1
|
Ding K, Jiang W, Zhan W, Xiong C, Chen J, Wang Y, Jia H, Lei M. The therapeutic potential of quercetin for cigarette smoking-induced chronic obstructive pulmonary disease: a narrative review. Ther Adv Respir Dis 2023; 17:17534666231170800. [PMID: 37154390 PMCID: PMC10170608 DOI: 10.1177/17534666231170800] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
Quercetin is a flavonoid with antioxidant and anti-inflammatory properties. Quercetin has potentially beneficial therapeutic effects for several diseases, including cigarette smoking-induced chronic obstructive pulmonary disease (CS-COPD). Many studies have shown that quercetin's antioxidant and anti-inflammatory properties have positive therapeutic potential for CS-COPD. In addition, quercetin's immunomodulatory, anti-cellular senescence, mitochondrial autophagy-modulating, and gut microbiota-modulating effects may also have therapeutic value for CS-COPD. However, there appears to be no review of the possible mechanisms of quercetin for treating CS-COPD. Moreover, the combination of quercetin with common therapeutic drugs for CS-COPD needs further refinement. Therefore, in this article, after introducing the definition and metabolism of quercetin, and its safety, we comprehensively presented the pathogenesis of CS-COPD related to oxidative stress, inflammation, immunity, cellular senescence, mitochondrial autophagy, and gut microbiota. We then reviewed quercetin's anti-CS-COPD effects, performed by influencing these mechanisms. Finally, we explored the possibility of using quercetin with commonly used drugs for treating CS-COPD, providing a basis for future screening of excellent drug combinations for treating CS-COPD. This review has provided meaningful information on quercetin's mechanisms and clinical use in treating CS-COPD.
Collapse
Affiliation(s)
- Kaixi Ding
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Jiang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenling Zhan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunping Xiong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jieling Chen
- Shehong Hospital of Traditional Chinese Medicine, Shehong, China
| | - Yu Wang
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Huanan Jia
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Ming Lei
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| |
Collapse
|
2
|
Liu G, Jarnicki AG, Paudel KR, Lu W, Wadhwa R, Philp AM, Van Eeckhoutte H, Marshall JE, Malyla V, Katsifis A, Fricker M, Hansbro NG, Dua K, Kermani NZ, Eapen MS, Tiotiu A, Chung KF, Caramori G, Bracke K, Adcock IM, Sohal SS, Wark PA, Oliver BG, Hansbro PM. Adverse roles of mast cell chymase-1 in COPD. Eur Respir J 2022; 60:2101431. [PMID: 35777766 DOI: 10.1183/13993003.01431-2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/08/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND COPD is the third leading cause of death worldwide. Cigarette smoke (CS)-induced chronic inflammation inducing airway remodelling, emphysema and impaired lung function is the primary cause. Effective therapies are urgently needed. Human chymase (hCMA)1 and its orthologue mCMA1/mouse mast cell protease (mMCP)5 are exocytosed from activated mast cells and have adverse roles in numerous disorders, but their role in COPD is unknown. METHODS We evaluated hCMA1 levels in lung tissues of COPD patients. We used mmcp5-deficient (-/-) mice to evaluate this protease's role and potential for therapeutic targeting in CS-induced experimental COPD. In addition, we used ex vivo/in vitro studies to define mechanisms. RESULTS The levels of hCMA1 mRNA and CMA1+ mast cells were increased in lung tissues from severe compared to early/mild COPD patients, non-COPD smokers and healthy controls. Degranulated mast cell numbers and mMCP5 protein were increased in lung tissues of wild-type mice with experimental COPD. mmcp5 -/- mice were protected against CS-induced inflammation and macrophage accumulation, airway remodelling, emphysema and impaired lung function in experimental COPD. CS extract challenge of co-cultures of mast cells from wild-type, but not mmcp5 -/- mice with wild-type lung macrophages increased in tumour necrosis factor (TNF)-α release. It also caused the release of CMA1 from human mast cells, and recombinant hCMA-1 induced TNF-α release from human macrophages. Treatment with CMA1 inhibitor potently suppressed these hallmark features of experimental COPD. CONCLUSION CMA1/mMCP5 promotes the pathogenesis of COPD, in part, by inducing TNF-α expression and release from lung macrophages. Inhibiting hCMA1 may be a novel treatment for COPD.
Collapse
Affiliation(s)
- Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Andrew G Jarnicki
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Keshav R Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Wenying Lu
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, Australia
| | - Ridhima Wadhwa
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Ashleigh M Philp
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
- St Vincent's Medical School, University of New South Wales Medicine, University of New South Wales, Sydney, Australia
| | - Hannelore Van Eeckhoutte
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Jacqueline E Marshall
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Vamshikrishna Malyla
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Angelica Katsifis
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Michael Fricker
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Kamal Dua
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
| | - Nazanin Z Kermani
- Data Science Institute, Department of Computing, Imperial College London, London, UK
| | - Mathew S Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, Australia
| | - Angelica Tiotiu
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Pulmonology, University Hospital of Nancy, Nancy, France
| | - K Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Gaetano Caramori
- UOC di Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Ken Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sukhwinder S Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, Australia
| | - Peter A Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia
| | - Brian G Oliver
- Woolcock Institute and School of Life Science, Faculty of Science Life Science, University of Technology Sydney, Sydney, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia
| |
Collapse
|
3
|
Taucher E, Mykoliuk I, Lindenmann J, Smolle-Juettner FM. Implications of the Immune Landscape in COPD and Lung Cancer: Smoking Versus Other Causes. Front Immunol 2022; 13:846605. [PMID: 35386685 PMCID: PMC8978964 DOI: 10.3389/fimmu.2022.846605] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 12/30/2022] Open
Abstract
Cigarette smoking is reported in about one third of adults worldwide. A strong relationship between cigarette smoke exposure and chronic obstructive pulmonary disease (COPD) as well as lung cancer has been proven. However, about 15% of lung cancer cases, and between one fourth and one third of COPD cases, occur in never-smokers. The effects of cigarette smoke on the innate as well as the adaptive immune system have been widely investigated. It is assumed that certain immunologic features contribute to lung cancer and COPD development in the absence of smoking as the major risk factor. In this article, we review different immunological aspects of lung cancer and COPD with a special focus on non-smoking related risk factors.
Collapse
Affiliation(s)
- Elisabeth Taucher
- Division of Pulmonology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Iurii Mykoliuk
- Division of Thoracic Surgery, Department of Surgery, Medical University Graz, Graz, Austria
| | - Joerg Lindenmann
- Division of Thoracic Surgery, Department of Surgery, Medical University Graz, Graz, Austria
| | | |
Collapse
|
4
|
Moschovis PP, Lu M, Hayden D, Yonker LM, Lombay J, Taveras E, Arauz Boudreau A, Triant VA, Foulkes AS, Bassett I, Hibberd PL, Kinane TB. Effect modification by age of the association between obstructive lung diseases, smoking, and COVID-19 severity. BMJ Open Respir Res 2021; 8:8/1/e001038. [PMID: 34740944 PMCID: PMC8573665 DOI: 10.1136/bmjresp-2021-001038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/11/2021] [Indexed: 11/21/2022] Open
Abstract
Introduction Obstructive lung diseases (asthma and chronic obstructive pulmonary disease (COPD)) and smoking are associated with greater risk of respiratory infections and hospitalisations, but conflicting data exist regarding their association with severity of COVID-19, and few studies have evaluated whether these associations differ by age. Objectives To examine the associations between asthma, COPD and smoking on the severity of COVID-19 among a cohort of hospitalised patients, and to test for effect modification by age. Methods We performed a retrospective analysis of electronic health record data of patients admitted to Massachusetts General Hospital, assigning the maximal WHO Clinical Progression Scale score for each patient during the first 28 days following hospital admission. Using ordered logistic regression, we measured the association between maximal severity score and asthma, COPD and smoking and their interaction with age. Measurements and main results Among 1391 patients hospitalised with COVID-19, we found an increased risk of severe disease among patients with COPD and prior smoking, independent of age. We also found evidence of effect modification by age with asthma and current smoking; in particular, asthma was associated with decreased COVID-19 severity among older adults, and current smoking was associated with decreased severity among younger patients. Conclusions This cohort study identifies age as a modifying factor for the association between asthma and smoking on severity of COVID-19. Our findings highlight the complexities of determining risk factors for COVID-19 severity, and suggest that the effect of risk factors may vary across the age spectrum.
Collapse
Affiliation(s)
- Peter P Moschovis
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mengdi Lu
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Douglas Hayden
- Biostatistics Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lael M Yonker
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jesiel Lombay
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Elsie Taveras
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexy Arauz Boudreau
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Virginia A Triant
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Andrea S Foulkes
- Biostatistics Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ingrid Bassett
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Patricia L Hibberd
- Department of Global Health, Boston University School of Public Health, Boston, Massachusetts, USA
| | - T Bernard Kinane
- Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Dobric A, De Luca SN, Spencer SJ, Bozinovski S, Saling MM, McDonald CF, Vlahos R. Novel pharmacological strategies to treat cognitive dysfunction in chronic obstructive pulmonary disease. Pharmacol Ther 2021; 233:108017. [PMID: 34626675 DOI: 10.1016/j.pharmthera.2021.108017] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/19/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a major incurable global health burden and currently the 3rd largest cause of death in the world, with approximately 3.23 million deaths per year. Globally, the financial burden of COPD is approximately €82 billion per year and causes substantial morbidity and mortality. Importantly, much of the disease burden and health care utilisation in COPD is associated with the management of its comorbidities and viral and bacterial-induced acute exacerbations (AECOPD). Recent clinical studies have shown that cognitive dysfunction is present in up to 60% of people with COPD, with impairments in executive function, memory, and attention, impacting on important outcomes such as quality of life, hospitalisation and survival. The high prevalence of cognitive dysfunction in COPD may also help explain the insufficient adherence to therapeutic plans and strategies, thus worsening disease progression in people with COPD. However, the mechanisms underlying the impaired neuropathology and cognition in COPD remain largely unknown. In this review, we propose that the observed pulmonary oxidative burden and inflammatory response of people with COPD 'spills over' into the systemic circulation, resulting in damage to the brain and leading to cognitive dysfunction. As such, drugs targeting the lungs and comorbidities concurrently represent an exciting and unique therapeutic opportunity to treat COPD and cognitive impairments, which may lead to the production of novel targets to prevent and reverse the debilitating and life-threatening effects of cognitive dysfunction in COPD.
