1
|
Rosazza T, Eigentler L, Earl C, Davidson FA, Stanley‐Wall NR. Bacillus subtilis extracellular protease production incurs a context-dependent cost. Mol Microbiol 2023; 120:105-121. [PMID: 37380434 PMCID: PMC10952608 DOI: 10.1111/mmi.15110] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/30/2023]
Abstract
Microbes encounter a wide range of polymeric nutrient sources in various environmental settings, which require processing to facilitate growth. Bacillus subtilis, a bacterium found in the rhizosphere and broader soil environment, is highly adaptable and resilient due to its ability to utilise diverse sources of carbon and nitrogen. Here, we explore the role of extracellular proteases in supporting growth and assess the cost associated with their production. We provide evidence of the essentiality of extracellular proteases when B. subtilis is provided with an abundant, but polymeric nutrient source and demonstrate the extracellular proteases as a shared public good that can operate over a distance. We show that B. subtilis is subjected to a public good dilemma, specifically in the context of growth sustained by the digestion of a polymeric food source. Furthermore, using mathematical simulations, we uncover that this selectively enforced dilemma is driven by the relative cost of producing the public good. Collectively, our findings reveal how bacteria can survive in environments that vary in terms of immediate nutrient accessibility and the consequent impact on the population composition. These findings enhance our fundamental understanding of how bacteria respond to diverse environments, which has importance to contexts ranging from survival in the soil to infection and pathogenesis scenarios.
Collapse
Affiliation(s)
- Thibault Rosazza
- Division of Molecular Microbiology, School of Life ScienceUniversity of DundeeDundeeUK
| | - Lukas Eigentler
- Division of Molecular Microbiology, School of Life ScienceUniversity of DundeeDundeeUK
- Mathematics, School of Science and EngineeringUniversity of DundeeDundeeUK
- Present address:
Evolutionary Biology DepartmentUniversität BielefeldKonsequenz 45Bielefeld33615Germany
| | - Chris Earl
- Division of Molecular Microbiology, School of Life ScienceUniversity of DundeeDundeeUK
| | | | | |
Collapse
|
2
|
Völler M, Addante A, Rulff H, von Lospichl B, Gräber SY, Duerr J, Lauster D, Haag R, Gradzielski M, Mall MA. An optimized protocol for assessment of sputum macrorheology in health and muco-obstructive lung disease. Front Physiol 2022; 13:912049. [PMID: 35991170 PMCID: PMC9388721 DOI: 10.3389/fphys.2022.912049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/29/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Airway mucus provides important protective functions in health and abnormal viscoelasticity is a hallmark of muco-obstructive lung diseases such as cystic fibrosis (CF). However, previous studies of sputum macrorheology from healthy individuals and patients with CF using different experimental protocols yielded in part discrepant results and data on a systematic assessment across measurement settings and conditions remain limited. Objectives: The aim of this study was to develop an optimized and reliable protocol for standardized macrorheological measurements of airway mucus model systems and native human sputum from healthy individuals and patients with muco-obstructive lung disease. Methods: Oscillatory rheological shear measurements were performed using bovine submaxillary mucin (BSM) at different concentrations (2% and 10% solids) and sputum samples from healthy controls (n = 10) and patients with CF (n = 10). Viscoelastic properties were determined by amplitude and frequency sweeps at 25°C and 37°C with or without solvent trap using a cone-plate geometry. Results: Under saturated atmosphere, we did not observe any temperature-dependent differences in 2% and 10% BSM macrorheology, whereas in the absence of evaporation control 10% BSM demonstrated a significantly higher viscoelasticity at 37°C. Similarly, during the measurements without evaporation control at 37°C we observed a substantial increase in the storage modulus G′ and the loss modulus G″ of the highly viscoelastic CF sputum but not in the healthy sputum. Conclusion: Our data show systematically higher viscoelasticity of CF compared to healthy sputum at 25°C and 37°C. For measurements at the higher temperature using a solvent trap to prevent evaporation is essential for macrorheological analysis of mucus model systems and native human sputum. Another interesting finding is that the viscoelastic properties are not much sensitive to the applied experimental deformation and yield robust results despite their delicate consistency. The optimized protocol resulting from this work will facilitate standardized quantitative assessment of abnormalities in viscoelastic properties of airway mucus and response to muco-active therapies in patients with CF and other muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Mirjam Völler
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Annalisa Addante
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Hanna Rulff
- Institute of Chemistry, Technische Universität Berlin, Berlin, Germany
| | | | - Simon Y. Gräber
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Lung Research (DZL), Associated Partner Site, Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Duerr
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Daniel Lauster
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Michael Gradzielski
- Institute of Chemistry, Technische Universität Berlin, Berlin, Germany
- *Correspondence: Michael Gradzielski, ; Marcus A. Mall,
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Lung Research (DZL), Associated Partner Site, Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: Michael Gradzielski, ; Marcus A. Mall,
| |
Collapse
|
3
|
Batson BD, Zorn BT, Radicioni G, Livengood SS, Kumagai T, Dang H, Ceppe A, Clapp PW, Tunney M, Elborn JS, McElvaney NG, Muhlebach MS, Boucher RC, Tiemeyer M, Wolfgang MC, Kesimer M. Cystic Fibrosis Airway Mucus Hyperconcentration Produces a Vicious Cycle of Mucin, Pathogen, and Inflammatory Interactions that Promotes Disease Persistence. Am J Respir Cell Mol Biol 2022; 67:253-265. [PMID: 35486871 PMCID: PMC9348562 DOI: 10.1165/rcmb.2021-0359oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 04/29/2022] [Indexed: 11/24/2022] Open
Abstract
The dynamics describing the vicious cycle characteristic of cystic fibrosis (CF) lung disease, initiated by stagnant mucus and perpetuated by infection and inflammation, remain unclear. Here we determine the effect of the CF airway milieu, with persistent mucoobstruction, resident pathogens, and inflammation, on the mucin quantity and quality that govern lung disease pathogenesis and progression. The concentrations of MUC5AC and MUC5B were measured and characterized in sputum samples from subjects with CF (N = 44) and healthy subjects (N = 29) with respect to their macromolecular properties, degree of proteolysis, and glycomics diversity. These parameters were related to quantitative microbiome and clinical data. MUC5AC and MUC5B concentrations were elevated, 30- and 8-fold, respectively, in CF as compared with control sputum. Mucin parameters did not correlate with hypertonic saline, inhaled corticosteroids, or antibiotics use. No differences in mucin parameters were detected at baseline versus during exacerbations. Mucin concentrations significantly correlated with the age and sputum human neutrophil elastase activity. Although significantly more proteolytic cleavages were detected in CF mucins, their macromolecular properties (e.g., size and molecular weight) were not significantly different than control mucins, likely reflecting the role of S-S bonds in maintaining multimeric structures. No evidence of giant mucin macromolecule reflecting oxidative stress-induced cross-linking was found. Mucin glycomic analysis revealed significantly more sialylated glycans in CF, and the total abundance of nonsulfated O-glycans correlated with the relative abundance of pathogens. Collectively, the interaction of mucins, pathogens, epithelium, and inflammatory cells promotes proteomic and glycomic changes that reflect a persistent mucoobstructive, infectious, and inflammatory state.
Collapse
Affiliation(s)
- Bethany D. Batson
- Marsico Lung Institute/Cystic Fibrosis Research Center
- Department of Pathology and Laboratory Medicine
| | - Bryan T. Zorn
- Marsico Lung Institute/Cystic Fibrosis Research Center
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Giorgia Radicioni
- Marsico Lung Institute/Cystic Fibrosis Research Center
- Department of Pathology and Laboratory Medicine
| | - Stephanie S. Livengood
- Marsico Lung Institute/Cystic Fibrosis Research Center
- Department of Pathology and Laboratory Medicine
| | - Tadahiro Kumagai
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia
| | - Hong Dang
- Marsico Lung Institute/Cystic Fibrosis Research Center
| | - Agathe Ceppe
- Marsico Lung Institute/Cystic Fibrosis Research Center
| | | | - Michael Tunney
- Queen’s University, Belfast, Northern Ireland, United Kingdom; and
| | - J. Stuart Elborn
- Queen’s University, Belfast, Northern Ireland, United Kingdom; and
| | - Noel G. McElvaney
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | | | | | - Michael Tiemeyer
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia
| | - Matthew C. Wolfgang
- Marsico Lung Institute/Cystic Fibrosis Research Center
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Mehmet Kesimer
- Marsico Lung Institute/Cystic Fibrosis Research Center
- Department of Pathology and Laboratory Medicine
| |
Collapse
|
4
|
Mucolytic bacteria: prevalence in various pathological diseases. World J Microbiol Biotechnol 2021; 37:176. [PMID: 34519941 DOI: 10.1007/s11274-021-03145-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023]
Abstract
All mucins are highly glycosylated and a key constituent of the mucus layer that is vigilant against pathogens in many organ systems of animals and humans. The viscous layer is organized in bilayers, i.e., an outer layer that is loosely arranged, variable in thickness, home to the commensal microbiota that grows in the complex environment, and an innermost layer that is stratified, non-aspirated, firmly adherent to the epithelial cells and devoid of any microorganisms. The O-glycosylation moiety represents the site of adhesion for pathogens and due to the increase of motility, mucolytic activity, and upregulation of virulence factors, some microorganisms can circumvent the component of the mucus layer and cause disruption in organ homeostasis. A dysbiotic microbiome, defective mucus barrier, and altered immune response often result in various diseases. In this review, paramount emphasis is given to the role played by the bacterial species directly or indirectly involved in mucin degradation, alteration in mucus secretion or its composition or mucin gene expression, which instigates many diseases in the digestive, respiratory, and other organ systems. A systematic view can help better understand the etiology of some complex disorders such as cystic fibrosis, ulcerative colitis and expand our knowledge about mucin degraders to develop new therapeutic approaches to correct ill effects caused by these mucin-dwelling pathogens.
