PMCID: PMC7010305 DOI: 10.1371/journal.pone.0228919] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 01/22/2020] [Indexed: 01/17/2023] Open
Abstract
Cigarette smoking and tuberculosis are a significant cause of death worldwide. Several epidemiological studies have demonstrated cigarette smoking is a risk factor for tuberculosis. Electronic cigarettes have recently appeared as a healthier alternative to conventional smoking, although their impact in tuberculosis is not well understood. The aim of this study was to explore the effect of electronic cigarettes in phagocytosis of Mycobacterium tuberculosis and cytokines production. In vitro infection was carried out by exposing THP-1 macrophages to four electronic vapor extracts and the intracellular burden of M. tuberculosis was determined. The percentage of infection was evaluated by confocal microscopy and the cytokine production by Luminex. A reduction of intracellular M. tuberculosis burden in THP-1 macrophages was found after its exposure to electronic vapor extract; the same trend was observed by confocal microscopy when Mycobacterium bovis BCG-GFP strain was used. Electronic cigarettes stimulate a pro-inflammatory cytokine response. We conclude that electronic cigarettes impair the phagocytic function and the cytokine response to M. tuberculosis.
Collapse
Affiliation(s)
- Andromeda-Celeste Gómez
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, CIBER Enfermedades Respiratorias (CIBERES), Badalona, Catalonia, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Bellaterra (Cerdanyola del Vallès), Barcelona, Catalonia, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Barcelona, Catalonia, Spain
| | - Pablo Rodríguez-Fernández
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, CIBER Enfermedades Respiratorias (CIBERES), Badalona, Catalonia, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Bellaterra (Cerdanyola del Vallès), Barcelona, Catalonia, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Barcelona, Catalonia, Spain
| | - Raquel Villar-Hernández
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, CIBER Enfermedades Respiratorias (CIBERES), Badalona, Catalonia, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Barcelona, Catalonia, Spain
| | - Isidre Gibert
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), Bellaterra (Cerdanyola del Vallès), Barcelona, Catalonia, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Barcelona, Catalonia, Spain
| | - Beatriz Muriel-Moreno
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, CIBER Enfermedades Respiratorias (CIBERES), Badalona, Catalonia, Spain
| | - Alicia Lacoma
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, CIBER Enfermedades Respiratorias (CIBERES), Badalona, Catalonia, Spain
| | - Cristina Prat-Aymerich
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, CIBER Enfermedades Respiratorias (CIBERES), Badalona, Catalonia, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Barcelona, Catalonia, Spain
| | - Jose Domínguez
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, CIBER Enfermedades Respiratorias (CIBERES), Badalona, Catalonia, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Barcelona, Catalonia, Spain
| |
Collapse
|
30
|
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: 55] [Impact Index Per Article: 11.0] [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
|
31
|
Methylation of Inflammatory Cells in Lung Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1255:63-72. [PMID: 32949390 DOI: 10.1007/978-981-15-4494-1_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
32
|
Valdez-Miramontes CE, Trejo Martínez LA, Torres-Juárez F, Rodríguez Carlos A, Marin-Luévano SP, de Haro-Acosta JP, Enciso-Moreno JA, Rivas-Santiago B. Nicotine modulates molecules of the innate immune response in epithelial cells and macrophages during infection with M. tuberculosis. Clin Exp Immunol 2019; 199:230-243. [PMID: 31631328 DOI: 10.1111/cei.13388] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2019] [Indexed: 01/12/2023] Open
Abstract
Smoking increases susceptibility to becoming infected with and developing tuberculosis. Among the components of cigarette smoke, nicotine has been identified as the main immunomodulatory molecule; however, its effect on the innate immune system is unknown. In the present study, the effect of nicotine on molecules of the innate immune system was evaluated. Lung epithelial cells and macrophages were infected with Mycobacterium tuberculosis (Mtb) and/or treated with nicotine. The results show that nicotine alone decreases the expression of the Toll-like receptors (TLR)-2, TLR-4 and NOD-2 in all three cell types, as well as the production of the SP-D surfactant protein in type II pneumocytes. Moreover, it was observed that nicotine decreases the production of interleukin (IL)-6 and C-C chemokine ligand (CCL)5 during Mtb infection in epithelial cells (EpCs), whereas in macrophages derived from human monocytes (MDMs) there is a decrease in IL-8, IL-6, tumor necrosis factor (TNF)-α, IL-10, CCL2, C-X-C chemokine ligand (CXCL)9 and CXCL10 only during infection with Mtb. Although modulation of the expression of cytokines and chemokines appears to be partially mediated by the nicotinic acetylcholine receptor α7, blocking this receptor found no effect on the expression of receptors and SP-D. In summary, it was found that nicotine modulates the expression of innate immunity molecules necessary for the defense against tuberculosis.