Collapse
Affiliation(s)
- Aleksandar Dobric
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Simone N De Luca
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Sarah J Spencer
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia; ARC Centre of Excellence for Nanoscale Biophotonics, RMIT University, Melbourne, VIC, Australia
| | - Steven Bozinovski
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Michael M Saling
- Clinical Neuropsychology, The University of Melbourne and Austin Health, VIC, Australia
| | - Christine F McDonald
- Institute for Breathing and Sleep, Austin Health, Melbourne, VIC, Australia; Department of Respiratory & Sleep Medicine, The University of Melbourne and Austin Health, Melbourne, VIC, Australia
| | - Ross Vlahos
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia.
| |
Collapse
|
6
|
Gangwar A, Paul S, Arya A, Ahmad Y, Bhargava K. Altitude acclimatization via hypoxia-mediated oxidative eustress involves interplay of protein nitrosylation and carbonylation: A redoxomics perspective. Life Sci 2021; 296:120021. [PMID: 34626604 DOI: 10.1016/j.lfs.2021.120021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/22/2021] [Accepted: 09/30/2021] [Indexed: 12/17/2022]
Abstract
AIM Hypoxia is an important feature of multiple diseases like cancer and obesity and also an environmental stressor to high altitude travelers. Emerging research suggests the importance of redox signaling in physiological responses transforming the notion of oxidative stress into eustress and distress. However, the behavior of redox protein post-translational modifications (PTMs), and their correlation with stress acclimatization in humans remains sketchy. Scant information exists about modifications in redoxome during physiological exposure to environmental hypoxia. In this study, we investigated redox PTMs, nitrosylation and carbonylation, in context of extended environmental hypoxia exposure. METHODS The volunteers were confirmed to be free of any medical conditions and matched for age and weight. The human global redoxome and the affected networks were investigated using TMT-labeled quantitative proteo-bioinformatics and biochemical assays. The percolator PSM algorithm was used for peptide-spectrum match (PSM) validation in database searches. The FDR for peptide matches was set to 0.01. 1-way ANOVA and Tukey's Multiple Comparison test were used for biochemical assays. p-value<0.05 was considered statistically significant. Three independent experiments (biological replicates) were performed. Results were presented as Mean ± standard error of mean (SEM). KEY FINDINGS This investigation revealed direct and indirect interplay between nitrosylation and carbonylation especially within coagulation and inflammation networks; interlinked redox signaling (via nitrosylation‑carbonylation); and novel nitrosylation and carbonylation sites in individual proteins. SIGNIFICANCE This study elucidates the role of redox PTMs in hypoxia signaling favoring tolerance and survival. Also, we demonstrated direct and indirect interplay between nitrosylation and carbonylation is crucial to extended hypoxia tolerance.
Collapse
Affiliation(s)
- Anamika Gangwar
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India
| | - Subhojit Paul
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India
| | - Aditya Arya
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India
| | - Yasmin Ahmad
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India.
| | - Kalpana Bhargava
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India.
| |
Collapse
|
7
|
Lee KH, Woo J, Kim JY, Lee CH, Yoo CG. Cigarette smoke extract-induced downregulation of p300 is responsible for the impaired inflammatory cytokine response of macrophages. Cell Signal 2021; 85:110050. [PMID: 34044126 DOI: 10.1016/j.cellsig.2021.110050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 01/22/2023]
Abstract
Patients with chronic obstructive pulmonary disease (COPD) are susceptible to infection owing to the impaired immune function of alveolar macrophages. This is presumed to be caused, at least partially, by cigarette smoke (CS), which is a major risk factor for COPD. Although CS has been reported to inhibit Toll-like receptor (TLR) function and phagocytosis in macrophages, the molecular mechanism of CS-mediated impairment of macrophage immune function has not been completely elucidated. We investigated the effects of CS extracts (CSE) on macrophage immune function and its molecular mechanism. We assessed lipopolysaccharide (LPS, TLR4 ligand)-, Pam3CSK4 (TLR2 ligand)-, or CpG-oligodeoxynucleotide (TLR9 ligand)-induced IL-6, TNF-α, and IL-1β production in macrophages. Upregulation of IL-6, TNF-α, and IL-1β mRNA and protein by TLR ligands was suppressed on treatment with CSE. However, LPS-induced MAP kinase activation, IκBα degradation, and nuclear translocation of NF-κB were not impeded by CSE. In contrast, CSE significantly suppressed NF-κB transcriptional activity in the nucleus. We found that p300, which acetylates RelA/p65 at lysine 310, and acetyl-p65 (K310) were downregulated upon CSE treatment. Knock-down of p300 suppressed LPS-induced acetylation of NF-κB p65 and production of inflammatory cytokine. To summarize, these results suggest that CSE impair cytokine response by decreasing the expression levels of p300.
Collapse
Affiliation(s)
- Kyoung-Hee Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Jisu Woo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Ji Yeon Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Chang-Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Chul-Gyu Yoo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| |
Collapse
|
8
|
Papanicolaou A, Wang H, Satzke C, Vlahos R, Wilson N, Bozinovski S. Novel Therapies for Pneumonia-Associated Severe Asthma Phenotypes. Trends Mol Med 2020; 26:1047-1058. [PMID: 32828703 DOI: 10.1016/j.molmed.2020.07.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/17/2020] [Accepted: 07/21/2020] [Indexed: 12/13/2022]
Abstract
Distinct asthma phenotypes are emerging from well-defined cohort studies and appear to be associated with a history of pneumonia. Asthmatics are more susceptible to infections caused by Streptococcus pneumoniae; however, the mechanisms that underlie defective immunity to this pathogen are still being elucidated. Here, we discuss how alternatively activated macrophages (AAMs) in asthmatics are defective in bacterial phagocytosis and how respiratory viruses disrupt essential host immunity to cause bacterial dispersion deeper into the lungs. We also describe how respiratory pathogens instigate neutrophilic inflammation and amplify type-2 inflammation in asthmatics. Finally, we propose novel dual-acting strategies including granulocyte-colony-stimulating factor receptor (G-CSFR) antagonism and specialised pro-resolving mediators (SPMs) to suppress type-2 and neutrophilic inflammation without compromising pathogen clearance.
Collapse
Affiliation(s)
- Angelica Papanicolaou
- Chronic Infectious and Inflammatory Disease Research Program, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Hao Wang
- Chronic Infectious and Inflammatory Disease Research Program, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Catherine Satzke
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia; Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Ross Vlahos
- Chronic Infectious and Inflammatory Disease Research Program, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | | | - Steven Bozinovski
- Chronic Infectious and Inflammatory Disease Research Program, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.
| |
Collapse
|
9
|
Becerra-Diaz M, Song M, Heller N. Androgen and Androgen Receptors as Regulators of Monocyte and Macrophage Biology in the Healthy and Diseased Lung. Front Immunol 2020; 11:1698. [PMID: 32849595 PMCID: PMC7426504 DOI: 10.3389/fimmu.2020.01698] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
Androgens, the predominant male sex hormones, drive the development and maintenance of male characteristics by binding to androgen receptor (AR). As androgens are systemically distributed throughout the whole organism, they affect many tissues and cell types in addition to those in male sexual organs. It is now clear that the immune system is a target of androgen action. In the lungs, many immune cells express ARs and are responsive to androgens. In this review, we describe the effects of androgens and ARs on lung myeloid immune cells-monocytes and macrophages-as they relate to health and disease. In particular, we highlight the effect of androgens on lung diseases, such as asthma, chronic obstructive pulmonary disease and lung fibrosis. We also discuss the therapeutic use of androgens and how circulating androgens correlate with lung disease. In addition to human studies, we also discuss how mouse models have helped to uncover the effect of androgens on monocytes and macrophages in lung disease. Although the role of estrogen and other female hormones has been broadly analyzed in the literature, we focus on the new perspectives of androgens as modulators of the immune system that target myeloid cells during lung inflammation.
Collapse
Affiliation(s)
| | | | - Nicola Heller
- Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
10
|
Lee KH, Woo J, Kim J, Lee CH, Yoo CG. Cigarette smoke extract decreased basal and lipopolysaccharide-induced expression of MARCO via degradation of p300. Respirology 2020; 26:102-111. [PMID: 32512637 DOI: 10.1111/resp.13867] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/15/2020] [Accepted: 04/30/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND OBJECTIVE Alveolar macrophages of patients with COPD display impaired cytokine release and diminished phagocytosis. COPD exacerbations exhibit immune dysfunction towards the respiratory pathogens. CS and CSE were reported to aggravate bacterial infections in COPD patients. METHODS MARCO is highly expressed in lungs and is involved in pathogen clearance. We investigated the effect of CSE on MARCO expression and its regulatory mechanisms. After relevant siRNA transfection and treatment with CSE and/or LPS, we measured the levels of MARCO by q-RT PCR, immunoblotting and flow cytometry. Immunofluorescence staining and immunoprecipitation were used to evaluate the mechanism. RESULTS CSE decreased LPS-induced expression of MARCO mRNA and protein. Upregulation of MARCO by LPS was Nrf2-dependent. Nrf2 knockdown significantly suppressed LPS-induced increase in MARCO transcripts. CSE did not block nuclear translocation of Nrf2 in LPS-treated cells, but rather CSE itself strongly accumulated Nrf2 in the nucleus through the degradation of its cytoplasmic inhibitor, KEAP1. However, CSE markedly suppressed LPS-induced Nrf2 acetylation. Histone acetyltransferase p300/CBP directly acetylates Nrf2, which augments promoter-specific DNA binding of Nrf2. Our results reveal CSE-induced polyubiquitinylation and subsequent degradation of p300 via the proteasome. Pretreatment with proteasome inhibitors completely blocked CSE-induced degradation of p300 and suppression of MARCO expression. CONCLUSION These findings suggest that CSE decreases MARCO expression via the proteasomal degradation of p300 in macrophages, which may be in part responsible for impaired bacterial phagocytosis.