Collapse
|
5
|
Hassett DJ, Kovall RA, Schurr MJ, Kotagiri N, Kumari H, Satish L. The Bactericidal Tandem Drug, AB569: How to Eradicate Antibiotic-Resistant Biofilm Pseudomonas aeruginosa in Multiple Disease Settings Including Cystic Fibrosis, Burns/Wounds and Urinary Tract Infections. Front Microbiol 2021; 12:639362. [PMID: 34220733 PMCID: PMC8245851 DOI: 10.3389/fmicb.2021.639362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/07/2021] [Indexed: 11/13/2022] Open
Abstract
The life-threatening pandemic concerning multi-drug resistant (MDR) bacteria is an evolving problem involving increased hospitalizations, billions of dollars in medical costs and a remarkably high number of deaths. Bacterial pathogens have demonstrated the capacity for spontaneous or acquired antibiotic resistance and there is virtually no pool of organisms that have not evolved such potentially clinically catastrophic properties. Although many diseases are linked to such organisms, three include cystic fibrosis (CF), burn/blast wounds and urinary tract infections (UTIs), respectively. Thus, there is a critical need to develop novel, effective antimicrobials for the prevention and treatment of such problematic infections. One of the most formidable, naturally MDR bacterial pathogens is Pseudomonas aeruginosa (PA) that is particularly susceptible to nitric oxide (NO), a component of our innate immune response. This susceptibility sets the translational stage for the use of NO-based therapeutics during the aforementioned human infections. First, we discuss how such NO therapeutics may be able to target problematic infections in each of the aforementioned infectious scenarios. Second, we describe a recent discovery based on years of foundational information, a novel drug known as AB569. AB569 is capable of forming a "time release" of NO from S-nitrosothiols (RSNO). AB569, a bactericidal tandem consisting of acidified NaNO2 (A-NO2 -) and Na2-EDTA, is capable of killing all pathogens that are associated with the aforementioned disorders. Third, we described each disease state in brief, the known or predicted effects of AB569 on the viability of PA, its potential toxicity and highly remote possibility for resistance to develop. Finally, we conclude that AB569 can be a viable alternative or addition to conventional antibiotic regimens to treat such highly problematic MDR bacterial infections for civilian and military populations, as well as the economical burden that such organisms pose.
Collapse
Affiliation(s)
- Daniel J Hassett
- Department of Molecular Genetics, Biochemistry and Microbiology, Cincinnati, OH, United States
| | - Rhett A Kovall
- Department of Molecular Genetics, Biochemistry and Microbiology, Cincinnati, OH, United States
| | - Michael J Schurr
- Department of Immunology and Microbiology, University of Colorado Health Sciences, Denver, CO, United States
| | - Nalinikanth Kotagiri
- Division of Pharmacy, University of Colorado Health Sciences, Denver, CO, United States
| | - Harshita Kumari
- Division of Pharmacy, University of Colorado Health Sciences, Denver, CO, United States
| | - Latha Satish
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Shriners Hospitals for Children-Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
6
|
McKelvey MC, Brown R, Ryan S, Mall MA, Weldon S, Taggart CC. Proteases, Mucus, and Mucosal Immunity in Chronic Lung Disease. Int J Mol Sci 2021; 22:5018. [PMID: 34065111 PMCID: PMC8125985 DOI: 10.3390/ijms22095018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
Dysregulated protease activity has long been implicated in the pathogenesis of chronic lung diseases and especially in conditions that display mucus obstruction, such as chronic obstructive pulmonary disease, cystic fibrosis, and non-cystic fibrosis bronchiectasis. However, our appreciation of the roles of proteases in various aspects of such diseases continues to grow. Patients with muco-obstructive lung disease experience progressive spirals of inflammation, mucostasis, airway infection and lung function decline. Some therapies exist for the treatment of these symptoms, but they are unable to halt disease progression and patients may benefit from novel adjunct therapies. In this review, we highlight how proteases act as multifunctional enzymes that are vital for normal airway homeostasis but, when their activity becomes immoderate, also directly contribute to airway dysfunction, and impair the processes that could resolve disease. We focus on how proteases regulate the state of mucus at the airway surface, impair mucociliary clearance and ultimately, promote mucostasis. We discuss how, in parallel, proteases are able to promote an inflammatory environment in the airways by mediating proinflammatory signalling, compromising host defence mechanisms and perpetuating their own proteolytic activity causing structural lung damage. Finally, we discuss some possible reasons for the clinical inefficacy of protease inhibitors to date and propose that, especially in a combination therapy approach, proteases represent attractive therapeutic targets for muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Michael C. McKelvey
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Ryan Brown
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Sinéad Ryan
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany;
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- German Center for Lung Research (DZL), 35392 Gießen, Germany
| | - Sinéad Weldon
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| |
Collapse
|
7
|
Hunter RC. Host-Microbe Interactions: Wallowing in Mucus Mire. Curr Biol 2021; 31:R85-R88. [PMID: 33497639 DOI: 10.1016/j.cub.2020.11.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mucin glycoproteins confer robust protection against infection by regulating bacterial behavior and virulence gene expression, but the mechanistic bases are poorly understood. New work implicates glycan-based signaling through a carbohydrate-binding sensor kinase in mediating pathogen behavior at the mucosal interface.
Collapse
Affiliation(s)
- Ryan C Hunter
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55117, USA.
| |
Collapse
|
8
|
Boo HJ, Park SJ, Noh M, Min HY, Jeong LS, Lee HY. LJ-2698, an Adenosine A3 Receptor Antagonist, Alleviates Elastase-Induced Pulmonary Emphysema in Mice. Biomol Ther (Seoul) 2020; 28:250-258. [PMID: 32062956 PMCID: PMC7216744 DOI: 10.4062/biomolther.2019.162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/15/2019] [Accepted: 12/24/2019] [Indexed: 12/15/2022] Open
Abstract
Emphysema, a major component of chronic obstructive pulmonary disease (COPD), is a leading cause of human death worldwide. The progressive deterioration of lung function that occurs in the disease is caused by chronic inflammation of the airway and destruction of the lung parenchyma. Despite the main impact of inflammation on the pathogenesis of emphysema, current therapeutic regimens mainly offer symptomatic relief and preservation of lung function with little therapeutic impact. In the present study, we aimed to discover novel therapeutics that suppress the pathogenesis of emphysema. Here, we show that LJ-2698, a novel and highly selective antagonist of the adenosine A3 receptor, a G protein-coupled receptor involved in various inflammatory diseases, significantly reversed the elastase-induced destructive changes in murine lungs. We found that LJ-2698 significantly prevented elastase-induced airspace enlargement, resulting in restoration of pulmonary function without causing any obvious changes in body weight in mice. LJ-2698 was found to inhibit matrix metalloproteinase activity and pulmonary cell apoptosis in the murine lung. LJ-2698 treatment induced increases in anti-inflammatory cytokines in macrophages at doses that displayed no significant cytotoxicity in normal cell lines derived from various organs. Treatment with LJ-2698 significantly increased the number of anti-inflammatory M2 macrophages in the lungs. These results implicate the adenosine A3 receptor in the pathogenesis of emphysema. Our findings also demonstrate the potential of LJ-2698 as a novel therapeutic/preventive agent in suppressing disease development with limited toxicity.
Collapse
Affiliation(s)
- Hye-Jin Boo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - So Jung Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Myungkyung Noh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hye-Young Min
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Lak Shin Jeong
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Ho-Young Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
9
|
Rosen BH, Evans TIA, Moll SR, Gray JS, Liang B, Sun X, Zhang Y, Jensen-Cody CW, Swatek AM, Zhou W, He N, Rotti PG, Tyler SR, Keiser NW, Anderson PJ, Brooks L, Li Y, Pope RM, Rajput M, Hoffman EA, Wang K, Harris JK, Parekh KR, Gibson-Corley KN, Engelhardt JF. Infection Is Not Required for Mucoinflammatory Lung Disease in CFTR-Knockout Ferrets. Am J Respir Crit Care Med 2019; 197:1308-1318. [PMID: 29327941 DOI: 10.1164/rccm.201708-1616oc] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Classical interpretation of cystic fibrosis (CF) lung disease pathogenesis suggests that infection initiates disease progression, leading to an exuberant inflammatory response, excessive mucus, and ultimately bronchiectasis. Although symptomatic antibiotic treatment controls lung infections early in disease, lifelong bacterial residence typically ensues. Processes that control the establishment of persistent bacteria in the CF lung, and the contribution of noninfectious components to disease pathogenesis, are poorly understood. OBJECTIVES To evaluate whether continuous antibiotic therapy protects the CF lung from disease using a ferret model that rapidly acquires lethal bacterial lung infections in the absence of antibiotics. METHODS CFTR (cystic fibrosis transmembrane conductance regulator)-knockout ferrets were treated with three antibiotics from birth to several years of age and lung disease was followed by quantitative computed tomography, BAL, and histopathology. Lung disease was compared with CFTR-knockout ferrets treated symptomatically with antibiotics. MEASUREMENTS AND MAIN RESULTS Bronchiectasis was quantified from computed tomography images. BAL was evaluated for cellular differential and features of inflammatory cellular activation, bacteria, fungi, and quantitative proteomics. Semiquantitative histopathology was compared across experimental groups. We demonstrate that lifelong antibiotics can protect the CF ferret lung from infections for several years. Surprisingly, CF animals still developed hallmarks of structural bronchiectasis, neutrophil-mediated inflammation, and mucus accumulation, despite the lack of infection. Quantitative proteomics of BAL from CF and non-CF pairs demonstrated a mucoinflammatory signature in the CF lung dominated by Muc5B and neutrophil chemoattractants and products. CONCLUSIONS These findings implicate mucoinflammatory processes in the CF lung as pathogenic in the absence of clinically apparent bacterial and fungal infections.