Collapse
Affiliation(s)
- C E Valdez-Miramontes
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico.,Research Center in Health Sciences and Biomedicine, San Luis Potosí, México
| | - L A Trejo Martínez
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
| | - F Torres-Juárez
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico.,Research Center in Health Sciences and Biomedicine, San Luis Potosí, México
| | - A Rodríguez Carlos
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico.,Research Center in Health Sciences and Biomedicine, San Luis Potosí, México
| | - S P Marin-Luévano
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico.,Research Center in Health Sciences and Biomedicine, San Luis Potosí, México
| | - J P de Haro-Acosta
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
| | - J A Enciso-Moreno
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
| | - B Rivas-Santiago
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
| |
Collapse
|
33
|
Arora S, Ahmad S, Irshad R, Goyal Y, Rafat S, Siddiqui N, Dev K, Husain M, Ali S, Mohan A, Syed MA. TLRs in pulmonary diseases. Life Sci 2019; 233:116671. [PMID: 31336122 PMCID: PMC7094289 DOI: 10.1016/j.lfs.2019.116671] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/16/2019] [Accepted: 07/19/2019] [Indexed: 12/21/2022]
Abstract
Toll-like receptors (TLRs) comprise a clan of proteins involved in identification and triggering a suitable response against pathogenic attacks. As lung is steadily exposed to multiple infectious agents, antigens and host-derived danger signals, the inhabiting stromal and myeloid cells of the lung express an aggregate of TLRs which perceive the endogenously derived damage-associated molecular patterns (DAMPs) along with pathogen associated molecular patterns (PAMPs) and trigger the TLR-associated signalling events involved in host defence. Thus, they form an imperative component of host defence activation in case of microbial infections as well as non-infectious pulmonary disorders such as interstitial lung disease, acute lung injury and airways disease, such as COPD and asthma. They also play an equally important role in lung cancer. Targeting the TLR signalling network would pave ways to the design of more reliable and effective vaccines against infectious agents and control deadly infections, desensitize allergens and reduce inflammation. Moreover, TLR agonists may act as adjuvants by increasing the efficiency of cancer vaccines, thereby contributing their role in treatment of lung cancer too. Overall, TLRs present a compelling and expeditiously bolstered area of research and addressing their signalling events would be of significant use in pulmonary diseases.
Collapse
Affiliation(s)
- Shweta Arora
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Shaniya Ahmad
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Rasha Irshad
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Yamini Goyal
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Sahar Rafat
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Neha Siddiqui
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Kapil Dev
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Mohammad Husain
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| | - Shakir Ali
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India.
| | - Anant Mohan
- Department of Pulmonary Medicine, AIIMS, New Delhi, India.
| | - Mansoor Ali Syed
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
34
|
Huang C, Shi G. Smoking and microbiome in oral, airway, gut and some systemic diseases. J Transl Med 2019; 17:225. [PMID: 31307469 PMCID: PMC6632217 DOI: 10.1186/s12967-019-1971-7] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 07/05/2019] [Indexed: 12/24/2022] Open
Abstract
The human microbiome harbors a diverse array of microbes which establishes a mutually beneficial relation with the host in healthy conditions, however, the dynamic homeostasis is influenced by both host and environmental factors. Smoking contributes to modifications of the oral, lung and gut microbiome, leading to various diseases, such as periodontitis, asthma, chronic obstructive pulmonary disease, Crohn’s disease, ulcerative colitis and cancers. However, the exact causal relationship between smoking and microbiome alteration remains to be further explored.
Collapse
Affiliation(s)
- Chunrong Huang
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197, Rui Jin Er Road, Shanghai, 200025, People's Republic of China.,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, 197, Rui Jin Er Road, Shanghai, 200025, People's Republic of China
| | - Guochao Shi
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197, Rui Jin Er Road, Shanghai, 200025, People's Republic of China. .,Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, 197, Rui Jin Er Road, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
35
|
Huang P, Wei S, Huang W, Wu P, Chen S, Tao A, Wang H, Liang Z, Chen R, Yan J, Zhang Q. Hydrogen gas inhalation enhances alveolar macrophage phagocytosis in an ovalbumin-induced asthma model. Int Immunopharmacol 2019; 74:105646. [PMID: 31200337 DOI: 10.1016/j.intimp.2019.05.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 05/14/2019] [Accepted: 05/17/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Maintaining an airway clear of bacteria, foreign particles and apoptotic cells by alveolar macrophages is very essential for lung homeostasis. In asthma, the phagocytic capacity of alveolar macrophages is significantly reduced, which is thought to be associated with increased oxidative stress. Hydrogen (H2) has been shown to exert potent antioxidant and anti-inflammatory effects, yet its effects on phagocytosis of alveolar macrophages are unknown. This study is aimed to evaluate the beneficial effects of hydrogen gas inhalation on alveolar macrophage phagocytosis in an ovalbumin (OVA)-induced murine asthma model. METHODS Female C57BL/6 mice were intraperitoneally sensitized with OVA before they were subject to airway challenge with aerosolized OVA. Hydrogen gas was delivered to the mice through inhalation twice a day (2 h once) for 7 consecutive days. Phagocytic function of alveolar macrophages isolated from bronchoalveolar lavage fluid was assessed by fluorescence-labeled Escherichia coli as well as flow cytometry. RESULTS Alveolar macrophages isolated from OVA-induced asthmatic mice showed decreased phagocytic capacity to Escherichia coli when compared with those of control mice. Defective phagocytosis in asthmatic mice was reversed by hydrogen gas inhalation. Hydrogen gas inhalation significantly alleviated OVA-induced airway hyperresponsiveness, inflammation and goblet cell hyperplasia, diminished TH2 response and decreased IL-4 as well as IgE levels, reduced malondialdehyde (MDA) production and increased superoxide dismutase (SOD) activity. Concomitantly, hydrogen gas inhalation inhibited NF-κB activation and markedly activated Nrf2 pathway in OVA-induced asthmatic mice. CONCLUSIONS Our findings demonstrated that hydrogen gas inhalation enhanced alveolar macrophage phagocytosis in OVA-induced asthmatic mice, which may be associated with the antioxidant effects of hydrogen gas and the activation of the Nrf2 pathway.