Collapse
Affiliation(s)
- Kyoung-Hee Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Jisu Woo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Jiyeon Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Chang-Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Chul-Gyu Yoo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
11
|
Zhou L, Le Y, Tian J, Yang X, Jin R, Gai X, Sun Y. Cigarette smoke-induced RANKL expression enhances MMP-9 production by alveolar macrophages. Int J Chron Obstruct Pulmon Dis 2018; 14:81-91. [PMID: 30587964 PMCID: PMC6304243 DOI: 10.2147/copd.s190023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background and purpose Cigarette smoke (CS) induces alveolar destruction through overproduction of proteinases including matrix metalloproteinase (MMP)-9 by alveolar macrophages (AMs). Receptor activator of nuclear factor-κB ligand (RANKL) functions in immune regulation and cytokine secretion; whether it is involved in CS-induced MMP-9 expression is unknown. The purpose of our study was to investigate the expression and functional role of RANKL pathway in MMP-9 production pertaining to the pathogenesis of COPD. Materials and methods We first localized RANKL and its receptor RANK in the lungs of mice exposed to long-term CS exposure. Next, we studied RANKL and RANK expression under CS extract (CSE) stimulation in vitro. Lastly, we studied the in vitro biological function of RANKL in CS-induced production of MMP-9. Results Both RANKL and RANK were highly expressed in AMs in CS-exposed mice, but not in the control mice. In vitro, CSE increased the expressions of RANKL and RANK in macrophages. AMs responded to CSE and RANKL stimulation by overexpressing MMP-9, and CSE-induced MMP-9 expression was partly blocked by using monoclonal anti-RANKL antibody. Conclusion RANKL/RANK pathway mediates CS-induced MMP-9 expression in AMs, suggesting a novel mechanism for CS-associated emphysema.
Collapse
Affiliation(s)
- Lu Zhou
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China,
| | - Yanqing Le
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China,
| | - Jieyu Tian
- Department of Respiratory Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xia Yang
- Department of Respiratory Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Rong Jin
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaoyan Gai
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China,
| | - Yongchang Sun
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China,
| |
Collapse
|
12
|
Wang H, Anthony D, Selemidis S, Vlahos R, Bozinovski S. Resolving Viral-Induced Secondary Bacterial Infection in COPD: A Concise Review. Front Immunol 2018; 9:2345. [PMID: 30459754 PMCID: PMC6232692 DOI: 10.3389/fimmu.2018.02345] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 09/21/2018] [Indexed: 11/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a leading cause of disability and death world-wide, where chronic inflammation accelerates lung function decline. Pathological inflammation is worsened by chronic bacterial lung infections and susceptibility to recurrent acute exacerbations of COPD (AECOPD), typically caused by viral and/or bacterial respiratory pathogens. Despite ongoing efforts to reduce AECOPD rates with inhaled corticosteroids, COPD patients remain at heightened risk of developing serious lung infections/AECOPD, frequently leading to hospitalization and infection-dependent delirium. Here, we review emerging mechanisms into why COPD patients are susceptible to chronic bacterial infections and highlight dysregulated inflammation and production of reactive oxygen species (ROS) as central causes. This underlying chronic infection leaves COPD patients particularly vulnerable to acute viral infections, which further destabilize host immunity to bacteria. The pathogeneses of bacterial and viral exacerbations are significant as clinical symptoms are more severe and there is a marked increase in neutrophilic inflammation and tissue damage. AECOPD triggered by a bacterial and viral co-infection increases circulating levels of the systemic inflammatory marker, serum amyloid A (SAA). SAA is a functional agonist for formyl peptide receptor 2 (FPR2/ALX), where it promotes chemotaxis and survival of neutrophils. Excessive levels of SAA can antagonize the protective actions of FPR2/ALX that involve engagement of specialized pro-resolving mediators, such as resolvin-D1. We propose that the anti-microbial and anti-inflammatory actions of specialized pro-resolving mediators, such as resolvin-D1 should be harnessed for the treatment of AECOPD that are complicated by the co-pathogenesis of viruses and bacteria.
Collapse
Affiliation(s)
- Hao Wang
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Desiree Anthony
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Ross Vlahos
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
13
|
Wang H, FitzPatrick M, Wilson NJ, Anthony D, Reading PC, Satzke C, Dunne EM, Licciardi PV, Seow HJ, Nichol K, Adcock IM, Chung KF, Anderson GP, Vlahos R, Wark P, Bozinovski S. CSF3R/CD114 mediates infection-dependent transition to severe asthma. J Allergy Clin Immunol 2018; 143:785-788.e6. [PMID: 30312710 DOI: 10.1016/j.jaci.2018.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/20/2018] [Accepted: 10/01/2018] [Indexed: 11/18/2022]
Affiliation(s)
- Hao Wang
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Meaghan FitzPatrick
- Department of Pharmacology & Therapeutics, Lung Health Research Centre, The University of Melbourne, Parkville, Victoria, Australia; CSL Limited, Parkville, Victoria, Australia
| | | | - Desiree Anthony
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Patrick C Reading
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia; Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Catherine Satzke
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia; Pneumococcal Research, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Eileen M Dunne
- Pneumococcal Research, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul V Licciardi
- Pneumococcal Research, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Huei Jiunn Seow
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Kristy Nichol
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Ian M Adcock
- Airways Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kian Fan Chung
- Airways Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Gary P Anderson
- Department of Pharmacology & Therapeutics, Lung Health Research Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Ross Vlahos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia; Department of Pharmacology & Therapeutics, Lung Health Research Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Peter Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia; Department of Pharmacology & Therapeutics, Lung Health Research Centre, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
14
|
Ramos CDO, Campos KKD, Costa GDP, Cangussú SD, Talvani A, Bezerra FS. Taurine treatment decreases inflammation and oxidative stress in lungs of adult mice exposed to cigarette smoke. Regul Toxicol Pharmacol 2018; 98:50-57. [PMID: 30026134 DOI: 10.1016/j.yrtph.2018.07.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 02/02/2023]
Abstract
Taurine is the major free amino acid found in mammalian cells and is known to be an antioxidant and membrane-stabilizing agent. This study aimed to evaluate the effects of taurine on oxidative stress and inflammatory response in the lungs of mice exposed to cigarette smoke. Fifty male C57BL/6 mice were divided into 5 groups: control group (CG), vehicle group (VG), taurine group (TG), cigarette smoke group (CSG), and cigarette smoke + taurine group (CSTG). For five consecutive days, CSG and CSTG were exposed to 4 cigarettes 3 times a day. Taurine administration was able to reduce total leukocytes in bronchoalveolar lavage fluid in CSTG compared to CSG. There was an increase in antioxidant superoxide dismutase and catalase activity in CSG compared to that in CG and TG, and a decrease in CSTG compared to CSG. There was an increase in the concentration of TNF and IL-17 in CSG and CSTG compared to CG and TG. There was an increase in the concentration of IL-22 in CSG compared to CG and TG, and a decrease in CSTG compared to CSG. The administration of taurine has been shown to reduce the inflammation and oxidative stress induced by short-term exposure to cigarette smoke.
Collapse
Affiliation(s)
- Camila de Oliveira Ramos
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG, Brazil
| | - Keila Karine Duarte Campos
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG, Brazil
| | - Guilherme de Paula Costa
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences(NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG, Brazil
| | - Sílvia Dantas Cangussú
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences(NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG, Brazil
| | - Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG, Brazil.
| |
Collapse
|
15
|
Hamon R, Tran HB, Roscioli E, Ween M, Jersmann H, Hodge S. Bushfire smoke is pro-inflammatory and suppresses macrophage phagocytic function. Sci Rep 2018; 8:13424. [PMID: 30194323 PMCID: PMC6128914 DOI: 10.1038/s41598-018-31459-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 08/03/2018] [Indexed: 12/03/2022] Open
Abstract
Bushfires are increasing in frequency and severity worldwide. Bushfire smoke contains organic/inorganic compounds including aldehydes and acrolein. We described suppressive effects of tobacco smoke on the phagocytic capacity of airway macrophages, linked to secondary necrosis of uncleared apoptotic epithelial cells, persistence of non-typeable H. influenzae (NTHi), and inflammation. We hypothesised that bushfire smoke extract (BFSE) would similarly impair macrophage function. THP-1 or monocyte-derived macrophages (MDM) were exposed to 1-10% BFSE prepared from foliage of 5 common Australian native plants (genus Acacia or Eucalyptus), or 10% cigarette smoke extract (CSE). Phagocytic recognition receptors were measured by flow cytometry; pro-inflammatory cytokines and caspase 1 by immunofluorescence or cytometric bead array; viability by LDH assay; and capsase-3/PARP by western blot. BFSE significantly decreased phagocytosis of apoptotic cells or NTHi by both THP-1 macrophages and MDM vs air control, consistent with the effects of CSE. BFSE significantly decreased MDM expression of CD36, CD44, SR-A1, CD206 and TLR-2 and increased active IL-1β, caspase-1 and secreted IL-8. BFSE dose-dependently decreased THP-1 macrophage viability (5-fold increase in LDH at 10%) and significantly increased active caspase-3. BFSE impairs macrophage function to a similar extent as CSE, highlighting the need for further research, especially in patients with pre-existing lung disease.