Collapse
Affiliation(s)
- Bradley H Rosen
- 1 Department of Anatomy & Cell Biology.,2 Department of Medicine
| | | | | | | | - Bo Liang
- 1 Department of Anatomy & Cell Biology
| | | | | | | | | | | | - Nan He
- 1 Department of Anatomy & Cell Biology
| | - Pavana G Rotti
- 1 Department of Anatomy & Cell Biology.,4 Department of Biomedical Engineering, College of Engineering, and
| | | | | | | | | | | | | | | | | | - Kai Wang
- 7 Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa; and
| | - J Kirk Harris
- 8 Department of Pediatrics, University of Colorado, Aurora, Colorado
| | | | | | | |
Collapse
|
10
|
Tang A, Caballero AR, Marquart ME, Bierdeman MA, O'Callaghan RJ. Mechanism of Pseudomonas aeruginosa Small Protease (PASP), a Corneal Virulence Factor. Invest Ophthalmol Vis Sci 2019; 59:5993-6002. [PMID: 30572344 PMCID: PMC6306078 DOI: 10.1167/iovs.18-25834] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Pseudomonas aeruginosa is the leading cause of contact lens-associated bacterial keratitis. Secreted bacterial proteases have a key role in keratitis, including the P. aeruginosa small protease (PASP), a proven corneal virulence factor. We investigated the mechanism of PASP and its importance to corneal toxicity. Methods PASP, a serine protease, was tested for activity on various substrates. The catalytic triad of PASP was sought by bioinformatic analysis and site-directed mutagenesis. All mutant constructs were expressed in a P. aeruginosa PASP-deficient strain; the resulting proteins were purified using ion-exchange, gel filtration, or affinity chromatography; and the proteolytic activity was assessed by gelatin zymography and a fluorometric assay. The purified PASP proteins with single amino acid changes were injected into rabbit corneas to determine their pathological effects. Results PASP substrates were cleaved at arginine or lysine residues. Alanine substitution of PASP residues Asp-29, His-34, or Ser-47 eliminated protease activity, whereas PASP with substitution for Ser-59 (control) retained activity. Computer modeling and Western blot analysis indicated that formation of a catalytic triad required dimer formation, and zymography demonstrated the protease activity of the homodimer, but not the monomer. PASP with the Ser-47 mutation, but not with the control mutation, lacked corneal toxicity, indicating the importance of protease activity. Conclusions PASP is a secreted serine protease that can cleave proteins at arginine or lysine residues and PASP activity requires dimer or larger aggregates to create a functional active site. Most importantly, proteolytic PASP molecules demonstrated highly significant toxicity for the rabbit cornea.
Collapse
Affiliation(s)
- Aihua Tang
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Armando R Caballero
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Mary E Marquart
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Michael A Bierdeman
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Richard J O'Callaghan
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
11
|
Abstract
INTRODUCTION Cystic fibrosis (CF) is a multisystem illness caused by abnormalities in the CF transmembrane conductance regulator (CFTR) gene and protein. CFTR is an ion channel regulating transport of chloride, bicarbonate, and water, and influencing sodium resorption. It is inherited as an autosomal recessive disorder, and with about 70,000 CF patients worldwide, it is the most common life shortening disease among persons of European descent. CFTR disease-causing mutations have been organized into six classes. : AREAS COVERED Recently, small molecule targeted therapy for specific classes of CFTR abnormalities have included CFTR correctors that decrease protein degradation and CFTR potentiators that increase channel open probability enhancing chloride transport. EXPERT OPINION Although there are many novel medications in preclinical and clinical testing, there is need for safe and effective CFTR modulating drugs and immunomodulatory medications to decrease the abundant neutrophilic inflammation response in the airway without unwanted adverse effects. Thymosin alpha 1 treatment of airway cells isolated from phe508del CF patients and from CF knockout mice, decreased inflammation, increased CFTR maturation, and facilitated translocation of CFTR protein to the plasma membrane increasing channel activity. If similar results are seen in humans with CF, thymosin alpha 1 has the unique potential to be a single molecule therapy for treating CF airway disease.
Collapse
Affiliation(s)
- Bruce K Rubin
- a Children's Hospital of Richmond , Virginia Commonwealth University , Richmond , VA , USA
| |
Collapse
|
12
|
Phospholipid transfer protein and alpha-1 antitrypsin regulate Hck kinase activity during neutrophil degranulation. Sci Rep 2018; 8:15394. [PMID: 30337619 PMCID: PMC6193999 DOI: 10.1038/s41598-018-33851-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/07/2018] [Indexed: 01/21/2023] Open
Abstract
Excessive neutrophil degranulation is a common feature of many inflammatory disorders, including alpha-1 antitrypsin (AAT) deficiency. Our group has demonstrated that phospholipid transfer protein (PLTP) prevents neutrophil degranulation but serine proteases, which AAT inhibits, cleave PLTP in diseased airways. We propose to identify if airway PLTP activity can be restored by AAT augmentation therapy and how PLTP subdues degranulation of neutrophils in AAT deficient subjects. Airway PLTP activity was lower in AAT deficient patients but elevated in the airways of patients on augmentation therapy. Functional AAT protein (from PiMM homozygotes) prevented PLTP cleavage unlike its mutated ZZ variant (PiZZ). PLTP lowered leukotriene B4 induced degranulation of primary, secondary and tertiary granules from neutrophils from both groups (n = 14/group). Neutrophils isolated from Pltp knockout mice have enhance neutrophil degranulation. Both AAT and PLTP reduced neutrophil degranulation and superoxide production, possibly though their inhibition of the Src tyrosine kinase, Hck. Src kinase inhibitors saracatinib and dasatinib reduced neutrophil degranulation and superoxide production. Therefore, AAT protects PLTP from proteolytic cleavage and both AAT and PLTP mediate degranulation, possibly via Hck tyrosine kinase inhibition. Deficiency of AAT could contribute to reduced lung PLTP activity and elevated neutrophil signaling associated with lung disease.
Collapse
|
13
|
Mallia-Milanes B, Dufour A, Philp C, Solis N, Klein T, Fischer M, Bolton CE, Shapiro S, Overall CM, Johnson SR. TAILS proteomics reveals dynamic changes in airway proteolysis controlling protease activity and innate immunity during COPD exacerbations. Am J Physiol Lung Cell Mol Physiol 2018; 315:L1003-L1014. [PMID: 30284925 DOI: 10.1152/ajplung.00175.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Dysregulated protease activity is thought to cause parenchymal and airway damage in chronic obstructive pulmonary disease (COPD). Multiple proteases have been implicated in COPD, and identifying their substrates may reveal new disease mechanisms and treatments. However, as proteases interact with many substrates that may be protease inhibitors or proteases themselves, these webs of protease interactions make the wider consequences of therapeutically targeting proteases difficult to predict. We therefore used a systems approach to determine protease substrates and protease activity in COPD airways. Protease substrates were determined by proteomics using the terminal amine isotopic labeling of substrates (TAILS) methodology in paired sputum samples during stable COPD and exacerbations. Protease activity and specific protein degradation in airway samples were assessed using Western blotting, substrate assays, and ex vivo cleavage assays. Two hundred ninety-nine proteins were identified in human COPD sputum, 125 of which were proteolytically processed, including proteases, protease inhibitors, mucins, defensins, and complement and other innate immune proteins. During exacerbations, airway neutrophils and neutrophil proteases increased and more proteins were cleaved, particularly at multiple sites, consistent with degradation and inactivation. During exacerbations, different substrates were processed, including protease inhibitors, mucins, and complement proteins. Exacerbations were associated with increasing airway elastase activity and increased processing of specific elastase substrates, including secretory leukocyte protease inhibitor. Proteolysis regulates multiple processes including elastase activity and innate immune proteins in COPD airways and differs during stable disease and exacerbations. The complexity of protease, inhibitor, and substrate networks makes the effect of protease inhibitors hard to predict which should be used cautiously.
