1
|
Rosen AL, Lint MA, Voelker DH, Gilbert NM, Tomera CP, Santiago-Borges J, Wallace MA, Hannan TJ, Burnham CAD, Hultgren SJ, Kau AL. Secretory leukocyte protease inhibitor protects against severe urinary tract infection in mice. mBio 2024; 15:e0255423. [PMID: 38270443 PMCID: PMC10865866 DOI: 10.1128/mbio.02554-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/15/2023] [Indexed: 01/26/2024] Open
Abstract
Millions suffer from urinary tract infections (UTIs) worldwide every year with women accounting for the majority of cases. Uropathogenic Escherichia coli (UPEC) causes most of these primary infections and leads to 25% becoming recurrent or chronic. To repel invading pathogens, the urinary tract mounts a vigorous innate immune response that includes the secretion of antimicrobial peptides (AMPs), rapid recruitment of phagocytes, and exfoliation of superficial umbrella cells. Here, we investigate secretory leukocyte protease inhibitor (SLPI), an AMP with antiprotease, antimicrobial, and immunomodulatory functions, known to play protective roles at other mucosal sites, but not well characterized in UTIs. Using a preclinical model of UPEC-caused UTI, we show that urine SLPI increases in infected mice and that SLPI is localized to bladder epithelial cells. UPEC-infected SLPI-deficient (Slpi-/-) mice suffer from higher urine bacterial burdens, prolonged bladder inflammation, and elevated urine neutrophil elastase (NE) levels compared to wild-type (Slpi+/+) controls. Combined with bulk bladder RNA sequencing, our data indicate that Slpi-/- mice have a dysregulated immune and tissue repair response following UTI. We also measure SLPI in urine samples from a small group of female subjects 18-49 years old and find that SLPI tends to be higher in the presence of a uropathogen, except in patients with a history of recent or recurrent UTI, suggesting a dysregulation of SLPI expression in these women. Taken together, our findings show SLPI promotes clearance of UPEC in mice and provides preliminary evidence that SLPI is likewise regulated in response to uropathogen exposure in women.IMPORTANCEAnnually, millions of people suffer from urinary tract infections (UTIs) and more than $3 billion are spent on work absences and treatment of these patients. While the early response to UTI is known to be important in combating urinary pathogens, knowledge of host factors that help curb infection is still limited. Here, we use a preclinical model of UTI to study secretory leukocyte protease inhibitor (SLPI), an antimicrobial protein, to determine how it protects the bladder against infection. We find that SLPI is increased during UTI, accelerates the clearance of bacteriuria, and upregulates genes and pathways needed to fight an infection while preventing prolonged bladder inflammation. In a small clinical study, we show SLPI is readily detectable in human urine and is associated with the presence of a uropathogen in patients without a previous history of UTI, suggesting SLPI may play an important role in protecting from bacterial cystitis.
Collapse
Affiliation(s)
- Anne L. Rosen
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael A. Lint
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dayne H. Voelker
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicole M. Gilbert
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Christopher P. Tomera
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jesús Santiago-Borges
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Meghan A. Wallace
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Thomas J. Hannan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Carey-Ann D. Burnham
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Scott J. Hultgren
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrew L. Kau
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
2
|
Abrami M, Biasin A, Tescione F, Tierno D, Dapas B, Carbone A, Grassi G, Conese M, Di Gioia S, Larobina D, Grassi M. Mucus Structure, Viscoelastic Properties, and Composition in Chronic Respiratory Diseases. Int J Mol Sci 2024; 25:1933. [PMID: 38339210 PMCID: PMC10856136 DOI: 10.3390/ijms25031933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
The respiratory mucus, a viscoelastic gel, effectuates a primary line of the airway defense when operated by the mucociliary clearance. In chronic respiratory diseases (CRDs), such as asthma, chronic obstructive pulmonary disease (COPD), and cystic fibrosis (CF), the mucus is overproduced and its solid content augments, changing its structure and viscoelastic properties and determining a derangement of essential defense mechanisms against opportunistic microbial (virus and bacteria) pathogens. This ensues in damaging of the airways, leading to a vicious cycle of obstruction and infection responsible for the harsh clinical evolution of these CRDs. Here, we review the essential features of normal and pathological mucus (i.e., sputum in CF, COPD, and asthma), i.e., mucin content, structure (mesh size), micro/macro-rheology, pH, and osmotic pressure, ending with the awareness that sputum biomarkers (mucins, inflammatory proteins and peptides, and metabolites) might serve to indicate acute exacerbation and response to therapies. There are some indications that old and novel treatments may change the structure, viscoelastic properties, and biomarker content of sputum; however, a wealth of work is still needed to embrace these measures as correlates of disease severity in association with (or even as substitutes of) pulmonary functional tests.
Collapse
Affiliation(s)
- Michela Abrami
- Department of Engineering and Architecture, University of Trieste, Via Valerio 6/A, I-34127 Trieste, Italy; (M.A.); (A.B.); (M.G.)
| | - Alice Biasin
- Department of Engineering and Architecture, University of Trieste, Via Valerio 6/A, I-34127 Trieste, Italy; (M.A.); (A.B.); (M.G.)
| | - Fabiana Tescione
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, P.le E. Fermi 1, I-80055 Portici, Italy; (F.T.); (D.L.)
| | - Domenico Tierno
- Clinical Department of Medical, Surgical and Health Sciences, Cattinara University Hospital, University of Trieste, Strada di Fiume 447, I-34149 Trieste, Italy; (D.T.); (G.G.)
| | - Barbara Dapas
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgieri 1, I-34127 Trieste, Italy;
| | - Annalucia Carbone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 121, I-71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Gabriele Grassi
- Clinical Department of Medical, Surgical and Health Sciences, Cattinara University Hospital, University of Trieste, Strada di Fiume 447, I-34149 Trieste, Italy; (D.T.); (G.G.)
| | - Massimo Conese
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 121, I-71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Sante Di Gioia
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 121, I-71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Domenico Larobina
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, P.le E. Fermi 1, I-80055 Portici, Italy; (F.T.); (D.L.)
| | - Mario Grassi
- Department of Engineering and Architecture, University of Trieste, Via Valerio 6/A, I-34127 Trieste, Italy; (M.A.); (A.B.); (M.G.)
| |
Collapse
|
3
|
Abstract
New methods and technologies within the field of lung biology are beginning to shed new light into the microbial world of the respiratory tract. Long considered to be a sterile environment, it is now clear that the human lungs are frequently exposed to live microbes and their by-products. The nature of the lung microbiome is quite distinct from other microbial communities inhabiting our bodies such as those in the gut. Notably, the microbiome of the lung exhibits a low biomass and is dominated by dynamic fluxes of microbial immigration and clearance, resulting in a bacterial burden and microbiome composition that is fluid in nature rather than fixed. As our understanding of the microbial ecology of the lung improves, it is becoming increasingly apparent that certain disease states can disrupt the microbial-host interface and ultimately affect disease pathogenesis. In this Review, we provide an overview of lower airway microbial dynamics in health and disease and discuss future work that is required to uncover novel therapeutic targets to improve lung health.
Collapse
|
4
|
Lin Z, Chen P, Yuan Z, Yang L, Miao L, Wang H, Xu D. Fine particulate matter, airway inflammation, stress response, non-specific immune function and buccal microbial diversity in young adults. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 308:119692. [PMID: 35772617 DOI: 10.1016/j.envpol.2022.119692] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/26/2022] [Accepted: 06/25/2022] [Indexed: 06/15/2023]
Abstract
Fine particulate matter (PM2.5) has been associated with risk of oral and respiratory diseases. However, the biological mechanisms of adverse oral and respiratory health response to PM2.5 fluctuation have not been well characterized. This study aims to explore the relationships of PM2.5 with airway inflammation, salivary biomarkers and buccal mucosa microbiota. We performed a panel study among 40 college students involving 4 follow-ups from August to October 2021 in Hefei, Anhui Province, China. Health outcomes included fractional exhaled nitric oxide (FeNO), salivary biomarkers [C-reactive protein (CRP), cortisol, lysozyme and alpha-amylase] and buccal mucosa microbial diversity. Linear mixed-effect models were applied to explore the cumulative impacts of PM2.5 on health indicators. PM2.5 was positively correlated with FeNO, CRP, cortisol and alpha-amylase, while negatively with lysozyme. Per 10-μg/m3 increase in PM2.5 was linked to maximum increments in FeNO of 10.71% (95%CI: 2.01%, 19.41%) at lag 0-24 h, in CRP of 7.10% (95%CI: 5.39%, 8.81%) at lag 0-24 h, in cortisol of 1.25% (95%CI: 0.44%, 2.07%) at lag 0-48 h, and in alpha-amylase of 2.12% (95%CI: 0.53%, 3.71%) at lag 0-24 h, while associated with maximum decrement in lysozyme of 0.53% (95%CI: 0.12%, 0.95%) at lag 0-72 h. Increased PM2.5 was linked to reduction in the richness and evenness of buccal microbe and o_Bacillales and o_Bacteroidales were identified as differential microbes after PM2.5 inhalation. Bio-information analysis indicated that immunity system pathway was the most important enriched abundant process altered by PM2.5 exposure. In summary, short-term PM2.5 exposure may impair oral and respiratory health by inducing inflammatory and stress responses, weakening immune function and altering buccal mucosa microbial diversity.