Collapse
Affiliation(s)
- Rhys Hamon
- Chronic Inflammatory Lung Disease Research Laboratory, Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia
- Department of Medicine, University of Adelaide, Adelaide, Australia
| | - Hai B Tran
- Chronic Inflammatory Lung Disease Research Laboratory, Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia
- Department of Medicine, University of Adelaide, Adelaide, Australia
| | - Eugene Roscioli
- Chronic Inflammatory Lung Disease Research Laboratory, Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia
- Department of Medicine, University of Adelaide, Adelaide, Australia
| | - Miranda Ween
- Chronic Inflammatory Lung Disease Research Laboratory, Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia
- Department of Medicine, University of Adelaide, Adelaide, Australia
| | - Hubertus Jersmann
- Chronic Inflammatory Lung Disease Research Laboratory, Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia
- Department of Medicine, University of Adelaide, Adelaide, Australia
| | - Sandra Hodge
- Chronic Inflammatory Lung Disease Research Laboratory, Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.
- Department of Medicine, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Aspirin-triggered resolvin D1 reduces pneumococcal lung infection and inflammation in a viral and bacterial coinfection pneumonia model. Clin Sci (Lond) 2017; 131:2347-2362. [DOI: 10.1042/cs20171006] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/27/2017] [Accepted: 08/03/2017] [Indexed: 01/15/2023]
Abstract
Formyl peptide receptor 2/lipoxin A4 (LXA4) receptor (Fpr2/ALX) co-ordinates the transition from inflammation to resolution during acute infection by binding to distinct ligands including serum amyloid A (SAA) and Resolvin D1 (RvD1). Here, we evaluated the proresolving actions of aspirin-triggered RvD1 (AT-RvD1) in an acute coinfection pneumonia model. Coinfection with Streptococcus pneumoniae and influenza A virus (IAV) markedly increased pneumococcal lung load and neutrophilic inflammation during the resolution phase. Fpr2/ALX transcript levels were increased in the lungs of coinfected mice, and immunohistochemistry identified prominent Fpr2/ALX immunoreactivity in bronchial epithelial cells and macrophages. Levels of circulating and lung SAA were also highly increased in coinfected mice. Therapeutic treatment with exogenous AT-RvD1 during the acute phase of infection (day 4–6 post-pneumococcal inoculation) significantly reduced the pneumococcal load. AT-RvD1 also significantly reduced neutrophil elastase (NE) activity and restored total antimicrobial activity in bronchoalveolar lavage (BAL) fluid (BALF) of coinfected mice. Pneumonia severity, as measured by quantitating parenchymal inflammation or alveolitis was significantly reduced with AT-RvD1 treatment, which also reduced the number of infiltrating lung neutrophils and monocytes/macrophages as assessed by flow cytometry. The reduction in distal lung inflammation in AT-RvD1-treated mice was not associated with a significant reduction in inflammatory and chemokine mediators. In summary, we demonstrate that in the coinfection setting, SAA levels were persistently increased and exogenous AT-RvD1 facilitated more rapid clearance of pneumococci in the lungs, while concurrently reducing the severity of pneumonia by limiting excessive leukocyte chemotaxis from the infected bronchioles to distal areas of the lungs.
Collapse
|
19
|
Dalle-Donne I, Colombo G, Gornati R, Garavaglia ML, Portinaro N, Giustarini D, Bernardini G, Rossi R, Milzani A. Protein Carbonylation in Human Smokers and Mammalian Models of Exposure to Cigarette Smoke: Focus on Redox Proteomic Studies. Antioxid Redox Signal 2017; 26:406-426. [PMID: 27393565 DOI: 10.1089/ars.2016.6772] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
SIGNIFICANCE Oxidative stress is one mechanism whereby tobacco smoking affects human health, as reflected by increased levels of several biomarkers of oxidative stress/damage isolated from tissues and biological fluids of active and passive smokers. Many investigations of cigarette smoke (CS)-induced oxidative stress/damage have been carried out in mammalian animal and cellular models of exposure to CS. Animal models allow the investigation of many parameters that are similar to those measured in human smokers. In vitro cell models may provide new information on molecular and functional differences between cells of smokers and nonsmokers. Recent Advances: Over the past decade or so, a growing number of researches highlighted that CS induces protein carbonylation in different tissues and body fluids of smokers as well as in in vivo and in vitro models of exposure to CS. CRITICAL ISSUES We review recent findings on protein carbonylation in smokers and models thereof, focusing on redox proteomic studies. We also discuss the relevance and limitations of these models of exposure to CS and critically assess the congruence between the smoker's condition and laboratory models. FUTURE DIRECTIONS The identification of protein targets is crucial for understanding the mechanism(s) by which carbonylated proteins accumulate and potentially affect cellular functions. Recent progress in redox proteomics allows the enrichment, identification, and characterization of specific oxidative protein modifications, including carbonylation. Therefore, redox proteomics can be a powerful tool to gain new insights into the onset and/or progression of CS-related diseases and to develop strategies to prevent and/or treat them. Antioxid. Redox Signal. 26, 406-426.
Collapse
Affiliation(s)
| | - Graziano Colombo
- 1 Department of Biosciences, Università degli Studi di Milano , Milan, Italy
| | - Rosalba Gornati
- 2 Department of Biotechnology and Life Sciences, University of Insubria , Varese, Italy
| | - Maria L Garavaglia
- 1 Department of Biosciences, Università degli Studi di Milano , Milan, Italy
| | - Nicola Portinaro
- 3 Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano and Pediatric Orthopaedic Unit, Humanitas Clinical and Research Center , Rozzano (Milan), Italy
| | | | - Giovanni Bernardini
- 2 Department of Biotechnology and Life Sciences, University of Insubria , Varese, Italy
| | - Ranieri Rossi
- 4 Department of Life Sciences, University of Siena , Siena, Italy
| | - Aldo Milzani
- 1 Department of Biosciences, Università degli Studi di Milano , Milan, Italy
| |
Collapse
|
20
|
Heulens N, Korf H, Mathyssen C, Everaerts S, De Smidt E, Dooms C, Yserbyt J, Gysemans C, Gayan-Ramirez G, Mathieu C, Janssens W. 1,25-Dihydroxyvitamin D Modulates Antibacterial and Inflammatory Response in Human Cigarette Smoke-Exposed Macrophages. PLoS One 2016; 11:e0160482. [PMID: 27513734 PMCID: PMC4981391 DOI: 10.1371/journal.pone.0160482] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 07/20/2016] [Indexed: 12/26/2022] Open
Abstract
Cigarette smoking is associated with increased inflammation and defective antibacterial responses in the airways. Interestingly, vitamin D has been shown to suppress inflammation and to improve antibacterial defense. However, it is currently unknown whether vitamin D may modulate inflammation and antibacterial defects in human cigarette smoke (CS)-exposed airways. To explore these unresolved issues, alveolar macrophages obtained from non-smoking and smoking subjects as well as human cigarette smoke extract (CSE)-treated THP-1 macrophages were stimulated with 1,25-dihydroxyvitamin D (1,25(OH)2D) to address inflammatory and antibacterial responses. Although basal levels of inflammatory cytokines and chemokines did not differ between non-smoking and smoking subjects, 1,25(OH)2D did reduce levels of IL-6, TNF-α and MCP-1 in alveolar macrophages in response to LPS/IFN-γ, although not statistically significant for TNF-α and IL-6 in smokers. CSE did not significantly alter vitamin D metabolism (expression levels of CYP24A1 or CYP27B1) in THP-1 macrophages. Furthermore, stimulation with 1,25(OH)2D reduced mRNA expression levels and/or protein levels of IL-8, TNF-α and MCP-1 in CSE-treated THP-1 macrophages. 1,25(OH)2D did not improve defects in phagocytosis of E. coli bacteria or the oxidative burst response in CSE-treated THP-1 macrophages or alveolar macrophages from smokers. However, 1,25(OH)2D significantly enhanced mRNA expression and/or protein levels of the antimicrobial peptide cathelicidin in alveolar macrophages and THP-1 macrophages, independently of CS exposure. In conclusion, our results provide the first evidence that vitamin D could be a new strategy for attenuating airway inflammation and improving antibacterial defense in CS-exposed airways.
Collapse
Affiliation(s)
- Nele Heulens
- Laboratory of Respiratory Diseases, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Hannelie Korf
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Carolien Mathyssen
- Laboratory of Respiratory Diseases, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Stephanie Everaerts
- Laboratory of Respiratory Diseases, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Elien De Smidt
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Christophe Dooms
- Laboratory of Respiratory Diseases, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jonas Yserbyt
- Laboratory of Respiratory Diseases, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Conny Gysemans
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Ghislaine Gayan-Ramirez
- Laboratory of Respiratory Diseases, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Chantal Mathieu
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Wim Janssens
- Laboratory of Respiratory Diseases, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
- * E-mail:
| |
Collapse
|
21
|
Low-Dose Oxygen Enhances Macrophage-Derived Bacterial Clearance following Cigarette Smoke Exposure. J Immunol Res 2016; 2016:1280347. [PMID: 27403445 PMCID: PMC4923598 DOI: 10.1155/2016/1280347] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/04/2016] [Accepted: 05/17/2016] [Indexed: 11/22/2022] Open
Abstract
Background. Chronic obstructive pulmonary disease (COPD) is a common, smoking-related lung disease. Patients with COPD frequently suffer disease exacerbations induced by bacterial respiratory infections, suggestive of impaired innate immunity. Low-dose oxygen is a mainstay of therapy during COPD exacerbations; yet we understand little about whether oxygen can modulate the effects of cigarette smoke on lung immunity. Methods. Wild-type mice were exposed to cigarette smoke for 5 weeks, followed by intratracheal instillation of Pseudomonas aeruginosa (PAO1) and 21% or 35–40% oxygen. After two days, lungs were harvested for PAO1 CFUs, and bronchoalveolar fluid was sampled for inflammatory markers. In culture, macrophages were exposed to cigarette smoke and oxygen (40%) for 24 hours and then incubated with PAO1, followed by quantification of bacterial phagocytosis and inflammatory markers. Results. Mice exposed to 35–40% oxygen after cigarette smoke and PAO1 had improved survival and reduced lung CFUs and inflammation. Macrophages from these mice expressed less TNF-α and more scavenger receptors. In culture, macrophages exposed to cigarette smoke and oxygen also demonstrated decreased TNF-α secretion and enhanced phagocytosis of PAO1 bacteria. Conclusions. Our findings demonstrate a novel, protective role for low-dose oxygen following cigarette smoke and bacteria exposure that may be mediated by enhanced macrophage phagocytosis.