Collapse
Affiliation(s)
- Brendan Mallia-Milanes
- Division of Respiratory Medicine and National Institute for Health Research Nottingham Biomedical Research Centre Respiratory Theme, University of Nottingham , Nottingham , United Kingdom
| | - Antoine Dufour
- Departments of Oral Biological and Medical Sciences, Biochemistry and Molecular Biology and Centre for Blood Research, Life Sciences Institute, Faculty of Dentistry, University of British Columbia , Vancouver, British Columbia , Canada
| | - Christopher Philp
- Division of Respiratory Medicine and National Institute for Health Research Nottingham Biomedical Research Centre Respiratory Theme, University of Nottingham , Nottingham , United Kingdom.,Nottingham Molecular Pathology Node, University of Nottingham , Nottingham , United Kingdom
| | - Nestor Solis
- Departments of Oral Biological and Medical Sciences, Biochemistry and Molecular Biology and Centre for Blood Research, Life Sciences Institute, Faculty of Dentistry, University of British Columbia , Vancouver, British Columbia , Canada
| | - Theo Klein
- Departments of Oral Biological and Medical Sciences, Biochemistry and Molecular Biology and Centre for Blood Research, Life Sciences Institute, Faculty of Dentistry, University of British Columbia , Vancouver, British Columbia , Canada
| | - Marlies Fischer
- Division of Respiratory Medicine and National Institute for Health Research Nottingham Biomedical Research Centre Respiratory Theme, University of Nottingham , Nottingham , United Kingdom.,Nottingham Molecular Pathology Node, University of Nottingham , Nottingham , United Kingdom
| | - Charlotte E Bolton
- Division of Respiratory Medicine and National Institute for Health Research Nottingham Biomedical Research Centre Respiratory Theme, University of Nottingham , Nottingham , United Kingdom
| | - Steven Shapiro
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Christopher M Overall
- Departments of Oral Biological and Medical Sciences, Biochemistry and Molecular Biology and Centre for Blood Research, Life Sciences Institute, Faculty of Dentistry, University of British Columbia , Vancouver, British Columbia , Canada
| | - Simon R Johnson
- Division of Respiratory Medicine and National Institute for Health Research Nottingham Biomedical Research Centre Respiratory Theme, University of Nottingham , Nottingham , United Kingdom.,Nottingham Molecular Pathology Node, University of Nottingham , Nottingham , United Kingdom
| |
Collapse
|
14
|
Müller L, Murgia X, Siebenbürger L, Börger C, Schwarzkopf K, Sewald K, Häussler S, Braun A, Lehr CM, Hittinger M, Wronski S. Human airway mucus alters susceptibility of Pseudomonas aeruginosa biofilms to tobramycin, but not colistin. J Antimicrob Chemother 2018; 73:2762-2769. [DOI: 10.1093/jac/dky241] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/26/2018] [Indexed: 02/06/2023] Open
Affiliation(s)
- Laura Müller
- Fraunhofer Institute for Toxicology and Experimental Medicine (Fraunhofer ITEM), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Member of the REBIRTH Cluster of Excellence, Nikolai-Fuchs-Straße 1, Hannover, Germany
| | - Xabier Murgia
- Helmholtz Institute for Pharmaceutical Research (HIPS), Helmholtz Centre for Infection Research, Universitätscampus E8.1, Saarbrücken, Germany
- Korea Institute of Science and Technology, KIST Europe, Campus E7.1, Saarbrücken, Germany
| | | | | | - Konrad Schwarzkopf
- Department of Anaesthesia and Intensive Care, Klinikum Saarbrücken, Winterberg 1, Saarbrücken, Germany
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine (Fraunhofer ITEM), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Member of the REBIRTH Cluster of Excellence, Nikolai-Fuchs-Straße 1, Hannover, Germany
| | - Susanne Häussler
- Helmholtz Institute for Infection Research, Inhoffenstraße 7, Braunschweig, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Straße 7, Hannover, Germany
| | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine (Fraunhofer ITEM), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Member of the REBIRTH Cluster of Excellence, Nikolai-Fuchs-Straße 1, Hannover, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research (HIPS), Helmholtz Centre for Infection Research, Universitätscampus E8.1, Saarbrücken, Germany
- PharmBioTec GmbH, Science Park 1, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus, Saarbrücken, Germany
| | | | - Sabine Wronski
- Fraunhofer Institute for Toxicology and Experimental Medicine (Fraunhofer ITEM), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Member of the REBIRTH Cluster of Excellence, Nikolai-Fuchs-Straße 1, Hannover, Germany
| |
Collapse
|
15
|
Bakshani CR, Morales-Garcia AL, Althaus M, Wilcox MD, Pearson JP, Bythell JC, Burgess JG. Evolutionary conservation of the antimicrobial function of mucus: a first defence against infection. NPJ Biofilms Microbiomes 2018; 4:14. [PMID: 30002868 PMCID: PMC6031612 DOI: 10.1038/s41522-018-0057-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/05/2018] [Accepted: 06/06/2018] [Indexed: 02/07/2023] Open
Abstract
Mucus layers often provide a unique and multi-functional hydrogel interface between the epithelial cells of organisms and their external environment. Mucus has exceptional properties including elasticity, changeable rheology and an ability to self-repair by re-annealing, and is therefore an ideal medium for trapping and immobilising pathogens and serving as a barrier to microbial infection. The ability to produce a functional surface mucosa was an important evolutionary step, which evolved first in the Cnidaria, which includes corals, and the Ctenophora. This allowed the exclusion of non-commensal microbes and the subsequent development of the mucus-lined digestive cavity seen in higher metazoans. The fundamental architecture of the constituent glycoprotein mucins is also evolutionarily conserved. Although an understanding of the biochemical interactions between bacteria and the mucus layer are important to the goal of developing new antimicrobial strategies, they remain relatively poorly understood. This review summarises the physicochemical properties and evolutionary importance of mucus, which make it so successful in the prevention of bacterial infection. In addition, the strategies developed by bacteria to counteract the mucus layer are also explored.
Collapse
Affiliation(s)
- Cassie R Bakshani
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Ana L Morales-Garcia
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mike Althaus
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Matthew D Wilcox
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Jeffrey P Pearson
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - John C Bythell
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - J Grant Burgess
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
16
|
McElvaney OJ, Gunaratnam C, Reeves EP, McElvaney NG. A specialized method of sputum collection and processing for therapeutic interventions in cystic fibrosis. J Cyst Fibros 2018; 18:203-211. [PMID: 29960875 DOI: 10.1016/j.jcf.2018.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/12/2018] [Accepted: 06/04/2018] [Indexed: 11/30/2022]
Abstract
Cystic fibrosis (CF) lung disease is characterized by aggressive neutrophil-dominated inflammation mediated in large part by neutrophil elastase (NE), an omnivorous protease released by activated or disintegrating neutrophils and a key therapeutic target. To date, several short-term studies have shown that anti-NE compounds can inhibit NE and have anti-inflammatory effects. However, progression to large-scale or multicenter clinical trials has been hampered by the fact that the current gold standard methodology of evaluating airway NE inhibition, bronchoalveolar lavage (BAL), is invasive, difficult to standardize across sites and excludes those with severe lung disease. Attempts to utilize sputum that is either spontaneously expectorated (SS) or induced (IS) have been hindered by poor reproducibility, often due to the various processing methods employed. In this study, we evaluate TEmperature-controlled Two-step Rapid Isolation of Sputum (TETRIS), a specialized method for the acquisition and processing of SS and IS. Using TETRIS, we show for the first time that NE activity and cytokine levels are comparable in BAL, SS and IS samples taken from the same people with CF (PWCF) on the same day once this protocol is used. We correlate biomarkers in TETRIS-processed IS and clinical outcome measures including FEV1, and show stability and reproducible inhibition of NE over time in IS processed by TETRIS. The data offer a tremendous opportunity to evaluate prognosis and therapeutic interventions in CF and to study the full spectrum of people with PWCF, many of whom have been excluded from previous studies due to being unfit for BAL or unable to expectorate sputum.
Collapse
Affiliation(s)
- O J McElvaney
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - C Gunaratnam
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - E P Reeves
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - N G McElvaney
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland.
| |
Collapse
|
17
|
Abdullah LH, Coakley R, Webster MJ, Zhu Y, Tarran R, Radicioni G, Kesimer M, Boucher RC, Davis CW, Ribeiro CMP. Mucin Production and Hydration Responses to Mucopurulent Materials in Normal versus Cystic Fibrosis Airway Epithelia. Am J Respir Crit Care Med 2018; 197:481-491. [PMID: 29099608 PMCID: PMC5821906 DOI: 10.1164/rccm.201706-1139oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 11/03/2017] [Indexed: 01/01/2023] Open
Abstract
RATIONALE Cystic fibrosis (CF) airways disease produces a mucoobstructive lung phenotype characterized by airways mucus plugging, epithelial mucous cell metaplasia/hyperplasia, chronic infection, and inflammation. Simultaneous biochemical and functional in vivo studies of mucin synthesis and secretion from CF airways are not available. In vitro translational models may quantitate differential CF versus normal mucin and fluid secretory responses to infectious/inflammatory stimuli. OBJECTIVES We tested the hypothesis that CF airways exhibit defective epithelial fluid, but not mucin, secretory responses to bacterial/inflammatory host products. METHODS Well-differentiated primary human bronchial epithelial cultures were exposed to supernatant from mucopurulent material (SMM) from human CF airways as a test of bacterial/inflammatory host product stimulus. Human bronchial epithelia (HBE) with normal CF transmembrane conductance regulator function were compared with ΔF508/ΔF508 CF HBE. MEASUREMENTS AND MAIN RESULTS Acute (up to 60 min) SMM exposure promoted mucin secretion, but mucins were degraded by the proteolytic enzymes present in SMM. Chronic SMM exposure induced upregulation of mucin synthesis and storage and generated absolute increases in basal and stimulated mucin release in normal and CF cultures. These responses were similar in normal and CF cultures. In contrast, SMM produced a coordinated CF transmembrane conductance regulator-mediated Cl- secretory response in normal HBE, but not in CF HBE. The absence of the fluid secretory response in CF produced quantitatively more dehydrated mucus. CONCLUSIONS Our study reveals the interplay between regulation of mucin and fluid secretion rates in inflamed versus noninflamed conditions and why a hyperconcentrated mucus is produced in CF airways.