Collapse
Affiliation(s)
- Peikai Huang
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Department of Respiratory Medicine, Huizhou Municipal Central Hospital, Huizhou, China
| | - Shushan Wei
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weihua Huang
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Penghui Wu
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuyu Chen
- The Second Affiliated Hospital of Guangzhou Medical University, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Ailin Tao
- The Second Affiliated Hospital of Guangzhou Medical University, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Hongyu Wang
- Firestone Institute for Respiratory Health, The Research Institute of St. Joe's Hamilton, St. Joseph's Healthcare; Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Zhenyu Liang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Rongchang Chen
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jie Yan
- The Second Affiliated Hospital of Guangzhou Medical University, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.
| | - Qingling Zhang
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
36
|
Van Damme P, Leroux-Roels G, Vandermeulen C, De Ryck I, Tasciotti A, Dozot M, Moraschini L, Testa M, Arora AK. Safety and immunogenicity of non-typeable Haemophilus influenzae-Moraxella catarrhalis vaccine. Vaccine 2019; 37:3113-3122. [DOI: 10.1016/j.vaccine.2019.04.041] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 03/26/2019] [Accepted: 04/14/2019] [Indexed: 01/28/2023]
|
37
|
Provost KA, Smith M, Miller-Larsson A, Gudleski GD, Sethi S. Bacterial regulation of macrophage bacterial recognition receptors in COPD are differentially modified by budesonide and fluticasone propionate. PLoS One 2019; 14:e0207675. [PMID: 30677037 PMCID: PMC6345465 DOI: 10.1371/journal.pone.0207675] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 11/05/2018] [Indexed: 11/18/2022] Open
Abstract
Rationale Patients with COPD have an increased risk for community-acquired pneumonia, which is further increased by inhaled corticosteroids. Objective To assess effects of the corticosteroids, budesonide and fluticasone propionate, on macrophage bacterial responses in COPD. Methods Monocyte-derived macrophages (MDMs) generated from blood monocytes from 10 non-smoker controls (NoS), 20 smokers without COPD (Sm), and 40 subjects with moderate to severe COPD (21 ex-smokers (COPD-ES) and 19 current smokers (COPD-S)) were pre-treated with budesonide or fluticasone (10 nM—1 μM) and challenged with live non-typeable Haemophilus influenzae (NTHI) or Streptococcus pneumoniae (SP). Cell surface bacterial recognition receptor expression (flow cytometry) and cytokine release (bead array) were analyzed. Results NTHI and SP reduced bacterial recognition receptor expression on MDMs from COPD and Sm, but not NoS (except TLR4). SR-AI and MARCO were reduced by both NTHI and SP, whereas other receptors by either NTHI or SP. Among COPD subjects, COPD-ES demonstrated a greater number of reductions as compared to COPD-S. NTHI reduced SR-AI, MARCO, CD11b, CD35 and CD206 in COPD-ES while only SR-AI and CD11b in COPD-S. SP reduced SRA-1, CD1d, TLR2 and TLR4 in both COPD-ES and COPD-S, and reduced MARCO and CD93 only in COPD-ES. All receptors reduced in COPD by NTHI and most by SP, were also reduced in Sm. Budesonide counteracted the receptor reductions induced by both NTHI (CD206 p = 0.03, MARCO p = 0.08) and SP (SR-AI p = 0.02) in COPD-ES. Fluticasone counteracted only SP-induced reductions in TLR2 (p = 0.008 COPD-ES and p = 0.04 COPD-S) and TLR4 (p = 0.02 COPD-ES). Cytokine release was equivalently reduced by both corticosteroids. Conclusions Reduction in macrophage bacterial recognition receptors during bacterial exposure could provide a mechanism for the increased pneumonia risk in COPD. Differential effects of budesonide and fluticasone propionate on macrophage bacterial recognition receptor expression may contribute to the higher pneumonia incidence reported with fluticasone propionate.