Collapse
Affiliation(s)
- Zhijing Lin
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China.
| | - Ping Chen
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Zhi Yuan
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Liyan Yang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Lin Miao
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - Dexiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
5
|
Malvisi L, Yarraguntla A, Mortier MC, Osman K, Cleary DW, Sente B, Pascal TG, Weynants V, Clarke SC, Taddei L, Wilkinson TMA, Devaster JM, Devos N. Impact of bacterial strain acquisition in the lung of patients with COPD: the AERIS study. Infect Dis (Lond) 2022; 54:784-793. [PMID: 35794793 DOI: 10.1080/23744235.2022.2092648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Bacterial infections are associated with acute exacerbations of chronic obstructive pulmonary disease (AECOPD), but the mechanism is incompletely understood. METHOD In a COPD observational study (NCT01360398), sputum samples were collected monthly at the stable state and exacerbation. Post-hoc analyses of 1307 non-typeable Haemophilus influenzae (NTHi) isolates from 20 patients and 756 Moraxella catarrhalis isolates from 38 patients in one year of follow-up were conducted by multilocus sequence typing (MLST). All isolates came from cultured sputum samples that were analyzed for bacterial species presence, apparition (infection not detected at the preceding visit), or acquisition (first-time infection), with the first study visit as a baseline. Strain apparition or new strain acquisition was analyzed by MLST. The odds ratio (OR) of experiencing an exacerbation vs. stable state was estimated by conditional logistic regression modelling, stratified by patient. RESULTS The culture results confirmed a significant association with exacerbation only for NTHi species presence (OR 2.28; 95% confidence interval [CI]: 1.12-4.64) and strain apparition (OR 2.38; 95% CI: 1.08-5.27). For M. catarrhalis, although confidence intervals overlapped, the association with exacerbation for first-time species acquisition (OR 5.99; 2.75-13.02) appeared stronger than species presence (OR 3.67; 2.10-6.40), new strain acquisition (OR 2.94; 1.43-6.04), species apparition (OR 4.18; 2.29-7.63), and strain apparition (OR 2.78; 1.42-5.42). This may suggest that previous M. catarrhalis colonization may modify the risk of exacerbation associated with M. catarrhalis infection. CONCLUSIONS The results confirm that NTHi and M. catarrhalis infections are associated with AECOPD but suggest different dynamic mechanisms in triggering exacerbations.
Collapse
Affiliation(s)
| | | | | | - Karen Osman
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - David W Cleary
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK.,Southampton NIHR Biomedical Research Centre, University Hospital Southampton Foundation NHS Trust, Southampton, UK
| | | | | | | | - Stuart C Clarke
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK.,Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | | | - Tom M A Wilkinson
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK.,Southampton NIHR Biomedical Research Centre, University Hospital Southampton Foundation NHS Trust, Southampton, UK.,Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | | | | | | |
Collapse
|
6
|
Page LK, Staples KJ, Spalluto CM, Watson A, Wilkinson TMA. Influence of Hypoxia on the Epithelial-Pathogen Interactions in the Lung: Implications for Respiratory Disease. Front Immunol 2021; 12:653969. [PMID: 33868294 PMCID: PMC8044850 DOI: 10.3389/fimmu.2021.653969] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Under normal physiological conditions, the lung remains an oxygen rich environment. However, prominent regions of hypoxia are a common feature of infected and inflamed tissues and many chronic inflammatory respiratory diseases are associated with mucosal and systemic hypoxia. The airway epithelium represents a key interface with the external environment and is the first line of defense against potentially harmful agents including respiratory pathogens. The protective arsenal of the airway epithelium is provided in the form of physical barriers, and the production of an array of antimicrobial host defense molecules, proinflammatory cytokines and chemokines, in response to activation by receptors. Dysregulation of the airway epithelial innate immune response is associated with a compromised immunity and chronic inflammation of the lung. An increasing body of evidence indicates a distinct role for hypoxia in the dysfunction of the airway epithelium and in the responses of both innate immunity and of respiratory pathogens. Here we review the current evidence around the role of tissue hypoxia in modulating the host-pathogen interaction at the airway epithelium. Furthermore, we highlight the work needed to delineate the role of tissue hypoxia in the pathophysiology of chronic inflammatory lung diseases such as asthma, cystic fibrosis, and chronic obstructive pulmonary disease in addition to novel respiratory diseases such as COVID-19. Elucidating the molecular mechanisms underlying the epithelial-pathogen interactions in the setting of hypoxia will enable better understanding of persistent infections and complex disease processes in chronic inflammatory lung diseases and may aid the identification of novel therapeutic targets and strategies.
Collapse
Affiliation(s)
- Lee K. Page
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
| | - Karl J. Staples
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - C. Mirella Spalluto
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - Alastair Watson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
- Birmingham Medical School, University of Birmingham, Birmingham, United Kingdom
| | - Tom M. A. Wilkinson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
7
|
Ritchie AI, Wedzicha JA. Definition, Causes, Pathogenesis, and Consequences of Chronic Obstructive Pulmonary Disease Exacerbations. Clin Chest Med 2020; 41:421-438. [PMID: 32800196 PMCID: PMC7423341 DOI: 10.1016/j.ccm.2020.06.007] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Andrew I Ritchie
- National Heart and Lung Institute, Guy Scadding Building, Imperial College London, Dovehouse Street, London SW3 6JY, United Kingdom
| | - Jadwiga A Wedzicha
- National Heart and Lung Institute, Guy Scadding Building, Imperial College London, Dovehouse Street, London SW3 6JY, United Kingdom.
| |
Collapse
|
8
|
Bouquet J, Tabor DE, Silver JS, Nair V, Tovchigrechko A, Griffin MP, Esser MT, Sellman BR, Jin H. Microbial burden and viral exacerbations in a longitudinal multicenter COPD cohort. Respir Res 2020; 21:77. [PMID: 32228581 PMCID: PMC7104712 DOI: 10.1186/s12931-020-01340-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/23/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease characterized by frequent exacerbation phenotypes independent of disease stage. Increasing evidence shows that the microbiota plays a role in disease progression and severity, but long-term and international multicenter assessment of the variations in viral and bacterial communities as drivers of exacerbations are lacking. METHODS Two-hundred severe COPD patients from Europe and North America were followed longitudinally for 3 years. We performed nucleic acid detection for 20 respiratory viruses and 16S ribosomal RNA gene sequencing to evaluate the bacterial microbiota in 1179 sputum samples collected at stable, acute exacerbation and follow-up visits. RESULTS Similar viral and bacterial taxa were found in patients from the USA compared to Bulgaria and Czech Republic but their microbiome diversity was significantly different (P < 0.001) and did not impact exacerbation rates. Virus infection was strongly associated with exacerbation events (P < 5E-20). Human rhinovirus (13.1%), coronavirus (5.1%) and influenza virus (3.6%) constitute the top viral pathogens in triggering exacerbation. Moraxella and Haemophilus were 5-fold and 1.6-fold more likely to be the dominating microbiota during an exacerbation event. Presence of Proteobacteria such as Pseudomonas or Staphylococcus amongst others, were associated with exacerbation events (OR > 0.17; P < 0.02) but more strongly associated with exacerbation frequency (OR > 0.39; P < 4E-10), as confirmed by longitudinal variations and biotyping of the bacterial microbiota, and suggesting a role of the microbiota in sensitizing the lung. CONCLUSIONS This study highlights bacterial taxa in lung sensitization and viral triggers in COPD exacerbations. It provides a global overview of the diverse targets for drug development and explores new microbiome analysis methods to guide future patient management applications.
Collapse
Affiliation(s)
- Jerome Bouquet
- Clinical Pharmacology & Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, South San Francisco, USA.
| | - David E Tabor
- Clinical Pharmacology & Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, South San Francisco, USA
| | - Jonathan S Silver
- Respiratory Inflammation and Autoimmunity, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Varsha Nair
- Clinical Pharmacology & Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, South San Francisco, USA
| | | | - M Pamela Griffin
- Respiratory Inflammation and Autoimmunity, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Mark T Esser
- Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Bret R Sellman
- Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Hong Jin
- Clinical Pharmacology & Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, South San Francisco, USA
| |
Collapse
|
9
|
Murphy TF, Brauer AL, Pettigrew MM, LaFontaine ER, Tettelin H. Persistence of Moraxella catarrhalis in Chronic Obstructive Pulmonary Disease and Regulation of the Hag/MID Adhesin. J Infect Dis 2020; 219:1448-1455. [PMID: 30496439 DOI: 10.1093/infdis/jiy680] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/26/2018] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Persistence of bacterial pathogens in the airways has profound consequences on the course and pathogenesis of chronic obstructive pulmonary disease (COPD). Patients with COPD continuously acquire and clear strains of Moraxella catarrhalis, a major pathogen in COPD. Some strains are cleared quickly and some persist for months to years. The mechanism of the variability in duration of persistence is unknown. METHODS Guided by genome sequences of selected strains, we studied the expression of Hag/MID, hag/mid gene sequences, adherence to human cells, and autoaggregation in longitudinally collected strains of M. catarrhalis from adults with COPD. RESULTS Twenty-eight of 30 cleared strains of M. catarrhalis expressed Hag/MID whereas 17 of 30 persistent strains expressed Hag/MID upon acquisition by patients. All persistent strains ceased expression of Hag/MID during persistence. Expression of Hag/MID in human airways was regulated by slipped-strand mispairing. Virulence-associated phenotypes (adherence to human respiratory epithelial cells and autoaggregation) paralleled Hag/MID expression in airway isolates. CONCLUSIONS Most strains of M. catarrhalis express Hag/MID upon acquisition by adults with COPD and all persistent strains shut off expression during persistence. These observations suggest that Hag/MID is important for initial colonization by M. catarrhalis and that cessation of expression facilitates persistence in COPD airways.