Collapse
Affiliation(s)
| | - Raymond Coakley
- Marsico Lung Institute and Cystic Fibrosis Research Center
- Department of Medicine
| | | | - Yunxiang Zhu
- Marsico Lung Institute and Cystic Fibrosis Research Center
| | - Robert Tarran
- Marsico Lung Institute and Cystic Fibrosis Research Center
- Department of Cell Biology and Physiology, and
| | | | - Mehmet Kesimer
- Marsico Lung Institute and Cystic Fibrosis Research Center
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Richard C. Boucher
- Marsico Lung Institute and Cystic Fibrosis Research Center
- Department of Medicine
| | - C. William Davis
- Marsico Lung Institute and Cystic Fibrosis Research Center
- Department of Cell Biology and Physiology, and
| | - Carla M. P. Ribeiro
- Marsico Lung Institute and Cystic Fibrosis Research Center
- Department of Medicine
- Department of Cell Biology and Physiology, and
| |
Collapse
|
18
|
Ohneck EJ, Arivett BA, Fiester SE, Wood CR, Metz ML, Simeone GM, Actis LA. Mucin acts as a nutrient source and a signal for the differential expression of genes coding for cellular processes and virulence factors in Acinetobacter baumannii. PLoS One 2018; 13:e0190599. [PMID: 29309434 PMCID: PMC5757984 DOI: 10.1371/journal.pone.0190599] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/18/2017] [Indexed: 12/27/2022] Open
Abstract
The capacity of Acinetobacter baumannii to persist and cause infections depends on its interaction with abiotic and biotic surfaces, including those found on medical devices and host mucosal surfaces. However, the extracellular stimuli affecting these interactions are poorly understood. Based on our previous observations, we hypothesized that mucin, a glycoprotein secreted by lung epithelial cells, particularly during respiratory infections, significantly alters A. baumannii's physiology and its interaction with the surrounding environment. Biofilm, virulence and growth assays showed that mucin enhances the interaction of A. baumannii ATCC 19606T with abiotic and biotic surfaces and its cytolytic activity against epithelial cells while serving as a nutrient source. The global effect of mucin on the physiology and virulence of this pathogen is supported by RNA-Seq data showing that its presence in a low nutrient medium results in the differential transcription of 427 predicted protein-coding genes. The reduced expression of ion acquisition genes and the increased transcription of genes coding for energy production together with the detection of mucin degradation indicate that this host glycoprotein is a nutrient source. The increased expression of genes coding for adherence and biofilm biogenesis on abiotic and biotic surfaces, the degradation of phenylacetic acid and the production of an active type VI secretion system further supports the role mucin plays in virulence. Taken together, our observations indicate that A. baumannii recognizes mucin as an environmental signal, which triggers a response cascade that allows this pathogen to acquire critical nutrients and promotes host-pathogen interactions that play a role in the pathogenesis of bacterial infections.
Collapse
Affiliation(s)
- Emily J. Ohneck
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| | - Brock A. Arivett
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| | - Steven E. Fiester
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| | - Cecily R. Wood
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| | - Maeva L. Metz
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| | - Gabriella M. Simeone
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| | - Luis A. Actis
- Department of Microbiology, Miami University, Oxford, OH, United States of America
| |
Collapse
|
19
|
Investment in secreted enzymes during nutrient-limited growth is utility dependent. Proc Natl Acad Sci U S A 2017; 114:E7796-E7802. [PMID: 28847943 DOI: 10.1073/pnas.1708580114] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Pathogenic bacteria secrete toxins and degradative enzymes that facilitate their growth by liberating nutrients from the environment. To understand bacterial growth under nutrient-limited conditions, we studied resource allocation between cellular and secreted components by the pathogenic bacterium Pseudomonas aeruginosa during growth on a protein substrate that requires extracellular digestion by secreted proteases. We identified a quantitative relationship between the rate of increase of cellular biomass under nutrient-limiting growth conditions and the rate of increase in investment in secreted proteases. Production of secreted proteases is stimulated by secreted signals that convey information about the utility of secreted proteins during nutrient-limited growth. Growth modeling using this relationship recapitulated the observed kinetics of bacterial growth on a protein substrate. The proposed regulatory strategy suggests a rationale for quorum-sensing-dependent stimulation of the production of secreted enzymes whereby investment in secreted enzymes occurs in proportion to the utility they confer. Our model provides a framework that can be applied toward understanding bacterial growth in many environments where growth rate is limited by the availability of nutrients.
Collapse
|
20
|
Marcos-López M, Espinosa Ruiz C, Rodger HD, O'Connor I, MacCarthy E, Esteban MÁ. Local and systemic humoral immune response in farmed Atlantic salmon (Salmo salar L.) under a natural amoebic gill disease outbreak. FISH & SHELLFISH IMMUNOLOGY 2017; 66:207-216. [PMID: 28501445 DOI: 10.1016/j.fsi.2017.05.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/05/2017] [Accepted: 05/09/2017] [Indexed: 06/07/2023]
Abstract
Amoebic gill disease (AGD), caused by the protozoan parasite Neoparamoeba perurans, is one of the most significant infectious diseases for Atlantic salmon (Salmo salar L.) mariculture. The present study investigated the humoral immune response (both local in gill mucus and systemic in serum) of farmed Atlantic salmon naturally infected with N. perurans in commercial sea pens, at two different stages of the disease and after freshwater treatment. Parameters analysed included activity of immune related enzymes (i.e. lysozyme, peroxidase, protease, anti-protease, esterase, alkaline phosphatase), IgM levels, and the terminal carbohydrate profile in the gill mucus. Overall, greater variations between groups were noted in the immune parameters determined in gill mucus than the equivalent in the serum. In gill mucus, IgM levels and peroxidase, lysozyme, esterase and protease activities were decreased in fish showing longer exposure time to the infection and higher disease severity, then showed a sequential increase after treatment. Results obtained highlight the capacity of gills to elicit a local response to the infection, indicate an impaired immune response at the later stages of the disease, and show partial reestablishment of the host immune status after freshwater treatment. In addition to providing data on the humoral response to AGD, this study increases knowledge on gill mucosal humoral immunity, since some of the parameters were analysed for the first time in gill mucus.
Collapse
Affiliation(s)
- Mar Marcos-López
- Marine and Freshwater Research Centre, Galway-Mayo Institute of Technology, Dublin Road, Galway, Co. Galway, Ireland; FishVet Group Ireland, Unit 7b Oranmore Business Park, Oranmore, Co. Galway, Ireland.
| | - Cristóbal Espinosa Ruiz
- Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, Murcia, Spain
| | - Hamish D Rodger
- FishVet Group Ireland, Unit 7b Oranmore Business Park, Oranmore, Co. Galway, Ireland
| | - Ian O'Connor
- Marine and Freshwater Research Centre, Galway-Mayo Institute of Technology, Dublin Road, Galway, Co. Galway, Ireland
| | - Eugene MacCarthy
- Marine and Freshwater Research Centre, Galway-Mayo Institute of Technology, Dublin Road, Galway, Co. Galway, Ireland
| | - M Ángeles Esteban
- Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, Murcia, Spain
| |
Collapse
|
21
|
Cowley AC, Thornton DJ, Denning DW, Horsley A. Aspergillosis and the role of mucins in cystic fibrosis. Pediatr Pulmonol 2017; 52:548-555. [PMID: 27870227 PMCID: PMC5396363 DOI: 10.1002/ppul.23618] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/20/2016] [Accepted: 09/20/2016] [Indexed: 01/08/2023]
Abstract
The prevalence of aspergillosis in CF patients has until recently been underestimated, but increasing evidence suggests that it may play an important role in the progression of CF lung disease. In healthy airways, Aspergillus fumigatus can be efficiently removed from the lung by mechanisms such as mucociliary clearance and cough. However, these mechanisms are defective in CF, allowing pathogens such as A. fumigatus to germinate and establish chronic infections within the airways. The precise means by which A. fumigatus contributes to CF lung disease remain largely unclear. As the first point of contact within the lung, and an important component of the innate immune system, it is likely that the mucus barrier plays an important role in this process. Study of the functional interplay between this vital protective barrier, and in particular its principal structural components, the polymeric gel-forming mucins, and CF pathogens such as A. fumigatus, is at an early stage. A. fumigatus protease activity has been shown to upregulate mucus production by inducing mucin mRNA and protein expression, and A. fumigatus proteases and glycosidases are able to degrade mucins. This may allow A. fumigatus to alter mucus barrier properties to promote fungal colonization of the airways and/or utilize mucins as a nutrient source. Moreover, conidial surface lectin binding to mucin glycans is a key aspect of clearance of Aspergillus from the lung in health but may be an important aspect of colonization, where mucociliary clearance is compromised, as in the CF lung. Here we discuss the nature of the mucus barrier and its mucin components in CF, and how they may be implicated in A. fumigatus infection. Pediatr Pulmonol 2017;52:548-555. © 2016 The Authors. Pediatric Pulmonology. Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Abigail C Cowley
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom.,Faculty of Biology, Medicine and Health, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - David J Thornton
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom.,Faculty of Biology, Medicine and Health, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - David W Denning
- Faculty of Biology, Medicine and Health, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, University of Manchester, Manchester, United Kingdom.,Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Alexander Horsley
- Faculty of Biology, Medicine and Health, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, University of Manchester, Manchester, United Kingdom.,Manchester Adult CF Centre, Manchester, United Kingdom
| |
Collapse
|
22
|
Han Z, Kautto L, Nevalainen H. Secretion of Proteases by an Opportunistic Fungal Pathogen Scedosporium aurantiacum. PLoS One 2017; 12:e0169403. [PMID: 28060882 PMCID: PMC5218550 DOI: 10.1371/journal.pone.0169403] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 12/16/2016] [Indexed: 01/31/2023] Open
Abstract
Scedosporium aurantiacum is an opportunistic filamentous fungus increasingly isolated from the sputum of cystic fibrosis patients, and is especially prevalent in Australia. At the moment, very little is known about the infection mechanism of this fungus. Secreted proteases have been shown to contribute to fungal virulence in several studies with other fungi. Here we have compared the profiles of proteases secreted by a clinical isolate Scedosporium aurantiacum (WM 06.482) and an environmental strain (WM 10.136) grown on a synthetic cystic fibrosis sputum medium supplemented with casein or mucin. Protease activity was assessed using class-specific substrates and inhibitors. Subtilisin-like and trypsin-like serine protease activity was detected in all cultures. The greatest difference in the secretion of proteases between the two strains occurred in mucin-supplemented medium, where the activities of the elastase-like, trypsin-like and aspartic proteases were, overall, 2.5–75 fold higher in the clinical strain compared to the environmental strain. Proteases secreted by the two strains in the mucin-supplemented medium were further analyzed by mass spectrometry. Six homologs of fungal proteases were identified from the clinical strain and five from the environmental strain. Of these, three were common for both strains including a subtilisin peptidase, a putative leucine aminopeptidase and a PA-SaNapH-like protease. Trypsin-like protease was identified by mass spectrometry only in the clinical isolate even though trypsin-like activity was present in all cultures. In contrast, high elastase-like activity was measured in the culture supernatant of the clinical strain but could not be identified by mass spectrometry searching against other fungi in the NCBI database. Future availability of an annotated genome will help finalise identification of the S. aurantiacum proteases.