Collapse
|
22
|
Neonatal pneumococcal colonisation caused by Influenza A infection alters lung function in adult mice. Sci Rep 2016; 6:22751. [PMID: 26940954 PMCID: PMC4778168 DOI: 10.1038/srep22751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/19/2016] [Indexed: 02/08/2023] Open
Abstract
There is emerging epidemiological data to suggest that upper respiratory tract bacterial colonisation in infancy may increase the risk of developing respiratory dysfunction later in life, and respiratory viruses are known to precipitate persistent colonisation. This study utilized a neonatal mouse model of Streptococcus pneumonia (SP) and influenza A virus (IAV) co-infection, where bronchoalveolar leukocyte infiltration had resolved by adulthood. Only co-infection resulted in persistent nasopharyngeal colonisation over 40 days and a significant increase in airway resistance in response to in vivo methacholine challenge. A significant increase in hysteresivity was also observed in IAV and co-infected mice, consistent with ventilatory heterogeneity and structural changes in the adult lung. Airway hyper-responsiveness was not associated with a detectable increase in goblet cell transdifferentiation, peribronchial smooth muscle bulk or collagen deposition in regions surrounding the airways. Increased reactivity was not observed in precision cut lung slices challenged with methacholine in vitro. Histologically, the airway epithelium appeared normal and expression of epithelial integrity markers (ZO-1, occludin-1 and E-cadherin) were not altered. In summary, neonatal co-infection led to persistent nasopharyngeal colonisation and increased airway responsiveness that was not associated with detectable smooth muscle or mucosal epithelial abnormalities, however increased hysteresivity may reflect ventilation heterogeneity.
Collapse
|
23
|
Bozinovski S, Vlahos R, Anthony D, McQualter J, Anderson G, Irving L, Steinfort D. COPD and squamous cell lung cancer: aberrant inflammation and immunity is the common link. Br J Pharmacol 2016; 173:635-48. [PMID: 26013585 PMCID: PMC4742298 DOI: 10.1111/bph.13198] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 04/30/2015] [Accepted: 05/14/2015] [Indexed: 12/25/2022] Open
Abstract
Cigarette smoking has reached epidemic proportions within many regions of the world and remains the highest risk factor for chronic obstructive pulmonary disease (COPD) and lung cancer. Squamous cell lung cancer is commonly detected in heavy smokers, where the risk of developing lung cancer is not solely defined by tobacco consumption. Although therapies that target common driver mutations in adenocarcinomas are showing some promise, they are proving ineffective in smoking-related squamous cell lung cancer. Since COPD is characterized by an excessive inflammatory and oxidative stress response, this review details how aberrant innate, adaptive and systemic inflammatory processes can contribute to lung cancer susceptibility in COPD. Activated leukocytes release increasing levels of proteases and free radicals as COPD progresses and tertiary lymphoid aggregates accumulate with increasing severity. Reactive oxygen species promote formation of reactive carbonyls that are not only tumourigenic through initiating DNA damage, but can directly alter the function of regulatory proteins involved in host immunity and tumour suppressor functions. Systemic inflammation is also markedly increased during infective exacerbations in COPD and the interplay between tumour-promoting serum amyloid A (SAA) and IL-17A is discussed. SAA is also an endogenous allosteric modifier of FPR2 expressed on immune and epithelial cells, and the therapeutic potential of targeting this receptor is proposed as a novel strategy for COPD-lung cancer overlap.
Collapse
Affiliation(s)
- Steven Bozinovski
- School of Health Sciences and Health Innovations Research Institute, RMIT University, Melbourne, Vic., Australia
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, Vic., Australia
| | - Ross Vlahos
- School of Health Sciences and Health Innovations Research Institute, RMIT University, Melbourne, Vic., Australia
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, Vic., Australia
| | - Desiree Anthony
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, Vic., Australia
| | - Jonathan McQualter
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, Vic., Australia
| | - Gary Anderson
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, Vic., Australia
| | - Louis Irving
- Department of Respiratory Medicine, The Royal Melbourne Hospital, Parkville, Vic., Australia
| | - Daniel Steinfort
- Department of Respiratory Medicine, The Royal Melbourne Hospital, Parkville, Vic., Australia
| |
Collapse
|
24
|
Bozinovski S, Vlahos R. Multifaceted Role for IL-17A in the Pathogenesis of Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2015; 191:1213-4. [DOI: 10.1164/rccm.201504-0714ed] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
25
|
O'Leary SM, Coleman MM, Chew WM, Morrow C, McLaughlin AM, Gleeson LE, O'Sullivan MP, Keane J. Cigarette smoking impairs human pulmonary immunity to Mycobacterium tuberculosis. Am J Respir Crit Care Med 2015; 190:1430-6. [PMID: 25390734 DOI: 10.1164/rccm.201407-1385oc] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Cigarette smoking is linked to important aspects of tuberculosis, such as susceptibility to infection, disease reactivation, mortality, transmission, and persistent infectiousness. The mechanistic basis for this remains poorly understood. OBJECTIVES To compare the functional impairment seen in human alveolar macrophages (AM) from nonsmokers, smokers, and ex-smokers after infection with Mycobacterium tuberculosis (Mtb). METHODS AM were acquired at bronchoscopy, and number and viability from smoking donors were compared with nonsmoking donors. AM were challenged in vitro with Mtb and intracellular bacterial viability was measured. Cytokine secretion was measured 24 hours postinfection by ELISA. Previously we determined the frequency of CD4(+)FoxP3(+) T cells in the presence or absence of allogeneic AM, and data were reanalyzed to separate the patient subjects according to smoking status. MEASUREMENTS AND MAIN RESULTS There were significantly more AM from smokers compared with nonsmokers or ex-smokers (P < 0.01). AM from smokers could not control intracellular Mtb growth. Nonsmokers' AM generated significantly more tumor necrosis factor (TNF)-α, IFN-γ, and IL-1β after Mtb infection compared with uninfected AM (P < 0.05). However, Mtb-infected AM from smokers did not secrete significantly more TNF-α, IFN-γ, and IL-1β compared with uninfected smokers' AM. AM taken from ex-smokers also failed to secrete significantly increased TNF-α, IFN-γ, and IL-1β after Mtb infection. Both smokers' and nonsmokers' AM induced FoxP3(+) T regulatory cell phenotype responses in allogeneic admixed T cells (>4.8 fold; P < 0.05). Even after Mtb infection, AM continued to drive this regulatory phenotype. CONCLUSIONS In smokers, the pulmonary compartment has a number of macrophage-specific immune impairments that provide some mechanistic explanations whereby cigarette smoking renders a patient susceptible to tuberculosis infection and disease.
Collapse
Affiliation(s)
- Seónadh M O'Leary
- 1 Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland; and
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Bozinovski S, Anthony D, Vlahos R. Targeting pro-resolution pathways to combat chronic inflammation in COPD. J Thorac Dis 2014; 6:1548-56. [PMID: 25478196 DOI: 10.3978/j.issn.2072-1439.2014.08.08] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 07/18/2014] [Indexed: 12/31/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is an inflammatory lung condition that is associated with irreversible airflow obstruction as a consequence of small airways disease, excessive mucus production and emphysema. Paradoxically, excessive inflammation fails to control microbial pathogens that not only colonise COPD airways, but also trigger acute exacerbations, which markedly increase inflammation underlying host tissue damage. Excessive production of leukocyte mobilising cytokines such as CXCL8 (IL-8) and leukotriene B4 (LTB4) in response to environmental stimuli (cigarette smoke and microbial products) are thought to maintain chronic inflammation, in conjunction with inefficient macrophage clearance of microbes and apoptotic neutrophils. In this perspective, we discuss an alternative view on why inflammation persists with a focus on why pro-resolution mediators such as lipoxin A4 (LXA4), D-series resolving and Annexin A1 fail to effectively switch off inflammation in COPD. These pro-resolving mediators converge on the G-protein coupled receptor, ALX/FPR2. This receptor is particularly relevant to COPD as the complex milieu of exogenous and host-derived mediators within the inflamed airways include agonists that potently activate ALX/FPR2, including Serum Amyloid A (SAA) and the cathelicidin, LL-37. There is emerging evidence to suggest that ALX/FPR2 can exist in alternative receptor conformations in an agonist-biased manner, which facilitates alternate functional receptor behaviors. Hence, the development of more stable pro-resolving analogs provides therapeutic opportunities to address ALX/FPR2 conformations to counteract pathogenic signaling and promote non-phlogistic clearance pathways essential for resolution of inflammation.