Collapse
Affiliation(s)
- Timothy F Murphy
- Division of Infectious Diseases, Department of Medicine, The State University of New York, Buffalo.,Department of Microbiology and Immunology, The State University of New York, Buffalo.,Clinical and Translational Research Center, University at Buffalo, The State University of New York, Buffalo
| | - Aimee L Brauer
- Division of Infectious Diseases, Department of Medicine, The State University of New York, Buffalo.,Clinical and Translational Research Center, University at Buffalo, The State University of New York, Buffalo
| | - Melinda M Pettigrew
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut
| | - Eric R LaFontaine
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens
| | - Hervé Tettelin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore
| |
Collapse
|
10
|
Hao W, Li M, Zhang C, Zhang Y, Du W. Increased levels of inflammatory biomarker CX3CL1 in patients with chronic obstructive pulmonary disease. Cytokine 2019; 126:154881. [PMID: 31629111 DOI: 10.1016/j.cyto.2019.154881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/03/2019] [Accepted: 10/07/2019] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To investigate the concentration of CX3CL1 in serum of patients with chronic obstructive pulmonary disease (COPD), and to evaluate the associations between the CX3CL1 level and systemic inflammation, small airway obstruction, and COPD assessment test (CAT) scores in COPD patients. METHODS Enzyme-linked immunosorbent assay were utilized to detect the CX3CL1 protein in serum separately from 64 patients with COPD and 53 healthy controls. RESULTS Compared with healthy non-smokers, healthy smokers and COPD non-smokers, serum CX3CL1 protein levels were significantly elevated in COPD smokers (258.33 ± 56.27 pg/mL versus 177.32 ± 43.21 pg/mL, 185.64 ± 47.03 pg/mL, and 226.55 ± 51.79 pg/mL, P < 0.05). Correlation analysis indicated that serum CX3CL1 in COPD smokers was negatively correlated with FEV1/FVC (justified r = -0.319, P < 0.001), FEV1/Pre (justified r = -0.476, P < 0.001), FEV3/FVC (justified r = -0.354, P < 0.001), MMEF25-75/Pre (justified r = -0.428, P < 0.001), but positively correlated with CRP (justified r = 0.331, P < 0.001) and MMP-12 (justified r = 0.352, P < 0.001). However, our results showed no significant correlation between serum CX3CL1 of COPD smokers and the diffusing capacity of the lung for carbon monoxide (DLCO) (justified r = 0.0397, P = 0.6025), but a positive correlation with COPD assessment test (CAT) scores (justified r = 0.367, P < 0.001). Finally, through multivariate linear analysis, statistical results demonstrated age (β = -0.2694, P = 0.005), FEV1/Pred (β = -0.2653, P = 0.003), CRP (β = 0.1427, P = 0.0478) and MMP-12 (β = 0.430, P < 0.001) are independent parameters associated with CX3CL1. CONCLUSION The results demonstrated that elevated circulating CX3CL1 level is associated with the systemic inflammation, small airway obstruction, and CAT scores in COPD patients, suggesting that CX3CL1 may play crucial roles in the pathogenesis of COPD. Blocking CX3CL1 might prevent the progression of chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Wendong Hao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China; Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yan'an University, Yan'an 716099, Shaanxi Province, PR China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China.
| | - Cailian Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yan'an University, Yan'an 716099, Shaanxi Province, PR China
| | - Yunqing Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Yan'an University, Yan'an 716099, Shaanxi Province, PR China
| | - Weiping Du
- Clinical Laboratory Diagnosis Department, The Affiliated Hospital of Yan'an University, Yan'an 716099, Shaanxi Province, PR China
| |
Collapse
|
11
|
Toraldo DM, Conte L. Influence of the Lung Microbiota Dysbiosis in Chronic Obstructive Pulmonary Disease Exacerbations: The Controversial Use of Corticosteroid and Antibiotic Treatments and the Role of Eosinophils as a Disease Marker. J Clin Med Res 2019; 11:667-675. [PMID: 31636780 PMCID: PMC6785281 DOI: 10.14740/jocmr3875] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 07/16/2019] [Indexed: 12/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a debilitating lung disease associated with loss of lung function, poorer quality of life, co-morbidities, significant mortality, and higher health care costs. Frequent acute exacerbations of COPD are sudden worsening of symptoms, the nature of which is associated with bacterial or viral infections. However, one-third of exacerbations remain of undetermined origin. Although it is largely discussed and controversial, current guidelines recommend treatment of exacerbations with bronchodilators, antibiotics, and systemic corticosteroids; this is despite being associated with limited benefits in term of reducing mortality, side effects and without paying attention to the heterogeneity of these exacerbations. Increasing evidence suggests that the lung microbiota plays an important role in COPD and numerous studies have reported differences in the microbiota between healthy and disease states, as well as between exacerbations and stable COPD, leading to the hypothesis that frequent acute exacerbation is more likely to experience significant changes in lung microbiota composition. These findings will need further examination to explain the causes of lung dysbiosis, namely microbial composition, the host response, including the recruitment of eosinophils, lifestyle, diet, cigarette smoking and the use of antibiotics and corticosteroids. It is now important to assess: 1) Whether alterations in the lung microbiota contribute to disease pathogenesis, especially in exacerbations of unknown origin; 2) The role of eosinophils; and 3) Whether the microbiota of the lung can be manipulated therapeutically to improve COPD exacerbation event and disease progression. In summary, we hypothesize that the alterations of the lung microbiota may explain the undetermined origins of exacerbations and that there is an urgent need to facilitate the design of intervention studies that aim at conserving the lung microbial flora.
Collapse
Affiliation(s)
- Domenico Maurizio Toraldo
- Department of Rehabilitation, Respiratory Care Unit, ASL/Lecce, Italy
- Both authors contributed equally to this manuscript
| | - Luana Conte
- Laboratory of Biomedical Physics and Environment, Department of Mathematics and Physics “E. De Giorgi”, University of Salento, Lecce, Italy
- Interdisciplinary Laboratory of Applied Research in Medicine (DReAM), University of Salento, “V. Fazzi” Hospital, Lecce, Italy
- Both authors contributed equally to this manuscript
| |
Collapse
|
12
|
Moghoofei M, Azimzadeh Jamalkandi S, Moein M, Salimian J, Ahmadi A. Bacterial infections in acute exacerbation of chronic obstructive pulmonary disease: a systematic review and meta-analysis. Infection 2019; 48:19-35. [PMID: 31482316 DOI: 10.1007/s15010-019-01350-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 08/16/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Due to the importance of Chronic obstructive pulmonary disease (COPD) as the fourth cause of mortality worldwide and the lack of studies evaluating the prevalence of bacterial infections in disease exacerbation, this systematic review and meta-analysis was performed to determine the prevalence rate of bacterial infections in COPD patients. METHODS PubMed, ISI Web of Science, and Scopus databases were systematically searched for population-based prevalence studies (1980-2018). MeSH terms for "Bacterial infections" and "AECOPD" were used as search keywords. The selected studies were filtered according to the inclusion and exclusion criteria. Fixed and random-effects models were used for estimation of summary effect sizes. Between-study heterogeneity, as well as publication bias, were calculated. RESULTS Finally, 118 out of 31,440 studies were selected. The overall estimation of the prevalence of bacterial infection was 49.59% [95% confidence interval (CI) 0.4418-0.55]. The heterogeneity in estimating the pooled prevalence of bacterial infections was shown in the studies (Cochran Q test: 6615, P < 0.0001, I2 = 98.23%). In addition, S. pneumoniae, H. influenzae, M. catarrhalis, A. baumannii, P. aeruginosa, and S. aureus were the most prevalent reported bacteria. CONCLUSIONS Our results as the first meta-analysis for the issue demonstrated that bacterial infections are an important risk factor for AECOPD. Further studies must be performed for understanding the exact role of bacterial agents in AECOPD and help physicians for more applicable preventive and therapeutic measurements.
Collapse
Affiliation(s)
- Mohsen Moghoofei
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sadegh Azimzadeh Jamalkandi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Masood Moein
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Salimian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Ahmadi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Neuropeptides SP and CGRP Diminish the Moraxella catarrhalis Outer Membrane Vesicle- (OMV-) Triggered Inflammatory Response of Human A549 Epithelial Cells and Neutrophils. Mediators Inflamm 2018; 2018:4847205. [PMID: 30174554 PMCID: PMC6098883 DOI: 10.1155/2018/4847205] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/07/2018] [Accepted: 06/30/2018] [Indexed: 12/30/2022] Open
Abstract
Neuropeptides such as substance P (SP) and calcitonin gene-related peptide (CGRP) play both pro- and anti-inflammatory activities and are produced during infection and inflammation. Moraxella catarrhalis is one of the leading infectious agents responsible for inflammatory exacerbation in chronic obstructive pulmonary disease (COPD). Since the airway inflammation in COPD is connected with activation of both epithelial cells and accumulated neutrophils, in this study we determined the in vitro effects of neuropeptides on the inflammatory potential of these cells in response to M. catarrhalis outer membrane vesicle (OMV) stimulant. The various OMV-mediated proinflammatory effects were demonstrated. Next, using hBD-2-pGL4[luc2] plasmid with luciferase reporter gene, SP and CGRP were shown to inhibit the IL-1β-dependent expression of potent neutrophil chemoattractant, hBD-2 defensin, in transfected A549 epithelial cells (type II alveolar cells) upon OMV stimulation. Both neuropeptides exerted antiapoptotic activity through rescuing a significant fraction of A549 cells from OMV-induced cell death and apoptosis. Finally, CGRP caused an impairment of specific but not azurophilic granule exocytosis from neutrophils as shown by evaluation of gelatinase-associated lipocalin (NGAL) or CD66b expression and elastase release, respectively. Concluding, these findings suggest that SP and CGRP mediate the dampening of proinflammatory action triggered by M. catarrhalis OMVs towards cells engaged in lung inflammation in vitro.
Collapse
|
14
|
van den Broek M, De Boeck I, Claes I, Nizet V, Lebeer S. Multifactorial inhibition of lactobacilli against the respiratory tract pathogen Moraxella catarrhalis. Benef Microbes 2018; 9:429-439. [DOI: 10.3920/bm2017.0101] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Probiotics, mainly lactic acid bacteria (LAB), are widely focused on gastrointestinal applications. However, recent microbiome studies indicate that LAB can be endogenous members of other human body sites such as the upper respiratory tract (URT). Interestingly, DNA-based microbiome research suggests an inverse correlation between the presence of LAB and the occurrence of potential pathogens, such as Moraxella catarrhalis, an important URT pathogen linked to otitis media, sinusitis and chronic obstructive pulmonary disease. However, a direct interaction between these microbes has not been explored in detail. This study investigated the direct antipathogenic effects of Lactobacillus species, including several well-documented probiotic strains, on M. catarrhalis using agar-based assays, time course analysis, biofilm assays and minimal inhibitory concentration (MIC) testing. These assays were performed using spent culture supernatans (SCS) at two pHs (4.3 and 7) and D- and/or L-lactic acid at three pHs (2, 4 and 7). In addition, cell line assays for adhesion competition and immunomodulation were used to substantiate the inhibitory effect of lactobacilli against M. catarrhalis. A proportion of Lactobacillus strains, including the model probiotic Lactobacillus rhamnosus GG, showed a strong and direct activity against M. catarrhalis. Screening of the activity of the SCS after different treatments demonstrated that lactic acid has an important antimicrobial activity against this pathogen – at least in vitro – with mean MIC values for D- and L-lactic acid varying between 0.5 and 27 g/l depending on the pH. Furthermore, L. rhamnosus GG also decreased the adhesion of M. catarrhalis to human airway epithelial Calu-3 cells with more than 50%, and the expression of mucin MUC5AC, pro-inflammatory cytokines interleukin (IL)-8, IL-1β, and tumor necrosis factor-α at least 1.2 fold. This study suggests that several lactobacilli and their key metabolite lactic acid are possible candidates for probiotic therapeutic interventions against URT infections.