Collapse
Affiliation(s)
- Zhiping Han
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, Australia
- Biomolecular Frontiers Research Centre, Macquarie University, Sydney, Australia
- * E-mail:
| | - Liisa Kautto
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, Australia
- Biomolecular Frontiers Research Centre, Macquarie University, Sydney, Australia
| | - Helena Nevalainen
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, Australia
- Biomolecular Frontiers Research Centre, Macquarie University, Sydney, Australia
| |
Collapse
|
23
|
Esther CR, Hill DB, Button B, Shi S, Jania C, Duncan EA, Doerschuk CM, Chen G, Ranganathan S, Stick SM, Boucher RC. Sialic acid-to-urea ratio as a measure of airway surface hydration. Am J Physiol Lung Cell Mol Physiol 2017; 312:L398-L404. [PMID: 28062483 DOI: 10.1152/ajplung.00398.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/23/2016] [Accepted: 12/23/2016] [Indexed: 11/22/2022] Open
Abstract
Although airway mucus dehydration is key to pathophysiology of cystic fibrosis (CF) and other airways diseases, measuring mucus hydration is challenging. We explored a robust method to estimate mucus hydration using sialic acid as a marker for mucin content. Terminal sialic acid residues from mucins were cleaved by acid hydrolysis from airway samples, and concentrations of sialic acid, urea, and other biomarkers were analyzed by mass spectrometry. In mucins purified from human airway epithelial (HAE), sialic acid concentrations after acid hydrolysis correlated with mucin concentrations (r2 = 0.92). Sialic acid-to-urea ratios measured from filters applied to the apical surface of cultured HAE correlated to percent solids and were elevated in samples from CF HAEs relative to controls (2.2 ± 1.1 vs. 0.93 ± 1.8, P < 0.01). Sialic acid-to-urea ratios were elevated in bronchoalveolar lavage fluid (BALF) from β-epithelial sodium channel (ENaC) transgenic mice, known to have reduced mucus hydration, and mice sensitized to house dust mite allergen. In a translational application, elevated sialic acid-to-urea ratios were measured in BALF from young children with CF who had airway infection relative to those who did not (5.5 ± 3.7 vs. 1.9 ± 1.4, P < 0.02) and could be assessed simultaneously with established biomarkers of inflammation. The sialic acid-to-urea ratio performed similarly to percent solids, the gold standard measure of mucus hydration. The method proved robust and has potential to serve as flexible techniques to assess mucin hydration, particularly in samples like BALF in which established methods such as percent solids cannot be utilized.
Collapse
Affiliation(s)
- Charles R Esther
- Pediatric Pulmonology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina;
| | - David B Hill
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Brian Button
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Shuai Shi
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Corey Jania
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Elizabeth A Duncan
- Rheumatology, Allergy, and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Claire M Doerschuk
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Pulmonary Diseases and Critical Care Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Gang Chen
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sarath Ranganathan
- Murdoch Children's Research Institute, University of Melbourne, Parkville, Australia; and
| | - Stephen M Stick
- Murdoch Children's Research Institute, University of Melbourne, Parkville, Australia; and
| | - Richard C Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
24
|
Alrahman MA, Yoon SS. Identification of essential genes of Pseudomonas aeruginosa for its growth in airway mucus. J Microbiol 2016; 55:68-74. [DOI: 10.1007/s12275-017-6515-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 10/21/2016] [Accepted: 10/21/2016] [Indexed: 11/30/2022]
|
25
|
Flynn JM, Niccum D, Dunitz JM, Hunter RC. Evidence and Role for Bacterial Mucin Degradation in Cystic Fibrosis Airway Disease. PLoS Pathog 2016; 12:e1005846. [PMID: 27548479 PMCID: PMC4993466 DOI: 10.1371/journal.ppat.1005846] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/05/2016] [Indexed: 02/01/2023] Open
Abstract
Chronic lung infections in cystic fibrosis (CF) patients are composed of complex microbial communities that incite persistent inflammation and airway damage. Despite the high density of bacteria that colonize the lower airways, nutrient sources that sustain bacterial growth in vivo, and how those nutrients are derived, are not well characterized. In this study, we examined the possibility that mucins serve as an important carbon reservoir for the CF lung microbiota. While Pseudomonas aeruginosa was unable to efficiently utilize mucins in isolation, we found that anaerobic, mucin-fermenting bacteria could stimulate the robust growth of CF pathogens when provided intact mucins as a sole carbon source. 16S rRNA sequencing and enrichment culturing of sputum also identified that mucin-degrading anaerobes are ubiquitous in the airways of CF patients. The collective fermentative metabolism of these mucin-degrading communities in vitro generated amino acids and short chain fatty acids (propionate and acetate) during growth on mucin, and the same metabolites were also found in abundance within expectorated sputum. The significance of these findings was supported by in vivo P. aeruginosa gene expression, which revealed a heightened expression of genes required for the catabolism of propionate. Given that propionate is exclusively derived from bacterial fermentation, these data provide evidence for an important role of mucin fermenting bacteria in the carbon flux of the lower airways. More specifically, microorganisms typically defined as commensals may contribute to airway disease by degrading mucins, in turn providing nutrients for pathogens otherwise unable to efficiently obtain carbon in the lung. Persistent CF lung infections are composed of hundreds of microbial taxa whose interactions contribute to respiratory failure. Despite their importance, the complex interplay between the lung microbiota and host environment is poorly understood. For example, the nutrients that sustain bacterial growth in vivo, and how those nutrients are derived, are not well characterized. We reveal that a subset of CF microbiota is capable of fermenting mucins for carbon and energy which, in-turn, can support the carbon demands of other respiratory pathogens in co-culture. Moreover, we show that metabolites consistent with mucin fermentation are abundant within airway secretions, highlighting a potential key role for fermentative metabolisms in CF lung disease. A thorough understanding of pathogen ecology in the CF airway and the nutritional dynamics that sustain their growth will have important implications for the design of new therapeutic strategies and the management of disease progression.
Collapse
Affiliation(s)
- Jeffrey M. Flynn
- Department of Microbiology & Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - David Niccum
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jordan M. Dunitz
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ryan C. Hunter
- Department of Microbiology & Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
26
|
Sumer-Bayraktar Z, Grant OC, Venkatakrishnan V, Woods RJ, Packer NH, Thaysen-Andersen M. Asn347 Glycosylation of Corticosteroid-binding Globulin Fine-tunes the Host Immune Response by Modulating Proteolysis by Pseudomonas aeruginosa and Neutrophil Elastase. J Biol Chem 2016; 291:17727-42. [PMID: 27339896 PMCID: PMC5016167 DOI: 10.1074/jbc.m116.735258] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/10/2016] [Indexed: 12/23/2022] Open
Abstract
Corticosteroid-binding globulin (CBG) delivers anti-inflammatory cortisol to inflamed tissues upon elastase-based proteolysis of the exposed reactive center loop (RCL). However, the molecular mechanisms that regulate the RCL proteolysis by co-existing host and bacterial elastases in inflamed/infected tissues remain unknown. We document that RCL-localized Asn(347) glycosylation fine-tunes the RCL cleavage rate by human neutrophil elastase (NE) and Pseudomonas aeruginosa elastase (PAE) by different mechanisms. NE- and PAE-generated fragments of native and exoglycosidase-treated blood-derived CBG of healthy individuals were monitored by gel electrophoresis and LC-MS/MS to determine the cleavage site(s) and Asn(347) glycosylation as a function of digestion time. The site-specific (Val(344)-Thr(345)) and rapid (seconds to minutes) NE-based RCL proteolysis was significantly antagonized by several volume-enhancing Asn(347) glycan features (i.e. occupancy, triantennary GlcNAc branching, and α1,6-fucosylation) and augmented by Asn(347) NeuAc-type sialylation (all p < 0.05). In contrast, the inefficient (minutes to hours) PAE-based RCL cleavage, which occurred equally well at Thr(345)-Leu(346) and Asn(347)-Leu(348), was abolished by the presence of Asn(347) glycosylation but was enhanced by sialoglycans on neighboring CBG N-sites. Molecular dynamics simulations of various Asn(347) glycoforms of uncleaved CBG indicated that multiple Asn(347) glycan features are modulating the RCL digestion efficiencies by NE/PAE. Finally, high concentrations of cortisol showed weak bacteriostatic effects toward virulent P. aeruginosa, which may explain the low RCL potency of the abundantly secreted PAE during host infection. In conclusion, site-specific CBG N-glycosylation regulates the bioavailability of cortisol in inflamed environments by fine-tuning the RCL proteolysis by endogenous and exogenous elastases. This study offers new molecular insight into host- and pathogen-based manipulation of the human immune system.