Collapse
Affiliation(s)
- Steven Bozinovski
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville 3010, Australia
| | - Desiree Anthony
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville 3010, Australia
| | - Ross Vlahos
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville 3010, Australia
| |
Collapse
|
27
|
Slattery ML, Lundgreen A, Torres-Mejia G, Wolff RK, Hines L, Baumgartner K, John EM. Diet and lifestyle factors modify immune/inflammation response genes to alter breast cancer risk and prognosis: the Breast Cancer Health Disparities Study. Mutat Res 2014; 770:19-28. [PMID: 25332681 PMCID: PMC4201121 DOI: 10.1016/j.mrfmmm.2014.08.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Tumor necrosis factor-α (TNF) and toll-like receptors (TLR) are important mediators of inflammation. We examined 10 of these genes with respect to breast cancer risk and mortality in a genetically admixed population of Hispanic/Native American (NA) (2111 cases, 2597 controls) and non-Hispanic white (NHW) (1481 cases, 1585 controls) women. Additionally, we explored if diet and lifestyle factors modified associations with these genes. Overall, these genes (collectively) were associated with breast cancer risk among women with >70% NA ancestry (P(ARTP) = 0.0008), with TLR1 rs7696175 being the primary risk contributor (OR 1.77, 95% CI 1.25, 2.51). Overall, TLR1 rs7696175 (HR 1.40, 95% CI 1.03, 1.91; P(adj) = 0.032), TLR4 rs5030728 (HR 1.96, 95% CI 1.30, 2.95; P(adj) = 0.014), and TNFRSF1A rs4149578 (HR 2.71, 95% CI 1.28, 5.76; P(adj) = 0.029) were associated with increased breast cancer mortality. We observed several statistically significant interactions after adjustment for multiple comparisons, including interactions between our dietary oxidative balance score and CD40LG and TNFSF1A; between cigarette smoking and TLR1, TLR4, and TNF; between body mass index (BMI) among pre-menopausal women and TRAF2; and between regular use of aspirin/non-steroidal anti-inflammatory drugs and TLR3 and TRA2. In conclusion, our findings support a contributing role of certain TNF-α and TLR genes in both breast cancer risk and survival, particularly among women with higher NA ancestry. Diet and lifestyle factors appear to be important mediators of the breast cancer risk associated with these genes.
Collapse
Affiliation(s)
- Martha L. Slattery
- University of Utah, Department of Medicine, 383 Colorow, Salt Lake City, UT 84108. 801-585-6955
| | - Abbie Lundgreen
- University of Utah, Department of Medicine, 383 Colorow, Salt Lake City, UT 84108. 801-585-6955
| | - Gabriela Torres-Mejia
- Instituto Nacional de Salud Pública, Centro de Investigación en Salud Poblacional, Av. Universidad No. 655, Col. Sta. Ma. Ahuacatitlán, Cuernavaca Morelos CP 62100, México
| | - Roger K. Wolff
- University of Utah, Department of Medicine, 383 Colorow, Salt Lake City, UT 84108. 801-585-6955
| | - Lisa Hines
- University of Colorado at Colorado Springs, Department of Biology, Colorado Springs, CO 80918
| | - Kathy Baumgartner
- Department of Epidemiology and Population Health, School of Public Health & Information Sciences, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40292
| | - Esther M. John
- Cancer Prevention Institute of California, Fremont, CA 94538, and Division of Epidemiology, Department of Health Research and Policy and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
28
|
Hansen MJ, Chan SPJ, Langenbach SY, Dousha LF, Jones JE, Yatmaz S, Seow HJ, Vlahos R, Anderson GP, Bozinovski S. IL-17A and serum amyloid A are elevated in a cigarette smoke cessation model associated with the persistence of pigmented macrophages, neutrophils and activated NK cells. PLoS One 2014; 9:e113180. [PMID: 25405776 PMCID: PMC4236152 DOI: 10.1371/journal.pone.0113180] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/20/2014] [Indexed: 12/29/2022] Open
Abstract
While global success in cessation advocacy has seen smoking rates fall in many developed countries, persistent lung inflammation in ex-smokers is an increasingly important clinical problem whose mechanistic basis remains poorly understood. In this study, candidate effector mechanisms were assessed in mice exposed to cigarette smoke (CS) for 4 months following cessation from long term CS exposure. BALF neutrophils, CD4+ and CD8+ T cells and lung innate NK cells remained significantly elevated following smoking cessation. Analysis of neutrophil mobilization markers showed a transition from acute mediators (MIP-2α, KC and G-CSF) to sustained drivers of neutrophil and macrophage recruitment and activation (IL-17A and Serum Amyoid A (SAA)). Follicle-like lymphoid aggregates formed with CS exposure and persisted with cessation, where they were in close anatomical proximity to pigmented macrophages, whose number actually increased 3-fold following CS cessation. This was associated with the elastolytic protease, MMP-12 (macrophage metallo-elastase) which remained significantly elevated post-cessation. Both GM-CSF and CSF-1 were significantly increased in the CS cessation group relative to the control group. In conclusion, we show that smoking cessation mediates a transition to accumulation of pigmented macrophages, which may contribute to the expanded macrophage population observed in COPD. These macrophages together with IL-17A, SAA and innate NK cells are identified here as candidate persistence determinants and, we suggest, may represent specific targets for therapies directed towards the amelioration of chronic airway inflammation.
Collapse
Affiliation(s)
- Michelle J. Hansen
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
- * E-mail:
| | - Sheau Pyng J. Chan
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Shenna Y. Langenbach
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Lovisa F. Dousha
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Jessica E. Jones
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Selcuk Yatmaz
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Huei Jiunn Seow
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Ross Vlahos
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Gary P. Anderson
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Steven Bozinovski
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| |
Collapse
|
29
|
Zhu L, Wang Y, Jie G, Chi Q, Zhou J, Cui B, Piao D, Zhao Y. Association between Toll-like receptor 4 and interleukin 17 gene polymorphisms and colorectal cancer susceptibility in Northeast China. Med Oncol 2014; 31:73. [PMID: 25216863 DOI: 10.1007/s12032-014-0073-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 06/10/2014] [Indexed: 12/15/2022]
Abstract
Limited studies about the effects of TLR 4 and IL17 polymorphisms (SNPs) on the risk of colorectal cancer (CRC) have yielded inconsistent results. Totally, 601 CRC patients and 627 controls were enrolled. Unconditional logistic regression was used to estimate the association between tagSNPs and susceptibility of CRC and the interactions effects of gene and environment on the risk of CRC. IL17 rs6973569 AG and AG/AA genotypes significantly decreased the risk of CRC compared with GG genotype (ORadjusted=0.72, 95% CI 0.55-0.94 and ORadjusted=0.74, 95 % CI 0.57-0.97). The haplotype G-T-G-C-A-G accounting for the largest proportion haplotypes increased the risk of CRC (OR=1.27, 95 % CI 1.06-1.53). However, G-C-C-T-A-G and G-C-G-C-A-G haplotypes decreased the susceptibility of CRC. Synergistic interactions between TLR 4 rs1927911 CT/TT and higher pungent food intake as well as IL17 rs6973569 AG/AA genotypes and higher intake of sausage food (ORi=1.72, 95% CI 1.04-2.84 and ORi=3.38, 95% CI 1.28-8.91) on the risk of CRC were observed. IL17 rs6973569 SNP might be an independent factor of susceptibility to CRC. TLR 4 haplotype of G-T-G-C-A-G may increase risk of CRC. Higher intake of pungent and sausage food synergistically interacted with TLR 4 and IL17 SNPs on the risk of CRC.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Epidemiology, Public Health College, Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, Heilongjiang, People's Republic of China,
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Vlahos R, Bozinovski S. Role of alveolar macrophages in chronic obstructive pulmonary disease. Front Immunol 2014; 5:435. [PMID: 25309536 PMCID: PMC4160089 DOI: 10.3389/fimmu.2014.00435] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 08/26/2014] [Indexed: 11/13/2022] Open
Abstract
Alveolar macrophages (AMs) represent a unique leukocyte population that responds to airborne irritants and microbes. This distinct microenvironment coordinates the maturation of long-lived AMs, which originate from fetal blood monocytes and self-renew through mechanisms dependent on GM-CSF and CSF-1 signaling. Peripheral blood monocytes can also replenish lung macrophages; however, this appears to occur in a stimuli specific manner. In addition to mounting an appropriate immune response during infection and injury, AMs actively coordinate the resolution of inflammation through efferocytosis of apoptotic cells. Any perturbation of this process can lead to deleterious responses. In chronic obstructive pulmonary disease (COPD), there is an accumulation of airway macrophages that do not conform to the classic M1/M2 dichotomy. There is also a skewed transcriptome profile that favors expression of wound-healing M2 markers, which is reflective of a deficiency to resolve inflammation. Endogenous mediators that can promote an imbalance in inhibitory M1 vs. healing M2 macrophages are discussed, as they are the plausible mechanisms underlying why AMs fail to effectively resolve inflammation and restore normal lung homeostasis in COPD.
Collapse
Affiliation(s)
- Ross Vlahos
- Department of Pharmacology and Therapeutics, Lung Health Research Centre, The University of Melbourne , Parkville, VIC , Australia
| | - Steven Bozinovski
- Department of Pharmacology and Therapeutics, Lung Health Research Centre, The University of Melbourne , Parkville, VIC , Australia
| |
Collapse
|
31
|
Metcalfe HJ, Lea S, Hughes D, Khalaf R, Abbott-Banner K, Singh D. Effects of cigarette smoke on Toll-like receptor (TLR) activation of chronic obstructive pulmonary disease (COPD) macrophages. Clin Exp Immunol 2014; 176:461-72. [PMID: 24528166 DOI: 10.1111/cei.12289] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2014] [Indexed: 01/08/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by an abnormal innate immune response. We have investigated the changes in the innate immune response of COPD alveolar macrophages exposed to both cigarette smoke and Toll-like receptor (TLR) stimulation. COPD and control alveolar macrophages were exposed to cigarette smoke extract (CSE) followed by TLR-2, -4 and -5 ligands [Pam3CSK4, lipopolysaccharide (LPS) and phase I flagellin (FliC), respectively] or non-typeable Haemophilus influenzae (NTHi). CSE exposure suppressed TLR-induced tumour necrosis factor (TNF)-α, interleukin (IL)-6, IL-10 and regulated on activation, normal T cell expressed and secreted (RANTES) production in both COPD and control alveolar macrophages, but had no effect on interleukin 8 (CXCL8) production. Similarly, CSE suppressed NTHi-induced TNF-α but not NTHi-induced CXCL8 production in COPD alveolar macrophages. Gene expression analysis showed that CSE suppressed LPS-induced TNF-α transcription but not CXCL8 transcription in COPD alveolar macrophages. The dampening effect of CSE on LPS-induced cytokine production was associated with a reduction in p38, extracellular signal regulated kinase (ERK) and p65 activation. In conclusion, CSE caused a reduced innate immune response in COPD alveolar macrophages, with the exception of persistent CXCL8 production. This could be a mechanism by which alveolar macrophages promote neutrophil chemotaxis under conditions of oxidative stress and bacterial exposure.