Collapse
Affiliation(s)
- M.F.L. van den Broek
- Department of Bioscience Engineering, Research Group Environmental Ecology and Applied Microbiology, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - I. De Boeck
- Department of Bioscience Engineering, Research Group Environmental Ecology and Applied Microbiology, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - I.J.J. Claes
- Department of Bioscience Engineering, Research Group Environmental Ecology and Applied Microbiology, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - V. Nizet
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California, San Diego, 9500 Gilman Drive, MC 0760, La Jolla, CA 92093-0760, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, MC 0687, La Jolla, CA 92093-0760, USA
| | - S. Lebeer
- Department of Bioscience Engineering, Research Group Environmental Ecology and Applied Microbiology, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| |
Collapse
|
15
|
Augustyniak D, Seredyński R, McClean S, Roszkowiak J, Roszniowski B, Smith DL, Drulis-Kawa Z, Mackiewicz P. Virulence factors of Moraxella catarrhalis outer membrane vesicles are major targets for cross-reactive antibodies and have adapted during evolution. Sci Rep 2018; 8:4955. [PMID: 29563531 PMCID: PMC5862889 DOI: 10.1038/s41598-018-23029-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/05/2018] [Indexed: 12/31/2022] Open
Abstract
Moraxella catarrhalis is a common human respiratory tract pathogen. Its virulence factors associated with whole bacteria or outer membrane vesicles (OMVs) aid infection, colonization and may induce specific antibodies. To investigate pathogen-host interactions, we applied integrated bioinformatic and immunoproteomic (2D-electrophoresis, immunoblotting, LC-MS/MS) approaches. We showed that OMV proteins engaged exclusively in complement evasion and colonization strategies, but not those involved in iron transport and metabolism, are major targets for cross-reacting antibodies produced against phylogenetically divergent M. catarrhalis strains. The analysis of 31 complete genomes of M. catarrhalis and other Moraxella revealed that OMV protein-coding genes belong to 64 orthologous groups, five of which are restricted to M. catarrhalis. This species showed a two-fold increase in the number of OMV protein-coding genes relative to its ancestors and animal-pathogenic Moraxella. The appearance of specific OMV factors and the increase in OMV-associated virulence proteins during M. catarrhalis evolution is an interesting example of pathogen adaptation to optimize colonization. This precisely targeted cross-reactive immunity against M. catarrhalis may be an important strategy of host defences to counteract this phenomenon. We demonstrate that cross-reactivity is closely associated with the anti-virulent antibody repertoire which we have linked with adaptation of this pathogen to the host.
Collapse
Affiliation(s)
- Daria Augustyniak
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Przybyszewskiego 63/77, 51-148, Wroclaw, Poland.
| | - Rafał Seredyński
- Department of Physiology, Wroclaw Medical University, T. Chalubinskiego 10, 50-368, Wroclaw, Poland.,Department of Physical Chemistry of Microorganisms, Institute of Genetics and Microbiology, University of Wroclaw, Przybyszewskiego 63/77, 51-148, Wroclaw, Poland
| | - Siobhán McClean
- School of Biomolecular and Biomedical Sciences, UCD O'Brien Centre for Science West, B304, Dublin, Ireland
| | - Justyna Roszkowiak
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Przybyszewskiego 63/77, 51-148, Wroclaw, Poland
| | - Bartosz Roszniowski
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Przybyszewskiego 63/77, 51-148, Wroclaw, Poland
| | - Darren L Smith
- Applied Sciences, University of Northumbria, Ellison Building EBD222, Newcastle upon Tyne, NE1 8ST, UK
| | - Zuzanna Drulis-Kawa
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Przybyszewskiego 63/77, 51-148, Wroclaw, Poland
| | - Paweł Mackiewicz
- Department of Genomics, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383, Wrocław, Poland.
| |
Collapse
|
16
|
Alnahas S, Hagner S, Raifer H, Kilic A, Gasteiger G, Mutters R, Hellhund A, Prinz I, Pinkenburg O, Visekruna A, Garn H, Steinhoff U. IL-17 and TNF-α Are Key Mediators of Moraxella catarrhalis Triggered Exacerbation of Allergic Airway Inflammation. Front Immunol 2017; 8:1562. [PMID: 29184554 PMCID: PMC5694487 DOI: 10.3389/fimmu.2017.01562] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 10/31/2017] [Indexed: 12/20/2022] Open
Abstract
Alterations of the airway microbiome are often associated with pulmonary diseases. For example, detection of the bacterial pathogen Moraxella catarrhalis in the upper airways is linked with an increased risk to develop or exacerbate asthma. However, the mechanisms by which M. catarrhalis augments allergic airway inflammation (AAI) remain unclear. We here characterized the cellular and soluble mediators of M. catarrhalis triggered excacerbation of AAI in wt and IL-17 deficient as well as in animals treated with TNF-α and IL-6 neutralizing antibodies. We compared the type of inflammatory response in M. catarrhalis infected, house dust mite (HDM)-allergic and animals infected with M. catarrhalis at different time points of HDM sensitization. We found that airway infection of mice with M. catarrhalis triggers a strong inflammatory response with massive neutrophilic infiltrates, high amounts of IL-6 and TNF-α and moderate levels of CD4+ T-cell-derived IFN-γ and IL-17. If bacterial infection occurred during HDM allergen sensitization, the allergic airway response was exacerbated, particularly by the expansion of Th17 cells and increased TNF-α levels. Neutralization of IL-17 or TNF-α but not IL-6 resulted in accelerated clearance of M. catarrhalis and effectively prevented infection-induced exacerbation of AAI. Taken together, our data demonstrate an essential role for TNF-α and IL-17 in infection-triggered exacerbation of AAI.
Collapse
Affiliation(s)
- Safa Alnahas
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Stefanie Hagner
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Member of the German Center for Lung Research, University of Marburg, Marburg, Germany
| | - Hartmann Raifer
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Ayse Kilic
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Member of the German Center for Lung Research, University of Marburg, Marburg, Germany
| | - Georg Gasteiger
- Institute of Medical Microbiology and Hygiene, FZI Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany
| | - Reinier Mutters
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Anne Hellhund
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Olaf Pinkenburg
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Alexander Visekruna
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Holger Garn
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Member of the German Center for Lung Research, University of Marburg, Marburg, Germany
| | - Ulrich Steinhoff
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| |
Collapse
|
17
|
Comparing microbiota profiles in induced and spontaneous sputum samples in COPD patients. Respir Res 2017; 18:164. [PMID: 28851370 PMCID: PMC5576328 DOI: 10.1186/s12931-017-0645-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 08/20/2017] [Indexed: 12/24/2022] Open
Abstract
Background Induced and spontaneous sputum are used to evaluate the airways microbiota. Whether the sputum types can be used interchangeably in microbiota research is unknown. Our aim was to compare microbiota in induced and spontaneous sputum from COPD patients sampled during the same consultation. Methods COPD patients from Bergen, Norway, were followed between 2006/2010, examined during the stable state and exacerbations. 30 patients delivered 36 sample pairs. DNA was extracted by enzymatic and mechanical lysis methods. The V3-V4 region of the 16S rRNA gene was PCR-amplified and prepared for paired-end sequencing. Illumina Miseq System was used for sequencing, and Quantitative Insights Into Microbial Ecology (QIIME) and Stata were used for bioinformatics and statistical analyses. Results Approximately 4 million sequences were sorted into 1004 different OTUs and further assigned to 106 different taxa. Pair-wise comparison of both taxonomic composition and beta-diversity revealed significant differences in one or both parameters in 1/3 of sample pairs. Alpha-diversity did not differ. Comparing abundances for each taxa identified, showed statistically significant differences between the mean abundances in induced versus spontaneous samples for 15 taxa when disease state was considered. This included potential pathogens like Haemophilus and Moraxella. Conclusion When studying microbiota in sputum samples one should take into consideration how samples are collected and avoid the usage of both induced and spontaneous sputum in the same study.
Collapse
|
18
|
Ponce-Gallegos MA, Ramírez-Venegas A, Falfán-Valencia R. Th17 profile in COPD exacerbations. Int J Chron Obstruct Pulmon Dis 2017; 12:1857-1865. [PMID: 28694696 PMCID: PMC5491572 DOI: 10.2147/copd.s136592] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
COPD is characterized by an ongoing inflammatory process of the airways that leads to obstruction or limitation of airflow. It is mainly associated with exposure to cigarette smoke. In addition, it is considered, at present, a serious public health problem, ranking fourth in mortality worldwide. Many cells participate in the pathophysiology of COPD, the most important are neutrophils, macrophages and CD4+ and CD8+ T cells. Neutrophil migration to the inflammation area could be mediated largely by cytokines related to CD4+ Th17 lymphocytes, because it has been shown that IL-17A, IL-17F and IL-22 act as inducers for CXCL8, CXCL1, CXCL5, G-CSF, and GM-CSF secretion by epithelial cells of the airways. The aims of these molecules are differentiation, proliferation and recruitment of neutrophils. Furthermore, it is believed that CD4+ lymphocytes Th17 may be involved in protection against pathogens for which Th1 and Th2 are not prepared to fight. In COPD exacerbations, there is an increased cellularity in the lung region and respiratory tract. Therefore, the increase in the number of neutrophils and macrophages in the airways and the increase in proinflammatory cytokines are directly related to the severity of exacerbations and that is the importance of the functions of Th17 profile in this entity.