Collapse
Affiliation(s)
- Karin A. Provost
- Veterans Health Administration, Veterans Affairs Western New York Healthcare System at Buffalo, Division of Pulmonary, Critical Care and Sleep Medicine, Buffalo, New York, United States of America
- University at Buffalo, State University of New York, Jacobs School of Medicine and Biomedical Sciences, Division of Pulmonary, Critical Care and Sleep Medicine, Buffalo, New York, United States of America
- * E-mail:
| | - Miyuki Smith
- Veterans Health Administration, Veterans Affairs Western New York Healthcare System at Buffalo, Division of Pulmonary, Critical Care and Sleep Medicine, Buffalo, New York, United States of America
| | | | - Gregory D. Gudleski
- University at Buffalo, State University of New York, Jacobs School of Medicine and Biomedical Sciences, Department of Medicine, Buffalo, New York, United States of America
| | - Sanjay Sethi
- Veterans Health Administration, Veterans Affairs Western New York Healthcare System at Buffalo, Division of Pulmonary, Critical Care and Sleep Medicine, Buffalo, New York, United States of America
- University at Buffalo, State University of New York, Jacobs School of Medicine and Biomedical Sciences, Division of Pulmonary, Critical Care and Sleep Medicine, Buffalo, New York, United States of America
| |
Collapse
|
38
|
Pro-inflammatory effects of extracellular Hsp70 and cigarette smoke in primary airway epithelial cells from COPD patients. Biochimie 2019; 156:47-58. [DOI: 10.1016/j.biochi.2018.09.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/25/2018] [Indexed: 11/18/2022]
|
39
|
Hulina-Tomašković A, Rajković MG, Somborac-Bačura A, Čeri A, Dabelić S, Rumora L. Extracellular Hsp70 modulates the inflammatory response of cigarette smoke extract in NCI-H292 cells. Exp Physiol 2018; 103:1704-1716. [PMID: 30298576 DOI: 10.1113/ep087180] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/08/2018] [Indexed: 12/19/2022]
Abstract
NEW FINDINGS What is the central question of this study? Does extracellular heat shock protein 70 (eHsp70) alter cigarette smoke extract (CSE)-induced inflammatory responses in NCI-H292 bronchial epithelial cells? What is the main finding and its importance? eHsp70 modulates inflammatory responses and TLR2, TLR4 and Hsp70 gene expression, and protects NCI-H292 cells against CSE-induced cytotoxicity. eHsp70 might be implicated in development of inflammatory diseases affected by cigarette smoke, such as COPD. ABSTRACT One of the major risk factors for development of chronic obstructive pulmonary disease (COPD) is cigarette smoke. Extracellular Hsp70 (eHsp70) is increased in sera of COPD patients, and can act as damage-associated molecular pattern (DAMP). In this study, we explored inflammatory parameters (cytokine concentrations, Toll-like receptor (TLR) 2 and 4 and Hsp70 expression, mitogen-activated protein kinases (MAPKs) and nuclear factor κB (NF-κB) activation, and cytotoxicity) after exposure of bronchial-epithelial NCI-H292 cells to cigarette smoke extract (CSE) alone (2.5 and 15%) and in combinations with recombinant human (rh) Hsp70 (0.3, 1 and 3 μg ml-1 ). We applied specific MAPKs, NF-κB and Hsp70 inhibitors to elucidate rhHsp70 inflammation-associated responses. CSE alone and combinations of 15% CSE with rhHsp70 stimulated IL-1α, IL-6 and IL-8 release. However, rhHsp70 applied with 2.5% CSE decreased secretion of cytokines indicating antagonistic effects. Individual and combined treatments with 2.5% CSE suppressed TLR2 expression. CSE at 15% induced TLR2 and TLR4 gene expression, whereas rhHsp70 abolished that effect. rhHsp70 and 15% CSE alone reduced, while their combination increased, intracellular Hsp70 mRNA level. CSE alone and in combination with rhHsp70 activated extracellular signal-regulated kinase and p38 MAPKs, while inhibition of MAPKs, NF-κB and Hsp70 attenuated IL-6 and IL-8 secretion. CSE at 15% reduced cell viability and induced apoptosis, as shown by MTS and caspases-3/7 assays. CSE at 2.5% alone stimulated lactate dehydrogenase release, but cellular membrane integrity remained intact in co-treatments with rhHsp70. rhHsp70 might modulate the inflammatory response of CSE and could also protect NCI-H292 cells against CSE cytotoxicity. Those effects are implemented via MAPK and NF-κB signalling pathways.
Collapse
Affiliation(s)
- Andrea Hulina-Tomašković
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Medical Biochemistry and Hematology, Zagreb, Croatia
| | - Marija Grdić Rajković
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Medical Biochemistry and Hematology, Zagreb, Croatia
| | - Anita Somborac-Bačura
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Medical Biochemistry and Hematology, Zagreb, Croatia
| | - Andrea Čeri
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Medical Biochemistry and Hematology, Zagreb, Croatia
| | - Sanja Dabelić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Biochemistry and Molecular Biology, Zagreb, Croatia
| | - Lada Rumora
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Medical Biochemistry and Hematology, Zagreb, Croatia
| |
Collapse
|
40
|
Gleeson LE, O’Leary SM, Ryan D, McLaughlin AM, Sheedy FJ, Keane J. Cigarette Smoking Impairs the Bioenergetic Immune Response to Mycobacterium tuberculosis Infection. Am J Respir Cell Mol Biol 2018; 59:572-579. [DOI: 10.1165/rcmb.2018-0162oc] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Laura E. Gleeson
- Department of Respiratory Medicine, and
- TB Immunology Research Group, School of Medicine, Trinity Translational Medicine Institute, St. James’s Hospital, Dublin, Ireland; and
| | - Seonadh M. O’Leary
- TB Immunology Research Group, School of Medicine, Trinity Translational Medicine Institute, St. James’s Hospital, Dublin, Ireland; and
| | | | | | - Frederick J. Sheedy
- TB Immunology Research Group, School of Medicine, Trinity Translational Medicine Institute, St. James’s Hospital, Dublin, Ireland; and
- Macrophage Homeostasis Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Joseph Keane
- Department of Respiratory Medicine, and
- TB Immunology Research Group, School of Medicine, Trinity Translational Medicine Institute, St. James’s Hospital, Dublin, Ireland; and
| |
Collapse
|
41
|
Effects of cigarette smoke on immunity, neuroinflammation and multiple sclerosis. J Neuroimmunol 2018; 329:24-34. [PMID: 30361070 DOI: 10.1016/j.jneuroim.2018.10.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 09/30/2018] [Accepted: 10/05/2018] [Indexed: 12/18/2022]
Abstract
Cigarette smoking is the most prominent significant cause of death and morbidity. It is recognised as a risk factor for a number of immune mediated, inflammatory diseases including multiple sclerosis (MS). Here, we review the complex immunological effects of smoking on the immune system, which include enhancement of inflammatory responses with a parallel reduction of some immune defences, resulting in an increased susceptibility to infection and a persistent proinflammatory environment. We discuss the effect of smoking on the susceptibility, clinical course, disability, and mortality in MS, the likely benefits of smoking cessation, and the specific immunological effects of smoking in MS. In conclusion, smoking is an important environmental risk factor for MS occurrence and outcome, and it acts in significant part through immunological mechanisms.