Collapse
Affiliation(s)
- H J Metcalfe
- The University of Manchester, Manchester Academic Health Science Centre, University Hospital South Manchester NHS Foundation Trust, NIHR South Manchester Respiratory and Allergy Clinical Research Facility, Manchester, UK
| | | | | | | | | | | |
Collapse
|
32
|
Anthony D, McQualter JL, Bishara M, Lim EX, Yatmaz S, Seow HJ, Hansen M, Thompson M, Hamilton JA, Irving LB, Levy BD, Vlahos R, Anderson GP, Bozinovski S. SAA drives proinflammatory heterotypic macrophage differentiation in the lung via CSF-1R-dependent signaling. FASEB J 2014; 28:3867-77. [PMID: 24846388 DOI: 10.1096/fj.14-250332] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 05/05/2014] [Indexed: 12/11/2022]
Abstract
Serum amyloid A (SAA) is expressed locally in chronic inflammatory conditions such as chronic obstructive pulmonary disease (COPD), where macrophages that do not accord with the classic M1/M2 paradigm also accumulate. In this study, the role of SAA in regulating macrophage differentiation was investigated in vitro using human blood monocytes from healthy subjects and patients with COPD and in vivo using an airway SAA challenge model in BALB/c mice. Differentiation of human monocytes with SAA stimulated the proinflammatory monokines IL-6 and IL-1β concurrently with the M2 markers CD163 and IL-10. Furthermore, SAA-differentiated macrophages stimulated with lipopolysaccharide (LPS) expressed markedly higher levels of IL-6 and IL-1β. The ALX/FPR2 antagonist WRW4 reduced IL-6 and IL-1β expression but did not significantly inhibit phagocytic and efferocytic activity. In vivo, SAA administration induced the development of a CD11c(high)CD11b(high) macrophage population that generated higher levels of IL-6, IL-1β, and G-CSF following ex vivo LPS challenge. Blocking CSF-1R signaling effectively reduced the number of CD11c(high)CD11b(high) macrophages by 71% and also markedly inhibited neutrophilic inflammation by 80%. In conclusion, our findings suggest that SAA can promote a distinct CD11c(high)CD11b(high) macrophage phenotype, and targeting this population may provide a novel approach to treating chronic inflammatory conditions associated with persistent SAA expression.
Collapse
Affiliation(s)
| | | | | | - Ee X Lim
- Department of Pharmacology and Therapeutics and
| | | | | | | | - Michelle Thompson
- Department of Respiratory Medicine, Royal Melbourne Hospital; Parkville, Victoria, Australia; and
| | - John A Hamilton
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Louis B Irving
- Department of Respiratory Medicine, Royal Melbourne Hospital; Parkville, Victoria, Australia; and
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ross Vlahos
- Department of Pharmacology and Therapeutics and
| | | | | |
Collapse
|
33
|
Colombo G, Clerici M, Giustarini D, Portinaro NM, Aldini G, Rossi R, Milzani A, Dalle-Donne I. Pathophysiology of tobacco smoke exposure: recent insights from comparative and redox proteomics. MASS SPECTROMETRY REVIEWS 2014; 33:183-218. [PMID: 24272816 DOI: 10.1002/mas.21392] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 05/23/2013] [Accepted: 05/23/2013] [Indexed: 06/02/2023]
Abstract
First-hand and second-hand tobacco smoke are causally linked to a huge number of deaths and are responsible for a broad spectrum of pathologies such as cancer, cardiovascular, respiratory, and eye diseases as well as adverse effects on female reproductive function. Cigarette smoke is a complex mixture of thousands of different chemical species, which exert their negative effects on macromolecules and biochemical pathways, both directly and indirectly. Many compounds can act as oxidants, pro-inflammatory agents, carcinogens, or a combination of these. The redox behavior of cigarette smoke has many implications for smoke related diseases. Reactive oxygen and nitrogen species (both radicals and non-radicals), reactive carbonyl compounds, and other species may induce oxidative damage in almost all the biological macromolecules, compromising their structure and/or function. Different quantitative and redox proteomic approaches have been applied in vitro and in vivo to evaluate, respectively, changes in protein expression and specific oxidative protein modifications induced by exposure to cigarette smoke and are overviewed in this review. Many gel-based and gel-free proteomic techniques have already been used successfully to obtain clues about smoke effects on different proteins in cell cultures, animal models, and humans. The further implementation with other sensitive screening techniques could be useful to integrate the comprehension of cigarette smoke effects on human health. In particular, the redox proteomic approach may also help identify biomarkers of exposure to tobacco smoke useful for preventing these effects or potentially predictive of the onset and/or progression of smoking-induced diseases as well as potential targets for therapeutic strategies.
Collapse
Affiliation(s)
- Graziano Colombo
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Smoking represents one of the most important preventable risk factors for the development of atherosclerosis. The present review aims at providing a comprehensive summary of published data from clinical and animal studies, as well as results of basic research on the proatherogenic effect of smoking. Extensive search and review of literature revealed a vast amount of data on the influence of cigarette smoke and its constituents on early atherogenesis, particularly on endothelial cells. Vascular dysfunction induced by smoking is initiated by reduced nitric oxide (NO) bioavailability and further by the increased expression of adhesion molecules and subsequent endothelial dysfunction. Smoking-induced increased adherence of platelets and macrophages provokes the development of a procoagulant and inflammatory environment. After transendothelial migration and activation, macrophages take up oxidized lipoproteins arising from oxidative modifications and transdifferentiate into foam cells. In addition to direct physical damage to endothelial cells, smoking induces tissue remodeling, and prothrombotic processes together with activation of systemic inflammatory signals, all of which contribute to atherogenic vessel wall changes. There are still great gaps in our knowledge about the effects of smoking on cardiovascular disease. However, we know that smoking cessation is the most effective measure for reversing damage that has already occurred and preventing fatal cardiovascular outcomes.
Collapse
Affiliation(s)
- Barbara Messner
- From the Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Vienna, Austria (B.M.); and Cardiac Surgery Research Laboratory Innsbruck, University Clinic for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria (D.B.)
| | - David Bernhard
- From the Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Vienna, Austria (B.M.); and Cardiac Surgery Research Laboratory Innsbruck, University Clinic for Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria (D.B.)
| |
Collapse
|
35
|
Vlahos R, Bozinovski S. Recent advances in pre-clinical mouse models of COPD. Clin Sci (Lond) 2014; 126:253-65. [PMID: 24144354 PMCID: PMC3878607 DOI: 10.1042/cs20130182] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 07/17/2013] [Accepted: 08/01/2013] [Indexed: 02/06/2023]
Abstract
COPD (chronic obstructive pulmonary disease) is a major incurable global health burden and will become the third largest cause of death in the world by 2020. It is currently believed that an exaggerated inflammatory response to inhaled irritants, in particular cigarette smoke, causes progressive airflow limitation. This inflammation, where macrophages, neutrophils and T-cells are prominent, leads to oxidative stress, emphysema, small airways fibrosis and mucus hypersecretion. The mechanisms and mediators that drive the induction and progression of chronic inflammation, emphysema and altered lung function are poorly understood. Current treatments have limited efficacy in inhibiting chronic inflammation, do not reverse the pathology of disease and fail to modify the factors that initiate and drive the long-term progression of disease. Therefore there is a clear need for new therapies that can prevent the induction and progression of COPD. Animal modelling systems that accurately reflect disease pathophysiology continue to be essential to the development of new therapies. The present review highlights some of the mouse models used to define the cellular, molecular and pathological consequences of cigarette smoke exposure and whether they can be used to predict the efficacy of new therapeutics for COPD.