Collapse
Affiliation(s)
- Marco Antonio Ponce-Gallegos
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico.,Medicine Academic Unit, Universidad Autónoma de Nayarit. Tepic, Nayarit, Mexico.,Interinstitutional Program for Strengthening Research and the Postgraduate in the Pacific (Dolphin), Tepic, Nayarit, México
| | - Alejandra Ramírez-Venegas
- Tobacco Smoking and COPD Research Department, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| |
Collapse
|
19
|
Leung JM, Tiew PY, Mac Aogáin M, Budden KF, Yong VFL, Thomas SS, Pethe K, Hansbro PM, Chotirmall SH. The role of acute and chronic respiratory colonization and infections in the pathogenesis of COPD. Respirology 2017; 22:634-650. [PMID: 28342288 PMCID: PMC7169176 DOI: 10.1111/resp.13032] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 02/09/2017] [Accepted: 02/09/2017] [Indexed: 12/16/2022]
Abstract
COPD is a major global concern, increasingly so in the context of ageing populations. The role of infections in disease pathogenesis and progression is known to be important, yet the mechanisms involved remain to be fully elucidated. While COPD pathogens such as Haemophilus influenzae, Moraxella catarrhalis and Streptococcus pneumoniae are strongly associated with acute exacerbations of COPD (AECOPD), the clinical relevance of these pathogens in stable COPD patients remains unclear. Immune responses in stable and colonized COPD patients are comparable to those detected in AECOPD, supporting a role for chronic colonization in COPD pathogenesis through perpetuation of deleterious immune responses. Advances in molecular diagnostics and metagenomics now allow the assessment of microbe-COPD interactions with unprecedented personalization and precision, revealing changes in microbiota associated with the COPD disease state. As microbial changes associated with AECOPD, disease severity and therapeutic intervention become apparent, a renewed focus has been placed on the microbiology of COPD and the characterization of the lung microbiome in both its acute and chronic states. Characterization of bacterial, viral and fungal microbiota as part of the lung microbiome has the potential to reveal previously unrecognized prognostic markers of COPD that predict disease outcome or infection susceptibility. Addressing such knowledge gaps will ultimately lead to a more complete understanding of the microbe-host interplay in COPD. This will permit clearer distinctions between acute and chronic infections and more granular patient stratification that will enable better management of these features and of COPD.
Collapse
Affiliation(s)
- Janice M. Leung
- Centre for Heart Lung InnovationVancouverBritish ColumbiaCanada
- Division of Respiratory Medicine, St Paul's HospitalUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Pei Yee Tiew
- Department of Respiratory and Critical Care MedicineSingapore General HospitalSingapore
| | - Micheál Mac Aogáin
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingapore
| | - Kurtis F. Budden
- Priority Research Centre for Healthy LungsUniversity of NewcastleNewcastleNew South WalesAustralia
- Hunter Medical Research InstituteNewcastleNew South WalesAustralia
| | | | - Sangeeta S. Thomas
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingapore
| | - Kevin Pethe
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingapore
| | - Philip M. Hansbro
- Priority Research Centre for Healthy LungsUniversity of NewcastleNewcastleNew South WalesAustralia
- Hunter Medical Research InstituteNewcastleNew South WalesAustralia
| | | |
Collapse
|
20
|
Persson LJP, Aanerud M, Hardie JA, Miodini Nilsen R, Bakke PS, Eagan TM, Hiemstra PS. Antimicrobial peptide levels are linked to airway inflammation, bacterial colonisation and exacerbations in chronic obstructive pulmonary disease. Eur Respir J 2017; 49:49/3/1601328. [PMID: 28298400 DOI: 10.1183/13993003.01328-2016] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 11/16/2016] [Indexed: 11/05/2022]
Abstract
Antimicrobial peptides (AMPs) are effectors of host defence against infection, inflammation and wound repair. We aimed to study AMP levels in stable chronic obstructive pulmonary disease (COPD) and during acute exacerbations of COPD (AECOPD), and to examine their relation to clinical parameters and inflammatory markers.The 3-year Bergen COPD Cohort Study included 433 COPD patients and 325 controls. Induced sputum was obtained and analysed for levels of the AMPs human cathelicidin (hCAP18/LL-37) and secretory leukocyte protease inhibitor (SLPI), and for the inflammatory markers interleukin (IL)-8, IL-6 and tumour necrosis factor-α (TNF-α) using immunoassays. Systemic hCAP18/LL-37 and vitamin D levels were also studied. Treating AMPs as response variables, non-parametric tests were applied for univariate comparison, and linear regression to obtain adjusted estimates. The risk of AECOPD was assessed by Cox proportional-hazard regression.Sputum AMP levels were higher in patients with stable COPD (n=215) compared to controls (n=45), and further changed during AECOPD (n=56), with increased hCAP18/LL-37 and decreased SLPI levels. Plasma hCAP18/LL-37 levels showed a similar pattern. In stable COPD, high sputum hCAP18/LL-37 levels were associated with increased risk of AECOPD, non-typeable Haemophilus influenzae colonisation, higher age, ex-smoking and higher levels of inflammatory markers.Altered levels of selected AMPs are linked to airway inflammation, infection and AECOPD, suggesting a role for these peptides in airway defence mechanisms in COPD.
Collapse
Affiliation(s)
- Louise J P Persson
- Dept of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Marianne Aanerud
- Dept of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Jon A Hardie
- Dept of Clinical Science, University of Bergen, Bergen, Norway
| | - Roy Miodini Nilsen
- Centre for Clinical Research, Haukeland University Hospital, Bergen, Norway
| | - Per S Bakke
- Dept of Clinical Science, University of Bergen, Bergen, Norway
| | - Tomas M Eagan
- Dept of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway.,Dept of Clinical Science, University of Bergen, Bergen, Norway
| | - Pieter S Hiemstra
- Dept of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
21
|
Krishna P, Jain A, Bisen PS. Microbiome diversity in the sputum of patients with pulmonary tuberculosis. Eur J Clin Microbiol Infect Dis 2016; 35:1205-10. [PMID: 27142586 DOI: 10.1007/s10096-016-2654-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 04/18/2016] [Indexed: 10/21/2022]
Abstract
TB is a worldwide pandemic. India has the highest burden of TB, with WHO statistics for 2013 giving an estimated incidence figure of 2.1 million cases for India out of a global incidence of 9 million. Microbiota have been shown to be associated with many disease conditions; however, only few studies have been reported for microbiota associated with TB infection. For the first time, we characterized the composition of microbiota of TB patients of India, using high-throughput 16S rRNA gene sequencing and compared it with healthy controls. Phylum-level analysis showed that the relative abundance of Firmicutes and Actinobacteria was significantly higher in TB samples and Neisseria and Veillonella were two dominant genera after Streptococcus. In our study, significantly different core genera in TB and normal population were found as compared with the reported studies. Also, the presence of diverse opportunistic pathogenic microbiota in TB patients increases the complexity and diversity of sputum microbiota. Characterization of the sputum microbiome is likely to provide important pathogenic insights into pulmonary tuberculosis.
Collapse
Affiliation(s)
- P Krishna
- Diagnostics R&D, Avantor Performance Materials India Limited (formerly RFCL Limited), New Delhi, 110020, India
| | - A Jain
- Diagnostics R&D, Avantor Performance Materials India Limited (formerly RFCL Limited), New Delhi, 110020, India
| | - P S Bisen
- School of Studies in Biotechnology, Jiwaji University, Gwalior, 474011 M.P., India.
| |
Collapse
|
22
|
Wang Z, Bafadhel M, Haldar K, Spivak A, Mayhew D, Miller BE, Tal-Singer R, Johnston SL, Ramsheh MY, Barer MR, Brightling CE, Brown JR. Lung microbiome dynamics in COPD exacerbations. Eur Respir J 2016; 47:1082-92. [PMID: 26917613 DOI: 10.1183/13993003.01406-2015] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 12/06/2015] [Indexed: 02/06/2023]
Abstract
Increasing evidence suggests that the lung microbiome plays an important role in chronic obstructive pulmonary disease (COPD) severity. However, the dynamics of the lung microbiome during COPD exacerbations and its potential role in disease aetiology remain poorly understood.We completed a longitudinal 16S ribosomal RNA survey of the lung microbiome on 476 sputum samples collected from 87 subjects with COPD at four visits defined as stable state, exacerbation, 2 weeks post-therapy and 6 weeks recovery.Our analysis revealed a dynamic lung microbiota where changes appeared to be associated with exacerbation events and indicative of specific exacerbation phenotypes. Antibiotic and steroid treatments appear to have differential effects on the lung microbiome. We depict a microbial interaction network for the lung microbiome and suggest that perturbation of a few bacterial operational taxonomic units, in particular Haemophilus spp., could greatly impact the overall microbial community structure. Furthermore, several serum and sputum biomarkers, in particular sputum interleukin-8, appear to be highly correlated with the structure and diversity of the microbiome.Our study furthers the understanding of lung microbiome dynamics in COPD patients and highlights its potential as a biomarker, and possibly a target, for future respiratory therapeutics.