Collapse
Affiliation(s)
- Zeynep Sumer-Bayraktar
- From the Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia and
| | - Oliver C Grant
- the Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| | - Vignesh Venkatakrishnan
- From the Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia and
| | - Robert J Woods
- the Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| | - Nicolle H Packer
- From the Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia and
| | - Morten Thaysen-Andersen
- From the Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia and
| |
Collapse
|
27
|
Ridley C, Kirkham S, Williamson SJ, Davis CW, Woodman P, Thornton DJ. Biosynthesis of the polymeric gel-forming mucin MUC5B. Am J Physiol Lung Cell Mol Physiol 2016; 310:L993-L1002. [PMID: 26993521 PMCID: PMC4896102 DOI: 10.1152/ajplung.00046.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/17/2016] [Indexed: 01/19/2023] Open
Abstract
MUC5B is a major polymeric mucin in the airway mucus gel and is an essential component of innate defense of the respiratory epithelium. Knowledge of the synthesis and intracellular processing of MUC5B is incomplete. We investigated the molecular details of MUC5B assembly in primary human bronchial epithelial cells (HBECs) grown at an air-liquid interface (ALI). Electrophoretic and centrifugal separations of intracellular forms of MUC5B probed with antibodies specific for non-O-glycosylated and O-glycosylated forms of the mucin identified three major intracellular populations of MUC5B (non-O-glycosylated monomer and dimer, and O-glycosylated polymers). Biophysical analysis of recombinant MUC5B COOH-terminus (CT5B; D4-B-C-CK) expressed in 293-EBNA cells showed that MUC5B dimerizes by disulfide linkage. Pulse-chase studies in the HBEC ALI cultures showed that non-O-glycosylated MUC5B was synthesized within 20 min of metabolic labeling and O-glycosylated, polymeric mucin within 2 h. Radiolabeled O-glycosylated mucin polymers were secreted within 2 h and the majority were released by 48 h. These data indicate that MUC5B follows a similar assembly to the related glycoprotein, von Willebrand factor (vWF); however, unlike vWF the MUC5B polypeptide shows no evidence of major proteolytic processing of D-domains during the production of the mature secreted polymeric mucin in normal and cystic fibrosis (CF) primary bronchial epithelial cells. In contrast, MUC5B D-domains were modified by neutrophil elastase, a protease commonly found in CF sputum, demonstrating that proteolytic degradation of MUC5B is an extracellular event in CF sputum. These results define the pathway for synthesis of MUC5B in primary human goblet cells.
Collapse
Affiliation(s)
- Caroline Ridley
- Wellcome Trust Centre for Cell-Matrix Research, The University of Manchester, Manchester, United Kingdom; Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom; and
| | - Sara Kirkham
- Wellcome Trust Centre for Cell-Matrix Research, The University of Manchester, Manchester, United Kingdom; Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom; and
| | - Sally J Williamson
- Wellcome Trust Centre for Cell-Matrix Research, The University of Manchester, Manchester, United Kingdom; Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom; and
| | - C William Davis
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina, Chapel Hill, North Carolina
| | - Philip Woodman
- Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom; and
| | - David J Thornton
- Wellcome Trust Centre for Cell-Matrix Research, The University of Manchester, Manchester, United Kingdom; Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom; and
| |
Collapse
|
28
|
Metabolism and Pathogenicity of Pseudomonas aeruginosa Infections in the Lungs of Individuals with Cystic Fibrosis. Microbiol Spectr 2016; 3. [PMID: 26350318 DOI: 10.1128/microbiolspec.mbp-0003-2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Individuals with the genetic disease cystic fibrosis (CF) accumulate mucus or sputum in their lungs. This sputum is a potent growth substrate for a range of potential pathogens, and the opportunistic bacterium Pseudomonas aeruginosa is generally most difficult of these to eradicate. As a result, P. aeruginosa infections are frequently maintained in the CF lung throughout life, and are the leading cause of death for these individuals. While great effort has been expended to better understand and treat these devastating infections, only recently have researchers begun to rigorously examine the roles played by specific nutrients in CF sputum to cue P. aeruginosa pathogenicity. This chapter summarizes the current state of knowledge regarding how P. aeruginosa metabolism in CF sputum affects initiation and maintenance of these infections. It contains an overview of CF lung disease and the mechanisms of P. aeruginosa pathogenicity. Several model systems used to study these infections are described with emphasis on the challenge of replicating the chronic infections observed in humans with CF. Nutrients present in CF sputum are surveyed, and the impacts of these nutrients on the infection are discussed. The chapter concludes by addressing the future of this line of research including the use of next-generation technologies and the potential for metabolism-based therapeutics.
Collapse
|
29
|
A small molecule neutrophil elastase inhibitor, KRP-109, inhibits cystic fibrosis mucin degradation. J Cyst Fibros 2015; 15:325-31. [PMID: 26526358 DOI: 10.1016/j.jcf.2015.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/24/2015] [Accepted: 10/14/2015] [Indexed: 01/31/2023]
Abstract
BACKGROUND Neutrophil elastase (NE) rapidly degrades gel-forming airway mucins in cystic fibrosis (CF) sputum. We hypothesized that KRP-109, a small molecule NE inhibitor, would inhibit CF mucin degradation in vitro. METHODS Sputa were collected from CF patients (n=5) chronically or intermittently infected with Pseudomonas aeruginosa (P.a.). Mucin degradation was analyzed using western blot. Protease inhibitor studies were performed using alpha1-proteinase inhibitor (A1-PI Prolastin®) and KRP-109. Elastase activity assays were performed using spectrophotometry. RESULTS There were significant differences in the amount of active NE in different CF sputum samples. KRP-109 decreased the NE driven mucin degradation in vitro. Pseudomonas elastases appeared to blunt elastase inhibition by A1-PI or KRP-109. CONCLUSION Inhibitors of neutrophil and Pseudomonas-derived elastases might rescue mucus clearance and reverse airway obstruction in CF.
Collapse
|
30
|
Chillappagari S, Preuss J, Licht S, Müller C, Mahavadi P, Sarode G, Vogelmeier C, Guenther A, Nahrlich L, Rubin BK, Henke MO. Altered protease and antiprotease balance during a COPD exacerbation contributes to mucus obstruction. Respir Res 2015; 16:85. [PMID: 26169056 PMCID: PMC4501272 DOI: 10.1186/s12931-015-0247-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/01/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Proteases have been shown to degrade airway mucin proteins and to damage the epithelium impairing mucociliary clearance. There are increased proteases in the COPD airway but changes in protease-antiprotease balance and mucin degradation have not been investigated during the course of a COPD exacerbation. We hypothesized that increased protease levels would lead to mucin degradation in acute COPD exacerbations. METHODS We measured neutrophil elastase (NE) and alpha 1 protease inhibitor (A1-PI) levels using immunoblotting, and conducted protease inhibitor studies, zymograms, elastin substrate assays and cigarette smoke condensate experiments to evaluate the stability of the gel-forming mucins, MUC5AC and MUC5B, before and 5-6 weeks after an acute pulmonary exacerbation of COPD (n = 9 subjects). RESULTS Unexpectedly, mucin concentration and mucin stability were highest at the start of the exacerbation and restored to baseline after 6 weeks. Consistent with these data, immunoblots and zymograms confirmed decreased NE concentration and activity and increased A1-PI at the start of the exacerbation. After recovery there was an increase in NE activity and a decrease in A1-PI levels. In vitro, protease inhibitor studies demonstrated that serine proteases played a key role in mucin degradation. Mucin stability was further enhanced upon treating with cigarette smoke condensate (CSC). CONCLUSION There appears to be rapid consumption of secreted proteases due to an increase in antiproteases, at the start of a COPD exacerbation. This leads to increased mucin gel stability which may be important in trapping and clearing infectious and inflammatory mediators, but this may also contribute acutely to mucus retention.