Collapse
Key Words
- acute exacerbations of chronic obstructive pulmonary disease (aecopd)
- chronic obstructive pulmonary disease (copd)
- emphysema
- inflammation
- skeletal muscle wasting
- smoking
- aecopd, acute exacerbations of copd
- bal, bronchoalveolar lavage
- balf, bal fluid
- copd, chronic obstructive pulmonary disease
- gm-csf, granulocyte/macrophage colony-stimulating factor
- gold, global initiative on chronic obstructive lung disease
- gpx, glutathione peroxidase
- hdac, histone deacetylation
- il, interleukin
- ltb4, leukotriene b4
- mapk, mitogen-activated protein kinase
- mcp-1, monocyte chemotactic protein-1
- mmp, matrix metalloproteinase
- ne, neutrophil elastase
- nf-κb, nuclear factor κb
- nrf2, nuclear erythroid-related factor 2
- o2•−, superoxide radical
- onoo−, peroxynitrite
- pde, phosphodiesterase
- pi3k, phosphoinositide 3-kinase
- ros, reactive oxygen species
- rv, rhinovirus
- slpi, secretory leucocyte protease inhibitor
- sod, superoxide dismutase
- tgf-β, transforming growth factor-β
- timp, tissue inhibitor of metalloproteinases
- tnf-α, tumour necrosis factor-α
- v/q, ventilation/perfusion
Collapse
Affiliation(s)
- Ross Vlahos
- *Lung Health Research Centre, Department of Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Steven Bozinovski
- *Lung Health Research Centre, Department of Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
36
|
Yoshida T, Nagai K, Inomata T, Ito Y, Betsuyaku T, Nishimura M. Relationship between neutrophil influx and oxidative stress in alveolar space in lipopolysaccharide-induced lung injury. Respir Physiol Neurobiol 2014; 191:75-83. [DOI: 10.1016/j.resp.2013.11.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 11/18/2013] [Accepted: 11/22/2013] [Indexed: 10/25/2022]
|
37
|
Bozinovski S, Anthony D, Anderson GP, Irving LB, Levy BD, Vlahos R. Treating neutrophilic inflammation in COPD by targeting ALX/FPR2 resolution pathways. Pharmacol Ther 2013; 140:280-9. [PMID: 23880288 DOI: 10.1016/j.pharmthera.2013.07.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 07/09/2013] [Indexed: 12/19/2022]
Abstract
Neutrophilic inflammation persists in COPD despite best current therapies and it is particularly resistant to inhaled glucocorticosteroids. Persistent neutrophil activation not only contributes to matrix breakdown, but can maintain inflammation through the release of endogenous damage associated molecule patterns (DAMPs). Inhibiting excessive neutrophilic inflammation is challenging as many pathogen recognition receptors can initiate migration and the targeting of downstream signaling molecules may compromise essential host defense mechanisms. Here, we discuss new strategies to combat this inflammation in COPD by focusing on the anti-inflammatory role of ALX/FPR2 receptors. ALX/FPR2 is a promiscuous G-protein coupled receptor (GPCR) responding to lipid and peptide agonists that can either switch on acute inflammation or promote resolution of inflammation. We highlight this receptor as an emerging target in the pathogenesis of COPD because known ALX/FPR2 endogenous agonists are enriched in COPD. Serum Amyloid A (SAA) has recently been discovered to be abundantly expressed in COPD and is a potent ALX/FPR2 agonist that unlike almost all other inflammatory chemoattractants, is induced by glucocorticosteroids. SAA not only initiates lung inflammation via ALX/FPR2 but can allosterically modify this receptor so that it no longer transduces pro-resolving signals from endogenous lipoxins that would otherwise promote tissue healing. We propose that there is an imbalance in endogenous and microbial ALX/FPR2 receptor agonists in the inflamed COPD lung environment that oppose protective anti-inflammatory and pro-resolution pathways. These insights open the possibility of targeting ALX/FPR2 receptors using synthetic agonists to resolve persistent neutrophilic inflammation without compromising essential host defense mechanisms.
Collapse
Affiliation(s)
- Steven Bozinovski
- Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Feldman C, Anderson R. Cigarette smoking and mechanisms of susceptibility to infections of the respiratory tract and other organ systems. J Infect 2013; 67:169-84. [PMID: 23707875 DOI: 10.1016/j.jinf.2013.05.004] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 05/08/2013] [Accepted: 05/14/2013] [Indexed: 01/04/2023]
Abstract
The predisposition of cigarette smokers for development of oral and respiratory infections caused by microbial pathogens is well recognised, with those infected with the human immunodeficiency virus (HIV) at particularly high risk. Smoking cigarettes has a suppressive effect on the protective functions of airway epithelium, alveolar macrophages, dendritic cells, natural killer (NK) cells and adaptive immune mechanisms, in the setting of chronic systemic activation of neutrophils. Cigarette smoke also has a direct effect on microbial pathogens to promote the likelihood of infective disease, specifically promotion of microbial virulence and antibiotic resistance. In addition to interactions between smoking and HIV infection, a number of specific infections/clinical syndromes have been associated epidemiologically with cigarette smoking, including those of the upper and lower respiratory tract, gastrointestinal tract, central nervous and other organ systems. Smoking cessation benefits patients in many ways, including reduction of the risk of infectious disease.
Collapse
Affiliation(s)
- Charles Feldman
- Division of Pulmonology, Department of Internal Medicine, Charlotte Maxeke Johannesburg Academic Hospital, South Africa.
| | | |
Collapse
|
40
|
Kuipers I, Bracke KR, Brusselle GG, Wouters EFM, Reynaert NL. Smoke decreases reversible oxidations S-glutathionylation and S-nitrosylation in mice. Free Radic Res 2012; 46:164-73. [PMID: 22145974 DOI: 10.3109/10715762.2011.647011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cigarette smoke causes irreversible oxidations in lungs, but its impact on reversible and physiologically relevant redox-dependent protein modifications remains to be investigated. Here the effect of cigarette smoke exposure in mice was investigated on the covalent binding of glutathione to protein thiols, known as S-glutathionylation (PSSG), which can be reversed by glutaredoxins (Grx). Also, protein S-nitrosylation (PSNO) which is the modification of protein thiols by NO and which is reversed by the enzyme alcohol dehydrogenase (ADH) 5 was examined. Both PSSG and PSNO levels in lung tissue were markedly decreased after 4 weeks of cigarette smoke exposure. This coincided with attenuated protein free thiol levels and increased protein carbonylation. The expression of NOX4, DHE sensitive oxidant production and iNOS levels were induced by smoke, whereas Grx1 mRNA expression and activity were attenuated. Free GSH levels, protein expression and activity of ADH5 were unaffected by smoke. Taken together, smoke exposure decreases reversible cysteine oxidations PSSG and PSNO and enhances protein carbonylation. These alterations are not associated with differences in some of the regulatory enzymes, but are likely the result of oxidative stress.
Collapse
Affiliation(s)
- Ine Kuipers
- Department of Respiratory Medicine, Nutrim School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | | | | | | | | |
Collapse
|
41
|
Hristova M, Spiess PC, Kasahara DI, Randall MJ, Deng B, van der Vliet A. The tobacco smoke component, acrolein, suppresses innate macrophage responses by direct alkylation of c-Jun N-terminal kinase. Am J Respir Cell Mol Biol 2012; 46:23-33. [PMID: 21778411 PMCID: PMC3262655 DOI: 10.1165/rcmb.2011-0134oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 07/07/2011] [Indexed: 12/21/2022] Open
Abstract
The respiratory innate immune system is often compromised by tobacco smoke exposure, and previous studies have indicated that acrolein, a reactive electrophile in tobacco smoke, may contribute to the immunosuppressive effects of smoking. Exposure of mice to acrolein at concentrations similar to those in cigarette smoke (5 ppm, 4 h) significantly suppressed alveolar macrophage responses to bacterial LPS, indicated by reduced induction of nitric oxide synthase 2, TNF-α, and IL-12p40. Mechanistic studies with bone marrow-derived macrophages or MH-S macrophages demonstrated that acrolein (1-30 μM) attenuated these LPS-mediated innate responses in association with depletion of cellular glutathione, although glutathione depletion itself was not fully responsible for these immunosuppressive effects. Inhibitory actions of acrolein were most prominent after acute exposure (<2 h), indicating the involvement of direct and reversible interactions of acrolein with critical signaling pathways. Among the key signaling pathways involved in innate macrophage responses, acrolein marginally affected LPS-mediated activation of nuclear factor (NF)-κB, and significantly suppressed phosphorylation of c-Jun N-terminal kinase (JNK) and activation of c-Jun. Using biotin hydrazide labeling, NF-κB RelA and p50, as well as JNK2, a critical mediator of innate macrophage responses, were revealed as direct targets for alkylation by acrolein. Mass spectrometry analysis of acrolein-modified recombinant JNK2 indicated adduction to Cys(41) and Cys(177), putative important sites involved in mitogen-activated protein kinase (MAPK) kinase (MEK) binding and JNK2 phosphorylation. Our findings indicate that direct alkylation of JNK2 by electrophiles, such as acrolein, may be a prominent and hitherto unrecognized mechanism in their immunosuppressive effects, and may be a major factor in smoking-induced effects on the immune system.
Collapse
Affiliation(s)
| | | | | | | | - Bin Deng
- Department of Biology and Proteomics Core Facility, University of Vermont, Burlington, Vermont
| | | |
Collapse
|
42
|
Slattery ML, Herrick JS, Bondurant KL, Wolff RK. Toll-like receptor genes and their association with colon and rectal cancer development and prognosis. Int J Cancer 2011; 130:2974-80. [PMID: 21792899 DOI: 10.1002/ijc.26314] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 07/01/2011] [Indexed: 01/11/2023]
Abstract
Toll-like receptors (TLRs) are mediators of inflammation in the gut and potentially important modulators of colon and rectal cancer risk. We use data from a population-based study of incident colon cancer cases (n = 1,555) and controls (n = 1,956) and rectal cancer cases (n = 754) and controls (n = 959). We evaluate genetic variation in TLR2 (six SNPs), TLR3 (four SNPs) and TLR4 (eight SNPs) with risk of developing colon or rectal cancer and survival after diagnosis. TLR3 rs11721827 was associated with rectal cancer (odds ratio [OR] 1.27, 95% confidence interval [CI] 1.02, 1.58 for AC/CC vs. AA genotype, Wald p = 0.035; adjusted p = 0.126); TLR3 rs3775292 and TLR4 rs11536898 were associated with colon cancer (OR 0.68, 95% CI 0.49, 0.95 for GG vs. CC/CG and OR 0.50. 95% CI 0.29, 0.87 for AA vs. CA/CC, respectively; Wald p = 0.023 and 0.015; adjusted p = 0.085 and 0.101, respectively). TLR2 rs7656411 and rs3804099, respectively, interacted with nonsteroidal anti-inflammatory drug (NSAID) use and cigarette smoking to alter risk of colon cancer (adjusted p = 0.034 and 0.077); TLR3 rs11721827 interacted with NSAID use to alter risk of colon cancer (adjusted p = 0.071). TLR3 rs3775292 interacted with dietary carbohydrates to alter colon cancer risk and with dietary carbohydrates and saturated fat to alter rectal cancer risk (adjusted p = 0.064, 0.0035 and 0.025, respectively). Multiple SNPs in TLR2 and TLR4 were associated with colon cancer survival. Although few independent associations with TLR genes were observed, we observed significant interaction of TLR2 and TLR3 with hypothesized lifestyle factors. Interaction with dietary factors remained significant for rectal cancer after adjustment for multiple comparisons.
Collapse
Affiliation(s)
- Martha L Slattery
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, UT 84108, USA.
| | | | | | | |
Collapse
|