Collapse
Affiliation(s)
- Zhang Wang
- Computational Biology, Target Sciences, GSK R&D, Collegeville, PA, USA These authors contributed equally
| | - Mona Bafadhel
- Respiratory Medicine Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK These authors contributed equally
| | - Koirobi Haldar
- Institute for Lung Health, National Institute for Health Research Respiratory Biomedical Research Unit, Dept of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK Dept of Health Sciences, University of Leicester, Leicester, UK
| | - Aaron Spivak
- Computational Biology, Target Sciences, GSK R&D, Collegeville, PA, USA
| | - David Mayhew
- Computational Biology, Target Sciences, GSK R&D, Collegeville, PA, USA
| | - Bruce E Miller
- Respiratory Therapy Area Unit, GSK R&D, King of Prussia, PA, USA
| | - Ruth Tal-Singer
- Respiratory Therapy Area Unit, GSK R&D, King of Prussia, PA, USA
| | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Mohammadali Yavari Ramsheh
- Institute for Lung Health, National Institute for Health Research Respiratory Biomedical Research Unit, Dept of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Michael R Barer
- Institute for Lung Health, National Institute for Health Research Respiratory Biomedical Research Unit, Dept of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Christopher E Brightling
- Institute for Lung Health, National Institute for Health Research Respiratory Biomedical Research Unit, Dept of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK Dept of Health Sciences, University of Leicester, Leicester, UK Both authors contributed equally
| | - James R Brown
- Computational Biology, Target Sciences, GSK R&D, Collegeville, PA, USA Both authors contributed equally
| |
Collapse
|
23
|
Liu G, Gradstedt H, Ermert D, Englund E, Singh B, Su YC, Johansson ME, Aspberg A, Agarwal V, Riesbeck K, Blom AM. Moraxella catarrhalis Evades Host Innate Immunity via Targeting Cartilage Oligomeric Matrix Protein. THE JOURNAL OF IMMUNOLOGY 2015; 196:1249-58. [DOI: 10.4049/jimmunol.1502071] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/23/2015] [Indexed: 12/13/2022]
|
24
|
Mocca B, Yin D, Gao Y, Wang W. Moraxella catarrhalis-produced nitric oxide has dual roles in pathogenicity and clearance of infection in bacterial-host cell co-cultures. Nitric Oxide 2015; 51:52-62. [PMID: 26537639 DOI: 10.1016/j.niox.2015.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 09/30/2015] [Accepted: 10/26/2015] [Indexed: 11/26/2022]
Abstract
In humans, the free radical nitric oxide (NO) is a concentration-dependent multifunctional signaling or toxic molecule that modulates various physiological and pathological processes, and innate immunity against bacterial infections. Because the expression of bacterial genes encoding nitrite reductase (AniA) and NO reductase (NorB) is highly upregulated in biofilms in vitro, it is important to investigate whether bacterial NO-metabolism might subvert host NO signaling and play pathogenic roles during infection. The Moraxella catarrhalis AniA and NorB directly function in production and reduction of NO. Using M. catarrhalis-human bronchial epithelial cell (HBEC) co-cultures, we recently reported AniA/nitrite-dependent cytotoxic effects on HBECs, including altered protein profiles of HBECs and induced HBEC apoptosis, suggesting bacterial nitrite reduction likely dysregulates host cell gene expression. To further clarify whether nitrite reduction-derived NO or nitrite-dependent stimulation of bacterial growth was responsible for adverse effects on HBECs, we monitored bacterial nitrite reduction, levels of NO in co-cultures and resulted dynamic effects on HBEC proliferation and bacterial viability. This study demonstrated that M. catarrhalis nitrite reduction-derived NO was responsible for observed adverse effects on HBECs at mid-to-late stages of infection. More importantly, our data showed that while nitrite promoted bacterial growth and biofilm formation at early hours of infection, nitrite reduction-derived NO was toxic towards M. catarrhalis in maturing biofilms, suggesting nitrite reduction-derived NO might be a possible dualistic mechanism by which M. catarrhalis promotes diseases and spontaneous resolutions.
Collapse
Affiliation(s)
- Brian Mocca
- Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993-0002, USA
| | - Dandan Yin
- Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993-0002, USA
| | - Yamei Gao
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993-0002, USA
| | - Wei Wang
- Division of Bacterial, Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993-0002, USA.
| |
Collapse
|
25
|
Nasopharyngeal Bacterial Carriage in the Conjugate Vaccine Era with a Focus on Pneumococci. J Immunol Res 2015; 2015:394368. [PMID: 26351646 PMCID: PMC4553195 DOI: 10.1155/2015/394368] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 05/29/2015] [Accepted: 06/10/2015] [Indexed: 12/17/2022] Open
Abstract
Seven-valent pneumococcal conjugate vaccine (PCV7) was included in the UK national immunisation program in 2006, and this was replaced by thirteen-valent PCV in 2010. During this time, the carriage of vaccine-type Streptococcus pneumoniae decreased but pneumococcal carriage remained stable due to increases in non-vaccine-type S. pneumoniae. Carriage studies have been undertaken in various countries to monitor vaccine-type replacement and to help predict the serotypes, which may cause invasive disease. There has been less focus on how conjugate vaccines indirectly affect colonization of other nasopharyngeal bacteria. If the nasopharynx is treated as a niche, then bacterial dynamics are accepted to occur. Alterations in these dynamics have been shown due to seasonal changes, antibiotic use, and sibling/day care interaction. It has been shown that, following PCV7 introduction, an eradication of pneumococcal vaccine types has resulted in increases in the abundance of other respiratory pathogens including Haemophilus influenzae and Staphylococcus aureus. These changes are difficult to attribute to PCV7 introduction alone and these studies do not account for further changes due to PCV13 implementation. This review aims to describe nasopharyngeal cocarriage of respiratory pathogens in the PCV era.
Collapse
|
26
|
Meyer M, Jaspers I. Respiratory protease/antiprotease balance determines susceptibility to viral infection and can be modified by nutritional antioxidants. Am J Physiol Lung Cell Mol Physiol 2015; 308:L1189-201. [PMID: 25888573 PMCID: PMC4587599 DOI: 10.1152/ajplung.00028.2015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/13/2015] [Indexed: 12/13/2022] Open
Abstract
The respiratory epithelium functions as a central orchestrator to initiate and organize responses to inhaled stimuli. Proteases and antiproteases are secreted from the respiratory epithelium and are involved in respiratory homeostasis. Modifications to the protease/antiprotease balance can lead to the development of lung diseases such as emphysema or chronic obstructive pulmonary disease. Furthermore, altered protease/antiprotease balance, in favor for increased protease activity, is associated with increased susceptibility to respiratory viral infections such as influenza virus. However, nutritional antioxidants induce antiprotease expression/secretion and decrease protease expression/activity, to protect against viral infection. As such, this review will elucidate the impact of this balance in the context of respiratory viral infection and lung disease, to further highlight the role epithelial cell-derived proteases and antiproteases contribute to respiratory immune function. Furthermore, this review will offer the use of nutritional antioxidants as possible therapeutics to boost respiratory mucosal responses and/or protect against infection.
Collapse
Affiliation(s)
- Megan Meyer
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ilona Jaspers
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
27
|
Abdillahi SM, Bober M, Nordin S, Hallgren O, Baumgarten M, Erjefält J, Westergren-Thorsson G, Bjermer L, Riesbeck K, Egesten A, Mörgelin M. Collagen VI Is Upregulated in COPD and Serves Both as an Adhesive Target and a Bactericidal Barrier for Moraxella catarrhalis. J Innate Immun 2015; 7:506-17. [PMID: 25925694 DOI: 10.1159/000381213] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 02/25/2015] [Indexed: 11/19/2022] Open
Abstract
Moraxella catarrhalis is a Gram-negative human mucosal commensal and pathogen. It is a common cause of exacerbation in chronic obstructive pulmonary disease (COPD). During the process of infection, host colonization correlates with recognition of host molecular patterns. Importantly, in COPD patients with compromised epithelial integrity the underlying extracellular matrix is exposed and provides potential adhesive targets. Collagen VI is a ubiquitous fibrillar component in the airway mucosa and has been attributed both adhesive and killing properties against Gram-positive bacteria. However, less is known regarding Gram-negative microorganisms. Therefore, in the present study, the interaction of M. catarrhalis with collagen VI was characterized. We found that collagen VI is upregulated in the airways of COPD patients and exposed upon epithelial desquamation. Ex vivo, we inoculated airway biopsies and fibroblasts from COPD patients with M. catarrhalis. The bacteria specifically adhered to collagen VI-containing matrix fibrils. In vitro, purified collagen VI microfibrils bound to bacterial surface structures. The primary adhesion target was mapped to the collagen VI α2-chain. Upon exposure to collagen VI, bacteria were killed by membrane destabilization in physiological conditions. These previously unknown properties of collagen VI provide novel insights into the extracellular matrix innate immunity by quickly entrapping and killing pathogen intruders.
Collapse
|
28
|
Påhlman LI, Jögi A, Gram M, Mori M, Egesten A. Hypoxia down-regulates expression of secretory leukocyte protease inhibitor in bronchial epithelial cells via TGF-β1. BMC Pulm Med 2015; 15:19. [PMID: 25851169 PMCID: PMC4379733 DOI: 10.1186/s12890-015-0016-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/20/2015] [Indexed: 11/10/2022] Open
Abstract
Background Secretory leukocyte protease inhibitor (SLPI) is a protein with anti-protease and antimicrobial properties that is constitutively secreted from the airway epithelium. The importance of maintaining a balance between proteases and anti-proteases, and robust innate defence mechanisms in the airways, is exemplified by inflammatory lung conditions such as chronic obstructive pulmonary disease (COPD) and cystic fibrosis (CF). Both conditions present with a high protease burden in the airways which leads to tissue destruction. These patients also have an impaired innate immune system in the lungs with bacterial colonization and frequent airway infections. Moreover, both diseases are associated with airway hypoxia due to inflammation and mucus plugs. The aim of the present study was to investigate the role of hypoxia on SLPI production from the airway epithelium. Methods Primary human bronchial epithelial cells were grown in sub-immersed cultures or as differentiated epithelium in air liquid interface cultures. Cells were incubated at 21% O2 (normoxia) or 1% O2 (hypoxia), and the release of SLPI was analysed with ELISA. RT-PCR was used to study the expression of SLPI and transforming growth factor β1 (TGF-β1). Results Hypoxia decreased the constitutive production of SLPI by bronchial epithelial cells. The multifunctional cytokine TGF-β1, which is known to affect SLPI expression, showed increased expression in hypoxic bronchial epithelial cells. When bronchial epithelial cells were exposed to exogenous TGF-β1 during normoxia, the SLPI production was down-regulated. Addition of TGF-β1-neutralizing antibodies partially restored SLPI production during hypoxia, showing that TGF-β1 is an important regulator of SLPI during hypoxic conditions. Conclusions The mechanism described here adds to our knowledge of the pathogenesis of severe pulmonary diseases associated with hypoxia, e.g. COPD and CF. The hypoxic down-regulation of SLPI may help explain the protease/anti-protease imbalance associated with these conditions and vulnerability to airway infections. Furthermore, it provides an interesting target for the treatment and prevention of exacerbation in these patients. Electronic supplementary material The online version of this article (doi:10.1186/s12890-015-0016-0) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Abstract
This article represents a review of the current literature on the role of infection in the pathogenesis of chronic obstructive pulmonary disease (COPD), in stable disease, exacerbations, and pneumonia. It outlines the complex interactions between respiratory pathogens and host immune defenses that underlie the clinical manifestations of infection in COPD.