Collapse
|
42
|
Chakraborty A, Boer JC, Selomulya C, Plebanski M, Royce SG. Insights into endotoxin-mediated lung inflammation and future treatment strategies. Expert Rev Respir Med 2018; 12:941-955. [PMID: 30221563 DOI: 10.1080/17476348.2018.1523009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Airway inflammatory disorders are prevalent diseases in need of better management and new therapeutics. Immunotherapies offer a solution to the problem of corticosteroid resistance. Areas covered: The current review focuses on lipopolysaccharide (Gram-negative bacterial endotoxin)-mediated inflammation in the lung and the animal models used to study related diseases. Endotoxin-induced lung pathology is usually initiated by antigen presenting cells (APC). We will discuss different subsets of APC including lung dendritic cells and macrophages, and their role in responding to endotoxin and environmental challenges. Expert commentary: The pharmacotherapeutic considerations to combat airway inflammation should cost-effectively improve quality of life with sustainable and safe strategies. Selectively targeting APCs in the lung offer the potential for a promising new strategy for the better management and treatment of inflammatory lung disease.
Collapse
Affiliation(s)
- Amlan Chakraborty
- a Department of Chemical Engineering , Monash University , Clayton , Australia.,b Department of Immunology and Pathology , Central Clinical School, Monash University , Melbourne , Australia
| | - Jennifer C Boer
- b Department of Immunology and Pathology , Central Clinical School, Monash University , Melbourne , Australia
| | - Cordelia Selomulya
- a Department of Chemical Engineering , Monash University , Clayton , Australia
| | - Magdalena Plebanski
- b Department of Immunology and Pathology , Central Clinical School, Monash University , Melbourne , Australia.,c School of Health and Biomedical Sciences and Enabling Capability platforms, Biomedical and Health Innovation , RMIT University , Melbourne , Australia
| | - Simon G Royce
- d Central Clinical School , Monash University , Clayton , Victoria , Australia.,e Department of Pharmacology , Monash University , Clayton , Australia
| |
Collapse
|
43
|
Mubarak RA, Roberts N, Mason RJ, Alper S, Chu HW. Comparison of pro- and anti-inflammatory responses in paired human primary airway epithelial cells and alveolar macrophages. Respir Res 2018; 19:126. [PMID: 29940963 PMCID: PMC6020222 DOI: 10.1186/s12931-018-0825-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/08/2018] [Indexed: 12/27/2022] Open
Abstract
Background Airway epithelial cells and alveolar macrophages (AMs) are the first line of defense in the lung during infection. Toll-like receptor (TLR) agonists have been extensively used to define the regulation of inflammation in these cells. However, previous studies were performed in non-paired airway epithelial cells and AMs. The major goal of our study was to compare the pro- and anti-inflammatory responses of paired human primary airway epithelial cells and AMs to TLR3 and TLR4 agonists. Methods Tracheobronchial epithelial cells (TBEC) and AMs from four smokers and four non-smokers without lung disease were cultured with or without Poly(I:C) (PIC) (a TLR3 agonist) or LPS (a TLR4 agonist) for 4, 24 and 48 h. The immune responses of paired cells were compared. Results TBEC and AMs showed stronger pro-inflammatory cytokine (e.g., IL-8) responses to PIC and LPS, respectively. TLR3 and TLR4 mRNA levels were similar in non-stimulated TBEC and AMs. However, PIC stimulation in AMs led to sustained up-regulation of the immune negative regulators Tollip and A20, which may render AMs less sensitive to PIC stimulation than TBEC. Unlike AMs, TBEC did not increase NF-κB activation after LPS stimulation. Interestingly, smoking status was correlated with less TLR3 and IRAK-M expression in non-stimulated TBEC, but not in AMs. PIC-stimulated TBEC and LPS-stimulated AMs from smokers vs. non-smokers produced more IL-8. Finally, we show that expression of A20 and IRAK-M is strongly correlated in the two paired cell types. Conclusions By using paired airway epithelial cells and AMs, this study reveals how these two critical types of lung cells respond to viral and bacterial pathogen associated molecular patterns, and provides rationale for modulating immune negative regulators to prevent excessive lung inflammation during respiratory infection. Electronic supplementary material The online version of this article (10.1186/s12931-018-0825-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Reem Al Mubarak
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Room A639, Denver, CO, 80206, USA
| | - Nicole Roberts
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Room A639, Denver, CO, 80206, USA
| | - Robert J Mason
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Room A639, Denver, CO, 80206, USA
| | - Scott Alper
- Department of Biomedical Research and Center for Genes, Environment, and Health, National Jewish Health, University of Colorado, 1400 Jackson Street, Denver, CO, 80206, USA. .,Department of Immunology and Microbiology, University of Colorado, 1400 Jackson Street, Denver, CO, 80206, USA.