Collapse
Affiliation(s)
- Kamen Rangelov
- Pulmonary and Critical Care Medicine, University at Buffalo, SUNY, 3435 Main Street, Buffalo, NY 14214, USA
| | - Sanjay Sethi
- Pulmonary, Critical Care, and Sleep Medicine, VA Western New York Healthcare System, University at Buffalo, The State University of New York, 3495 Bailey Avenue, Buffalo, NY 14215, USA.
| |
Collapse
|
30
|
Berenson CS, Kruzel RL, Eberhardt E, Dolnick R, Minderman H, Wallace PK, Sethi S. Impaired innate immune alveolar macrophage response and the predilection for COPD exacerbations. Thorax 2014; 69:811-8. [PMID: 24686454 DOI: 10.1136/thoraxjnl-2013-203669] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Alveolar macrophages (AM) in COPD have fundamentally impaired responsiveness to Toll-like receptor 2 (TLR2) and TLR4 ligands of non-typeable Haemophilus influenzae (NTHI). However, the contribution of innate immune dysfunction to exacerbations of COPD is unexplored. We hypothesised that impaired innate AM responses in COPD extend beyond NTHI to other pathogens and are linked with COPD exacerbations and severity. METHODS AMs, obtained by bronchoalveolar lavage from 88 volunteers with stable-to-moderate COPD, were incubated with respiratory pathogens (NTHI, Moraxella catarrhalis (MC), Streptococcus pneumoniae (SP) and TLR ligands lipopolysaccharide, Pam3Cys) and elicited IL-8 and TNF-α were measured by microsphere flow cytometry. NF-κB nuclear translocation was measured by colorimetric assay. AM TLR2 and TLR4 expression was determined by immunolabeling and quantitation of mean fluorescent indices. Participants were monitored prospectively for occurrence of COPD exacerbations for 1 year following bronchoscopy. Non-parametric analyses were used to compare exacerbation-prone and non-exacerbation-prone individuals. RESULTS 29 subjects had at least one exacerbation in the follow-up period (exacerbation-prone) and 59 remained exacerbation-free (non-exacerbation-prone). AMs of exacerbation-prone COPD donors were more refractory to cytokine induction by NTHI (p=0.02), MC (p=0.045) and SP (p=0.046), and to TLR2 (p=0.07) and TLR4 (p=0.028) ligands, and had diminished NF-κB nuclear activation, compared with non-exacerbation-prone counterparts. AMs of exacerbation-prone subjects were more refractory to TLR2 upregulation by MC and SP (p=0.04 each). CONCLUSIONS Our results support a paradigm of impaired innate responses of COPD AMs to respiratory pathogens, mediated by impaired TLR responses, underlying a propensity for exacerbations in COPD.
Collapse
Affiliation(s)
- Charles S Berenson
- Infectious Disease Division, Department of Veterans Affairs Western New York Healthcare System, State University of New York at Buffalo School of Medicine, Buffalo, New York, USA
| | - Ragina L Kruzel
- Infectious Disease Division, Department of Veterans Affairs Western New York Healthcare System, State University of New York at Buffalo School of Medicine, Buffalo, New York, USA
| | - Ellana Eberhardt
- Pulmonary, Critical Care and Sleep Division, Department of Veterans Affairs Western New York Healthcare System, State University of New York at Buffalo School of Medicine, Buffalo, New York, USA
| | - Ree Dolnick
- Department of Flow and Image Cytometry, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Hans Minderman
- Department of Flow and Image Cytometry, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Paul K Wallace
- Department of Flow and Image Cytometry, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Sanjay Sethi
- Pulmonary, Critical Care and Sleep Division, Department of Veterans Affairs Western New York Healthcare System, State University of New York at Buffalo School of Medicine, Buffalo, New York, USA
| |
Collapse
|
31
|
Meyer M, Bauer RN, Letang BD, Brighton L, Thompson E, Simmen RCM, Bonner J, Jaspers I. Regulation and activity of secretory leukoprotease inhibitor (SLPI) is altered in smokers. Am J Physiol Lung Cell Mol Physiol 2013; 306:L269-76. [PMID: 24285265 DOI: 10.1152/ajplung.00290.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A hallmark of cigarette smoking is a shift in the protease/antiprotease balance, in favor of protease activity. However, it has recently been shown that smokers have increased expression of a key antiprotease, secretory leukoprotease inhibitor (SLPI), yet the mechanisms involved in SLPI transcriptional regulation and functional activity of SLPI remain unclear. We examined SLPI mRNA and protein secretion in differentiated nasal epithelial cells (NECs) and nasal lavage fluid (NLF) from nonsmokers and smokers and demonstrated that SLPI expression is increased in NECs and NLF from smokers. Transcriptional regulation of SLPI expression was confirmed using SLPI promoter reporter assays followed by chromatin immunoprecipitation. The role of STAT1 in regulating SLPI expression was further elucidated using WT and stat1(-/-) mice. Our data demonstrate that STAT1 regulates SLPI transcription in epithelial cells and slpi protein in the lungs of mice. Additionally, we reveal that NECs from smokers have increased STAT1 mRNA/protein expression. Finally, we demonstrate that SLPI contained in the nasal mucosa of smokers is proteolytically cleaved but retains functional activity against neutrophil elastase. These results demonstrate that smoking enhances expression of SLPI in NECs in vitro and in vivo, and that this response is regulated by STAT1. In addition, despite posttranslational cleavage of SLPI, antiprotease activity against neutrophil elastase is enhanced in smokers. Together, our findings show that SLPI regulation and activity is altered in the nasal mucosa of smokers, which could have broad implications in the context of respiratory inflammation and infection.
Collapse
Affiliation(s)
- Megan Meyer
- Dept. of Pediatrics, Univ. of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7310.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Infectious Mechanisms Regulating Susceptibility to Acute Exacerbations of COPD. SMOKING AND LUNG INFLAMMATION 2013. [PMCID: PMC7115011 DOI: 10.1007/978-1-4614-7351-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Acute exacerbations of COPD (AECOPD) are defined by clinical criteria, outlined in the Global Initiative for Chronic Obstructive Lung Disease (GOLD) guidelines [1]. These include an acute increase in one or more of the following cardinal symptoms, beyond day to day variability: dyspnea, increased frequency or severity of cough and increased volume or change in character of sputum, which represent an acute increase in airway inflammation. The role of infection in the pathogenesis of COPD, acute exacerbation and disease progression has been a clinical and research question for many years, and the pendulum has swung from infection as a major cause of acute exacerbation and COPD (British Hypothesis) [2], to infection as an unrelated epiphomenon in acute exacerbation [3–5], and back again to infection as integral in the development of AECOPD and likely an important contributor to COPD progression [6–19]. Upwards of 80 % of AECOPD are driven by infectious stimuli, with 40–50 % associated with bacterial infection and 30–50 % associated with acute viral infection, with some exacerbations having dual bacterial and viral causation [20]. Much of the advancement in our understanding of the role of infection is AECOPD is due to the advancement of clinical and research tools that have allowed researchers to accurately characterize the microbial pathogens, and better understand the host-pathogen interactions (Table 1).
Collapse
|
33
|
Hoenderdos K, Condliffe A. The Neutrophil in Chronic Obstructive Pulmonary Disease. Too Little, Too Late or Too Much, Too Soon? Am J Respir Cell Mol Biol 2013; 48:531-9. [DOI: 10.1165/rcmb.2012-0492tr] [Citation(s) in RCA: 260] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
34
|
Su YC, Singh B, Riesbeck K. Moraxella catarrhalis: from interactions with the host immune system to vaccine development. Future Microbiol 2013; 7:1073-100. [PMID: 22953708 DOI: 10.2217/fmb.12.80] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Moraxella catarrhalis is a human-restricted commensal that over the last two decades has developed into an emerging respiratory tract pathogen. The bacterial species is equipped with various adhesins to facilitate its colonization. Successful evasion of the human immune system is a prerequisite for Moraxella infection. This strategy involves induction of an excessive proinflammatory response, intervention of granulocyte recruitment to the infection site, activation of selected pattern recognition receptors and cellular adhesion molecules to counteract the host bacteriolytic attack, as well as, finally, reprogramming of antigen presenting cells. Host immunomodulator molecules are also exploited by Moraxella to aid in resistance against complement killing and host bactericidal molecules. Thus, breaking the basis of Moraxella immune evasion mechanisms is fundamental for future invention of effective therapy in controlling Moraxella infection.
Collapse
Affiliation(s)
- Yu-Ching Su
- Medical Microbiology, Department of Laboratory Medicine Malmö, Lund University, Skåne University Hospital, Malmö, Sweden
| | | | | |
Collapse
|
35
|
Affiliation(s)
- Sanjay Sethi
- Department of MedicineUniversity at Buffalo, SUNYBuffalo, New YorkandDepartment of Veterans Affairs Western New York Healthcare SystemBuffalo, New York
| |
Collapse
|
36
|
Mallia P, Footitt J, Sotero R, Jepson A, Contoli M, Trujillo-Torralbo MB, Kebadze T, Aniscenko J, Oleszkiewicz G, Gray K, Message SD, Ito K, Barnes PJ, Adcock IM, Papi A, Stanciu LA, Elkin SL, Kon OM, Johnson M, Johnston SL. Rhinovirus infection induces degradation of antimicrobial peptides and secondary bacterial infection in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2012; 186:1117-24. [PMID: 23024024 DOI: 10.1164/rccm.201205-0806oc] [Citation(s) in RCA: 199] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Chronic obstructive pulmonary disease (COPD) exacerbations are associated with virus (mostly rhinovirus) and bacterial infections, but it is not known whether rhinovirus infections precipitate secondary bacterial infections. OBJECTIVES To investigate relationships between rhinovirus infection and bacterial infection and the role of antimicrobial peptides in COPD exacerbations. METHODS We infected subjects with moderate COPD and smokers and nonsmokers with normal lung function with rhinovirus. Induced sputum was collected before and repeatedly after rhinovirus infection and virus and bacterial loads measured with quantitative polymerase chain reaction and culture. The antimicrobial peptides secretory leukoprotease inhibitor (SLPI), elafin, pentraxin, LL-37, α-defensins and β-defensin-2, and the protease neutrophil elastase were measured in sputum supernatants. MEASUREMENTS AND MAIN RESULTS After rhinovirus infection, secondary bacterial infection was detected in 60% of subjects with COPD, 9.5% of smokers, and 10% of nonsmokers (P < 0.001). Sputum virus load peaked on Days 5-9 and bacterial load on Day 15. Sputum neutrophil elastase was significantly increased and SLPI and elafin significantly reduced after rhinovirus infection exclusively in subjects with COPD with secondary bacterial infections, and SLPI and elafin levels correlated inversely with bacterial load. CONCLUSIONS Rhinovirus infections are frequently followed by secondary bacterial infections in COPD and cleavage of the antimicrobial peptides SLPI and elafin by virus-induced neutrophil elastase may precipitate these secondary bacterial infections. Therapy targeting neutrophil elastase or enhancing innate immunity may be useful novel therapies for prevention of secondary bacterial infections in virus-induced COPD exacerbations.