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Room A639, Denver, CO, 80206, USA.
| |
Collapse
|
44
|
Strzelak A, Ratajczak A, Adamiec A, Feleszko W. Tobacco Smoke Induces and Alters Immune Responses in the Lung Triggering Inflammation, Allergy, Asthma and Other Lung Diseases: A Mechanistic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E1033. [PMID: 29883409 PMCID: PMC5982072 DOI: 10.3390/ijerph15051033] [Citation(s) in RCA: 370] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/08/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023]
Abstract
Many studies have been undertaken to reveal how tobacco smoke skews immune responses contributing to the development of chronic obstructive pulmonary disease (COPD) and other lung diseases. Recently, environmental tobacco smoke (ETS) has been linked with asthma and allergic diseases in children. This review presents the most actual knowledge on exact molecular mechanisms responsible for the skewed inflammatory profile that aggravates inflammation, promotes infections, induces tissue damage, and may promote the development of allergy in individuals exposed to ETS. We demonstrate how the imbalance between oxidants and antioxidants resulting from exposure to tobacco smoke leads to oxidative stress, increased mucosal inflammation, and increased expression of inflammatory cytokines (such as interleukin (IL)-8, IL-6 and tumor necrosis factor α ([TNF]-α). Direct cellular effects of ETS on epithelial cells results in increased permeability, mucus overproduction, impaired mucociliary clearance, increased release of proinflammatory cytokines and chemokines, enhanced recruitment of macrophages and neutrophils and disturbed lymphocyte balance towards Th2. The plethora of presented phenomena fully justifies a restrictive policy aiming at limiting the domestic and public exposure to ETS.
Collapse
Affiliation(s)
- Agnieszka Strzelak
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Aleksandra Ratajczak
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Aleksander Adamiec
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Wojciech Feleszko
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| |
Collapse
|
45
|
Ween MP, Whittall JJ, Hamon R, Reynolds PN, Hodge SJ. Phagocytosis and Inflammation: Exploring the effects of the components of E-cigarette vapor on macrophages. Physiol Rep 2018; 5:5/16/e13370. [PMID: 28867672 PMCID: PMC5582261 DOI: 10.14814/phy2.13370] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 06/26/2017] [Accepted: 06/28/2017] [Indexed: 01/24/2023] Open
Abstract
E‐cigarettes are perceived as harmless; however, evidence of their safety is lacking. New data suggests E‐cigarettes discharge a range of compounds capable of physiological damage to users. We previously established that cigarette smoke caused defective alveolar macrophage phagocytosis. The present study compared the effect E‐cigarette of components; E‐liquid flavors, nicotine, vegetable glycerine, and propylene glycol on phagocytosis, proinflammatory cytokine secretion, and phagocytic recognition molecule expression using differentiated THP‐1 macrophages. Similar to CSE, phagocytosis of NTHi bacteria was significantly decreased by E‐liquid flavoring (11.65–15.75%) versus control (27.01%). Nicotine also decreased phagocytosis (15.26%). E‐liquid, nicotine, and E‐liquid+ nicotine reduced phagocytic recognition molecules; SR‐A1 and TLR‐2. IL‐8 secretion increased with flavor and nicotine, while TNFα, IL‐1β, IL‐6, MIP‐1α, MIP‐1β, and MCP‐1 decreased after exposure to most flavors and nicotine. PG, VG, or PG:VG mix also induced a decrease in MIP‐1α and MIP‐1β. We conclude that E‐cigarettes can cause macrophage phagocytic dysfunction, expression of phagocytic recognition receptors and cytokine secretion pathways. As such, E‐cigarettes should be treated with caution by users, especially those who are nonsmokers.
Collapse
Affiliation(s)
- Miranda P Ween
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia .,School of Medicine, University of Adelaide, Adelaide, Australia
| | - Jonathan J Whittall
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,School of Medicine, University of Adelaide, Adelaide, Australia
| | - Rhys Hamon
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,School of Medicine, University of Adelaide, Adelaide, Australia
| | - Paul N Reynolds
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,School of Medicine, University of Adelaide, Adelaide, Australia
| | - Sandra J Hodge
- School of Medicine, University of Adelaide, Adelaide, Australia
| |
Collapse
|
46
|
Cessation from Smoking Improves Innate Host Defense and Clearance of Experimentally Inoculated Nasal Staphylococcus aureus. Infect Immun 2018; 86:IAI.00912-17. [PMID: 29311241 DOI: 10.1128/iai.00912-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/02/2018] [Indexed: 12/17/2022] Open
Abstract
Staphylococcus aureus nasal carriage is transient in most humans and usually benign, but dissemination of S. aureus to extranasal sites causes the majority of clinical infections, and S. aureus is a major cause of serious infections in the United States. A better understanding of innate nasal decolonization mechanisms is urgently needed, as are relevant models for studying S. aureus clearance. Here, we screened a population of healthy smokers for nasal S. aureus carriage and compared the participants' abilities to clear experimentally applied nasal S. aureus before and after completion of a smoking cessation program. We determined that cigarette smoking increases the mean nasal S. aureus load (2.6 × 104 CFU/swab) compared to the load observed in healthy nonsmokers (1.7 × 103 CFU/swab) and might increase the rate of S. aureus nasal carriage in otherwise-healthy adults: 22 of 99 smokers carried S. aureus at the screening visit, while only 4 of 30 nonsmokers screened positive during the same time period. Only 6 of 19 experimental inoculation studies in active smokers resulted in S. aureus clearance within the month of follow-up, while in the cessation group, 6 of 9 subjects cleared nasal S. aureus and carriage duration averaged 21 ± 4 days. Smoking cessation associated with enhanced expression of S. aureus-associated interleukin-1β (IL-1β) and granulocyte colony-stimulating factor (G-CSF) in nasal fluids. Participants who failed to clear S. aureus exhibited a higher nasal S. aureus load and elevated nasal interleukin-1 receptor antagonist (IL-1RA) expression at the preexperiment study visits. We conclude that smokers exhibit higher S. aureus loads than nonsmokers and that innate immune pathways, including G-CSF expression and signaling through the IL-1 axis, are important mediators of nasal S. aureus clearance.
Collapse
|
47
|
Bhat TA, Kalathil SG, Bogner PN, Miller A, Lehmann PV, Thatcher TH, Phipps RP, Sime PJ, Thanavala Y. Secondhand Smoke Induces Inflammation and Impairs Immunity to Respiratory Infections. THE JOURNAL OF IMMUNOLOGY 2018; 200:2927-2940. [PMID: 29555783 DOI: 10.4049/jimmunol.1701417] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/16/2018] [Indexed: 12/29/2022]
Abstract
Despite advocacy to reduce smoking-related diseases, >1 billion people worldwide continue to smoke. Smoking is immunosuppressive and an important etiological factor in the development of several human disorders including respiratory diseases like chronic obstructive pulmonary disease. However, there is a critical gap in the knowledge of the role of secondhand smoke (SHS) in inflammation and immunity. We therefore studied the influence of SHS on pulmonary inflammation and immune responses to respiratory infection by nontypeable Haemophilus influenzae (NTHI) recurrently found in chronic obstructive pulmonary disease patients. Chronic SHS-exposed mice were chronically infected with NTHI and pulmonary inflammation was evaluated by histology. Immune cell numbers and cytokines were measured by flow cytometry and ELISA, respectively. Chronic SHS exposure impaired NTHI P6 Ag-specific B and T cell responses following chronic NTHI infection as measured by ELISPOT assays, reduced the production of Abs in serum and bronchoalveolar lavage, and enhanced albumin leak into the bronchoalveolar lavage as determined by ELISA. Histopathological examination of lungs revealed lymphocytic accumulation surrounding airways and bronchovasculature following chronic SHS exposure and chronic infection. Chronic SHS exposure enhanced the levels of inflammatory cytokines IL-17A, IL-6, IL-1β, and TNF-α in the lungs, and impaired the generation of adaptive immunity following either chronic infection or P6 vaccination. Chronic SHS exposure diminished bacterial clearance from the lungs after acute NTHI challenge, whereas P6 vaccination improved clearance equivalent to the level seen in air-exposed, non-vaccinated mice. Our study provides unequivocal evidence that SHS exposure has long-term detrimental effects on the pulmonary inflammatory microenvironment and immunity to infection and vaccination.
Collapse
Affiliation(s)
- Tariq A Bhat
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | | | - Paul N Bogner
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Austin Miller
- Department of Biostatistics, Roswell Park Cancer Institute, Buffalo, NY 14263
| | | | - Thomas H Thatcher
- Department of Medicine, University of Rochester, Rochester, NY 14620; and
| | - Richard P Phipps
- Department of Medicine, University of Rochester, Rochester, NY 14620; and.,Department of Environmental Medicine, University of Rochester, Rochester, NY 14620
| | - Patricia J Sime
- Department of Medicine, University of Rochester, Rochester, NY 14620; and.,Department of Environmental Medicine, University of Rochester, Rochester, NY 14620
| | - Yasmin Thanavala
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263;
| |
Collapse
|
48
|
Dysregulated Functions of Lung Macrophage Populations in COPD. J Immunol Res 2018; 2018:2349045. [PMID: 29670919 PMCID: PMC5835245 DOI: 10.1155/2018/2349045] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 11/29/2017] [Indexed: 01/02/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a diverse respiratory disease characterised by bronchiolitis, small airway obstruction, and emphysema. Innate immune cells play a pivotal role in the disease's progression, and in particular, lung macrophages exploit their prevalence and strategic localisation to orchestrate immune responses. To date, alveolar and interstitial resident macrophages as well as blood monocytes have been described in the lungs of patients with COPD contributing to disease pathology by changes in their functional repertoire. In this review, we summarise recent evidence from human studies and work with animal models of COPD with regard to altered functions of each of these myeloid cell populations. We primarily focus on the dysregulated capacity of alveolar macrophages to secrete proinflammatory mediators and proteases, induce oxidative stress, engulf microbes and apoptotic cells, and express surface and intracellular markers in patients with COPD. In addition, we discuss the differences in the responses between alveolar macrophages and interstitial macrophages/monocytes in the disease and propose how the field should advance to better understand the implications of lung macrophage functions in COPD.