Collapse
Affiliation(s)
- Patrick Mallia
- National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
A novel group of Moraxella catarrhalis UspA proteins mediates cellular adhesion via CEACAMs and vitronectin. PLoS One 2012; 7:e45452. [PMID: 23049802 PMCID: PMC3458076 DOI: 10.1371/journal.pone.0045452] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 08/22/2012] [Indexed: 02/01/2023] Open
Abstract
Moraxella catarrhalis (Mx) is a common cause of otitis media and exacerbation of chronic obstructive pulmonary disease, an increasing worldwide problem. Surface proteins UspA1 and UspA2 of Mx bind to a number of human receptors and may function in pathogenesis. Genetic recombination events in the pathogen can generate hybrid proteins termed UspA2H. However, whether certain key functions (e.g. UspA1-specific CEACAM binding) can be exchanged between these adhesin families remains unknown. In this study, we have shown that Mx can incorporate the UspA1 CEACAM1-binding region not only into rare UspA1 proteins devoid of CEACAM-binding ability, but also into UspA2 which normally lack this capacity. Further, a screen of Mx isolates revealed the presence of novel UspA2 Variant proteins (UspA2V) in ∼14% of the CEACAM-binding population. We demonstrate that the expression of UspA2/2V with the CEACAM-binding domain enable Mx to bind both to cell surface CEACAMs and to integrins, the latter via vitronectin. Such properties of UspA2/2V have not been reported to date. The studies demonstrate that the UspA family is much more heterogeneous than previously believed and illustrate the in vivo potential for exchange of functional regions between UspA proteins which could convey novel adhesive functions whilst enhancing immune evasion.
Collapse
|
38
|
The interaction of α-chymotrypsin with one persistent organic pollutant (dicofol): Spectroscope and molecular modeling identification. Food Chem Toxicol 2012; 50:3298-305. [DOI: 10.1016/j.fct.2012.06.037] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 06/15/2012] [Accepted: 06/22/2012] [Indexed: 11/18/2022]
|
39
|
Beasley V, Joshi PV, Singanayagam A, Molyneaux PL, Johnston SL, Mallia P. Lung microbiology and exacerbations in COPD. Int J Chron Obstruct Pulmon Dis 2012; 7:555-69. [PMID: 22969296 PMCID: PMC3437812 DOI: 10.2147/copd.s28286] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the most common chronic respiratory condition in adults and is characterized by progressive airflow limitation that is not fully reversible. The main etiological agents linked with COPD are cigarette smoking and biomass exposure but respiratory infection is believed to play a major role in the pathogenesis of both stable COPD and in acute exacerbations. Acute exacerbations are associated with more rapid decline in lung function and impaired quality of life and are the major causes of morbidity and mortality in COPD. Preventing exacerbations is a major therapeutic goal but currently available treatments for exacerbations are not very effective. Historically, bacteria were considered the main infective cause of exacerbations but with the development of new diagnostic techniques, respiratory viruses are also frequently detected in COPD exacerbations. This article aims to provide a state-of-the art review of current knowledge regarding the role of infection in COPD, highlight the areas of ongoing debate and controversy, and outline emerging technologies and therapies that will influence future diagnostic and therapeutic pathways in COPD.
Collapse
|
40
|
Bacterium-generated nitric oxide hijacks host tumor necrosis factor alpha signaling and modulates the host cell cycle in vitro. J Bacteriol 2012; 194:4059-68. [PMID: 22636782 DOI: 10.1128/jb.00476-12] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In mammalian cells, nitric oxide (NO·) is an important signal molecule with concentration-dependent and often controversial functions of promoting cell survival and inducing cell death. An inducible nitric oxide synthase (iNOS) in various mammalian cells produces higher levels of NO· from l-arginine upon infections to eliminate pathogens. In this study, we reveal novel pathogenic roles of NO· generated by bacteria in bacterium-host cell cocultures using Moraxella catarrhalis, a respiratory tract disease-causing bacterium, as a biological producer of NO·. We recently demonstrated that M. catarrhalis cells that express the nitrite reductase (AniA protein) can produce NO· by reducing nitrite. Our study suggests that, in the presence of pathophysiological levels of nitrite, this opportunistic pathogen hijacks host cell signaling and modulates host gene expression through its ability to produce NO· from nitrite. Bacterium-generated NO· significantly increases the secretion of tumor necrosis factor alpha (TNF-α) and modulates the expression of apoptotic proteins, therefore triggering host cell programmed death partially through TNF-α signaling. Furthermore, our study reveals that bacterium-generated NO· stalls host cell division and directly results in the death of dividing cells by reducing the levels of an essential regulator of cell division. This study provides unique insight into why NO· may exert more severe cytotoxic effects on fast growing cells, providing an important molecular basis for NO·-mediated pathogenesis in infections and possible therapeutic applications of NO·-releasing molecules in tumorigenesis. This study strongly suggests that bacterium-generated NO· can play important pathogenic roles during infections.
Collapse
|
41
|
Döring G, Parameswaran IG, Murphy TF. Differential adaptation of microbial pathogens to airways of patients with cystic fibrosis and chronic obstructive pulmonary disease. FEMS Microbiol Rev 2011; 35:124-46. [PMID: 20584083 DOI: 10.1111/j.1574-6976.2010.00237.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Cystic fibrosis (CF), the most common autosomal recessive disorder in Caucasians, and chronic obstructive pulmonary disease (COPD), a disease of adults, are characterized by chronic lung inflammation, airflow obstruction and extensive tissue remodelling, which have a major impact on patients' morbidity and mortality. Airway inflammation is stimulated in CF by chronic bacterial infections and in COPD by environmental stimuli, particularly from smoking. Pseudomonas aeruginosa is the major bacterial pathogen in CF, while in COPD, Haemophilus influenzae is most frequently observed. Molecular studies indicate that during chronic pulmonary infection, P. aeruginosa clones genotypically and phenotypically adapt to the CF niche, resulting in a highly diverse bacterial community that is difficult to eradicate therapeutically. Pseudomonas aeruginosa clones from COPD patients remain within the airways only for limited time periods, do not adapt and are easily eradicated. However, in a subgroup of severely ill COPD patients, P. aeruginosa clones similar to those in CF persist. In this review, we will discuss the pathophysiology of lung disease in CF and COPD, the complex genotypic and phenotypic adaptation processes of the opportunistic bacterial pathogens and novel treatment options.
Collapse
Affiliation(s)
- Gerd Döring
- Institute of Medical Microbiology and Hygiene, Universitätsklinikum Tübingen, Tübingen, Germany.
| | | | | |
Collapse
|
42
|
Parameswaran GI, Sethi S, Murphy TF. Effects of bacterial infection on airway antimicrobial peptides and proteins in COPD. Chest 2011; 140:611-617. [PMID: 21349930 DOI: 10.1378/chest.10-2760] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Pathogenic bacteria colonize the airways of 30% to 40% of patients with COPD and cause approximately 50% of exacerbations. New strains of nontypeable Haemophilus influenzae (NTHI) and Moraxella catarrhalis are associated with exacerbations. Antimicrobial protein/peptides (AMPs) play important roles in innate lung defense against pathogens. To our knowledge, the changes in AMP baseline levels in respiratory secretions during bacterial colonization and exacerbation have not been described. The objective of this study was to elucidate the effects of the acquisition of a new strain of pathogenic bacteria on the airway levels of AMPs in patients with COPD. METHODS One hundred fifty-three samples from 11 patients were selected from COPD sputum samples collected prospectively over 6 years. Samples were grouped as culture-negative (no pathogenic bacteria), colonization, and exacerbation due to new strains of NTHI and M catarrhalis. Levels of lysozyme, lactoferrin, LL-37, and secretory leukocyte protease inhibitor (SLPI) were measured by enzyme-linked immunosorbent assay and compared among groups by paired analysis. RESULTS Compared with baseline, sputum lysozyme levels were significantly lower during colonization and exacerbation by NTHI (P = .001 and P = .013, respectively) and M catarrhalis (P = .007 and P = .018, respectively); SLPI levels were lower with exacerbation due to NTHI and M catarrhalis (P = .002 and P = .004, respectively), and during colonization by M catarrhalis (P = 032). Lactoferrin levels did not change significantly; LL-37 levels were higher during exacerbation by NTHI and M catarrhalis (P = .001 and P = .018, respectively). CONCLUSIONS Acquisition of NTHI and M catarrhalis is associated with significant changes in airway levels of AMPs, with larger changes in exacerbation. Airway AMP levels are likely to be important in pathogen clearance and clinical outcomes of infection in COPD.
Collapse
Affiliation(s)
- Ganapathi Iyer Parameswaran
- Division of Infectious Diseases, Department of Medicine, University at Buffalo, State University of New York, Buffalo, NY; VA Western New York Healthcare System, Buffalo, NY.
| | - Sanjay Sethi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University at Buffalo, State University of New York, Buffalo, NY; VA Western New York Healthcare System, Buffalo, NY
| | - Timothy F Murphy
- Division of Infectious Diseases, Department of Medicine, University at Buffalo, State University of New York, Buffalo, NY; Division of Microbiology, Department of Medicine, University at Buffalo, State University of New York, Buffalo, NY
| |
Collapse
|