Collapse
|
49
|
Haw TJ, Starkey MR, Pavlidis S, Fricker M, Arthurs AL, Nair PM, Liu G, Hanish I, Kim RY, Foster PS, Horvat JC, Adcock IM, Hansbro PM. Toll-like receptor 2 and 4 have opposing roles in the pathogenesis of cigarette smoke-induced chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2018; 314:L298-L317. [PMID: 29025711 PMCID: PMC5866502 DOI: 10.1152/ajplung.00154.2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 09/08/2017] [Accepted: 10/03/2017] [Indexed: 12/18/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of morbidity and death and imposes major socioeconomic burdens globally. It is a progressive and disabling condition that severely impairs breathing and lung function. There is a lack of effective treatments for COPD, which is a direct consequence of the poor understanding of the underlying mechanisms involved in driving the pathogenesis of the disease. Toll-like receptor (TLR)2 and TLR4 are implicated in chronic respiratory diseases, including COPD, asthma and pulmonary fibrosis. However, their roles in the pathogenesis of COPD are controversial and conflicting evidence exists. In the current study, we investigated the role of TLR2 and TLR4 using a model of cigarette smoke (CS)-induced experimental COPD that recapitulates the hallmark features of human disease. TLR2, TLR4, and associated coreceptor mRNA expression was increased in the airways in both experimental and human COPD. Compared with wild-type (WT) mice, CS-induced pulmonary inflammation was unaltered in TLR2-deficient ( Tlr2-/-) and TLR4-deficient ( Tlr4-/-) mice. CS-induced airway fibrosis, characterized by increased collagen deposition around small airways, was not altered in Tlr2-/- mice but was attenuated in Tlr4-/- mice compared with CS-exposed WT controls. However, Tlr2-/- mice had increased CS-induced emphysema-like alveolar enlargement, apoptosis, and impaired lung function, while these features were reduced in Tlr4-/- mice compared with CS-exposed WT controls. Taken together, these data highlight the complex roles of TLRs in the pathogenesis of COPD and suggest that activation of TLR2 and/or inhibition of TLR4 may be novel therapeutic strategies for the treatment of COPD.
Collapse
Affiliation(s)
- Tatt Jhong Haw
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Malcolm R Starkey
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
- Priority Research Centre for Grow Up Well, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Stelios Pavlidis
- The Airways Disease Section, National Heart and Lung Institute, Imperial College London , London , United Kingdom
| | - Michael Fricker
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Anya L Arthurs
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Prema M Nair
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Gang Liu
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Irwan Hanish
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor , Malaysia
| | - Richard Y Kim
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| | - Ian M Adcock
- The Airways Disease Section, National Heart and Lung Institute, Imperial College London , London , United Kingdom
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute and University of Newcastle, Callaghan, New South Wales , Australia
| |
Collapse
|
50
|
Mishra V, Banga J, Silveyra P. Oxidative stress and cellular pathways of asthma and inflammation: Therapeutic strategies and pharmacological targets. Pharmacol Ther 2018; 181:169-182. [PMID: 28842273 PMCID: PMC5743757 DOI: 10.1016/j.pharmthera.2017.08.011] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Asthma is a complex inflammatory disease characterized by airway inflammation and hyperresponsiveness. The mechanisms associated with the development and progression of asthma have been widely studied in multiple populations and animal models, and these have revealed involvement of various cell types and activation of intracellular signaling pathways that result in activation of inflammatory genes. Significant contributions of Toll-like-receptors (TLRs) and transcription factors such as NF-кB, have been reported as major contributors to inflammatory pathways. These have also recently been associated with mechanisms of oxidative biology. This is of important clinical significance as the observed inefficacy of current available treatments for severe asthma is widely attributed to oxidative stress. Therefore, targeting oxidizing molecules in conjunction with inflammatory mediators and transcription factors may present a novel therapeutic strategy for asthma. In this review, we summarize TLRs and NF-кB pathways in the context of exacerbation of asthma pathogenesis and oxidative biology, and we discuss the potential use of polyphenolic flavonoid compounds, known to target these pathways and possess antioxidant activity, as potential therapeutic agents for asthma.
Collapse
Affiliation(s)
- Vikas Mishra
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Departments of Pediatrics, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Jaspreet Banga
- The Feinstein Institute for Medical Research, Center for Autoimmune and Musculoskeletal Diseases, Manhasset, NY, USA
| | - Patricia Silveyra
- Departments of Pediatrics, The Pennsylvania State University, College of Medicine, Hershey, PA, USA; Biochemistry and Molecular Biology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.
| |
Collapse
|