Collapse
Affiliation(s)
- Shashi Chillappagari
- Department of Pediatrics, Justus Liebig University Giessen, Feulgenstrasse 12, Giessen, 35392, Germany.,Department of Internal Medicine, Justus-Liebig-University Giessen, Klinikstrasse-36, Giessen, 35392, Germany.,Department of Pulmonary Medicine, Philipps-University Marburg, Baldingerstrasse 1, Marburg, 35043, Germany.,Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Jenni Preuss
- Department of Pulmonary Medicine, Philipps-University Marburg, Baldingerstrasse 1, Marburg, 35043, Germany
| | - Sebastian Licht
- Department of Pulmonary Medicine, Philipps-University Marburg, Baldingerstrasse 1, Marburg, 35043, Germany
| | - Christian Müller
- Department of Pulmonary Medicine, Philipps-University Marburg, Baldingerstrasse 1, Marburg, 35043, Germany
| | - Poornima Mahavadi
- Department of Internal Medicine, Justus-Liebig-University Giessen, Klinikstrasse-36, Giessen, 35392, Germany.,Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Gaurav Sarode
- Asklepios Fachkliniken München-Gauting, Robert-Koch-Allee 2, Gauting, 82131, Germany.,Member of the Comprehensive Pneumology Center (CPC), Helmholtz Zentrum, Munich, Germany
| | - Claus Vogelmeier
- Department of Pulmonary Medicine, Philipps-University Marburg, Baldingerstrasse 1, Marburg, 35043, Germany
| | - Andreas Guenther
- Department of Internal Medicine, Justus-Liebig-University Giessen, Klinikstrasse-36, Giessen, 35392, Germany.,Member of the German Centre for Lung Research (DZL), Giessen, Germany.,Member of the European IPF Registry/Biobank, Giessen, Germany.,Lung Clinic Waldhof-Elgershausen, Greifenstein, Germany
| | - Lutz Nahrlich
- Department of Pediatrics, Justus Liebig University Giessen, Feulgenstrasse 12, Giessen, 35392, Germany.,Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Bruce K Rubin
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, 1001 East Marshall Street, Richmond, 23298, VA, USA
| | - Markus O Henke
- Department of Pulmonary Medicine, Philipps-University Marburg, Baldingerstrasse 1, Marburg, 35043, Germany. .,Asklepios Fachkliniken München-Gauting, Robert-Koch-Allee 2, Gauting, 82131, Germany. .,Member of the Comprehensive Pneumology Center (CPC), Helmholtz Zentrum, Munich, Germany.
| |
Collapse
|
31
|
Chandler JD, Min E, Huang J, Nichols DP, Day BJ. Nebulized thiocyanate improves lung infection outcomes in mice. Br J Pharmacol 2015; 169:1166-77. [PMID: 23586967 DOI: 10.1111/bph.12206] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 03/14/2013] [Accepted: 03/28/2013] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Nebulized saline solutions are used in the treatment of multiple pulmonary diseases including cystic fibrosis (CF), asthma and chronic obstructive pulmonary disease (COPD). The benefits of these therapies include improved lung function, phlegm clearance and fewer lung infections. The thiocyanate anion (SCN) is a normal component of the airway epithelial lining fluid (ELF) secreted by pulmonary epithelia with antioxidant and host defence functions. We sought to test if SCN could be nebulized to combat lung infection by bolstering innate immune defence and antioxidant capacity. EXPERIMENTAL APPROACH We established an effective antioxidant concentration of SCN in vitro using a bronchiolar epithelial cell line. We then developed a nebulization method of SCN in mice that increased ELF SCN above this concentration up to 12 h and used this method in a prolonged Pseudomonas aeruginosa infection model to test if increasing SCN improved host defence and infection outcomes. KEY RESULTS SCN protected against cytotoxicity in vitro from acute and sustained exposure to inflammation-associated oxidative stress. Nebulized SCN effectively reduced bacterial load, infection-mediated morbidity and airway inflammation in mice infected with P. aeruginosa. SCN also sustained adaptive increases in reduced GSH in infected mice. CONCLUSIONS AND IMPLICATIONS SCN is a dually protective molecule able to both enhance host defence and decrease tissue injury and inflammation as an antioxidant. Nebulized SCN could be developed to combat lung infections and inflammatory lung disease.
Collapse
Affiliation(s)
- J D Chandler
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | | | | | | | | |
Collapse
|
32
|
Chandler JD, Day BJ. Biochemical mechanisms and therapeutic potential of pseudohalide thiocyanate in human health. Free Radic Res 2015; 49:695-710. [PMID: 25564094 DOI: 10.3109/10715762.2014.1003372] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Thiocyanate (SCN(-)) is a ubiquitous molecule in mammalian biology, reaching up to mM concentrations in extracellular fluids. Two- electron oxidation of SCN(-) by H2O2 produces hypothiocyanous acid (HOSCN), a potent anti-microbial species. This reaction is catalyzed by chordate peroxidases (e.g., myeloperoxidase and lactoperoxidase), occurring in human secretory mucosa, including the oral cavity, airway, and alimentary tract, and regulates resident and transient flora as part of innate immunity. Increasing SCN(-) levels limits the concentrations of a family of 2-electron oxidants (H2O2, hypohalous acids, and haloamines) in favor of HOSCN formation, altering the oxidative impact on host tissue by substitution of repairable thiol and selenol oxidations instead of biomolecule degradation. This fine-tuning of inflammatory oxidation paradoxically associates with maintained host defense and decreased host injury during infections, due in part to phylogenetic differences in the thioredoxin reductase system between mammals and their pathogens. These differences could be exploited by pharmacologic use of SCN(-). Recent preclinical studies have identified anti-microbial and anti-inflammatory effects of SCN(-) in pulmonary and cardiovascular animal models, with implications for treatment of infectious lung disease and atherogenesis. Further research is merited to expand on these findings and identify other diseases where SCN(-) may be of use. High oral bioavailability and an increased knowledge of the biochemical effects of SCN(-) on a subset of pro-inflammatory reactions suggest clinical utility.
Collapse
|
33
|
Chillappagari S, Venkatesan S, Garapati V, Mahavadi P, Munder A, Seubert A, Sarode G, Guenther A, Schmeck BT, Tümmler B, Henke MO. Impaired TLR4 and HIF expression in cystic fibrosis bronchial epithelial cells downregulates hemeoxygenase-1 and alters iron homeostasis in vitro. Am J Physiol Lung Cell Mol Physiol 2014; 307:L791-9. [DOI: 10.1152/ajplung.00167.2014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Hemeoxygenase-1 (HO-1), an inducible heat shock protein, is upregulated in response to multiple cellular insults via oxidative stress, lipopolysaccharides (LPS), and hypoxia. In this study, we investigated in vitro the role of Toll-like receptor 4 (TLR4), hypoxia-inducible factor 1α (HIF-1α), and iron on HO-1 expression in cystic fibrosis (CF). Immunohistochemical analysis of TLR4, HO-1, ferritin, and HIF-1α were performed on lung sections of CFTR−/− and wild-type mice. CFBE41o- and 16HBE14o- cell lines were employed for in vitro analysis via immunoblotting, immunofluorescence, real-time PCR, luciferase reporter gene analysis, and iron quantification. We observed a reduced TLR4, HIF-1α, HO-1, and ferritin in CFBE41o- cell line and CF mice. Knockdown studies using TLR4-siRNA in 16HBE14o- revealed significant decrease of HO-1, confirming the role of TLR4 in HO-1 downregulation. Inhibition of HO-1 using tin protoporphyrin in 16HBE14o- cells resulted in increased iron levels, suggesting a probable role of HO-1 in iron accumulation. Additionally, sequestration of excess iron using iron chelators resulted in increased hypoxia response element response in CFBE41o- and 16HBE14o-, implicating a role of iron in HIF-1α stabilization and HO-1. To conclude, our in vitro results demonstrate that multiple regulatory factors, such as impaired TLR4 surface expression, increased intracellular iron, and decreased HIF-1α, downregulate HO-1 expression in CFBE41o- cells.
Collapse
Affiliation(s)
- Shashi Chillappagari
- Department of Medicine, Pulmonary Critical Care Philipps University, Marburg, Germany
- Institute for Lung Research, Philipps-University, Marburg, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Member of the German Center for Lung Research (DZL)
| | - Shalini Venkatesan
- Department of Internal Medicine, Justus-Liebig-University, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Member of the German Center for Lung Research (DZL)
| | - Virajith Garapati
- Department of Medicine, Pulmonary Critical Care Philipps University, Marburg, Germany
- Institute for Lung Research, Philipps-University, Marburg, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Member of the German Center for Lung Research (DZL)
| | - Poornima Mahavadi
- Department of Internal Medicine, Justus-Liebig-University, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Member of the German Center for Lung Research (DZL)
| | - Antje Munder
- Clinical Research Group ‘Molecular Pathology of Cystic Fibrosis and Pseudomonas Genomics’, Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Germany
| | - Andreas Seubert
- Department of Chemistry-Biochemistry, Philipps University, Marburg, Germany
| | - Gaurav Sarode
- Department of Medicine, Pulmonary Critical Care Philipps University, Marburg, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Andreas Guenther
- Department of Internal Medicine, Justus-Liebig-University, Giessen, Germany
- Lung Clinic Waldhof-Elgershausen, Greifenstein, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Member of the German Center for Lung Research (DZL)
| | - Bernd T. Schmeck
- Institute for Lung Research, Philipps-University, Marburg, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Member of the German Center for Lung Research (DZL)
| | - Burkhard Tümmler
- Clinical Research Group ‘Molecular Pathology of Cystic Fibrosis and Pseudomonas Genomics’, Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Germany
| | - Markus O. Henke
- Department of Medicine, Pulmonary Critical Care Philipps University, Marburg, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Member of the German Center for Lung Research (DZL)
- Pneumology, Asklepios Fachkliniken München-Gauting, Germany
- Comprehensive Pneumology Center (CPC), Helmholtz Zentrum, Munich, Germany
| |
Collapse
|
34
|
Rubin BK, Priftis KN, Schmidt HJ, Henke MO. Secretory hyperresponsiveness and pulmonary mucus hypersecretion. Chest 2014; 146:496-507. [PMID: 25091755 DOI: 10.1378/chest.13-2609] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The term bronchial hyperresponsiveness is generally used to describe a heightened airway smooth muscle bronchoconstrictor response measured by bronchoprovocation testing. However, the airway also responds to inflammation or bronchoprovocation with increased mucus secretion. We use the term "secretory hyperresponsiveness" to mean increased mucus secretion either intrinsically or in response to bronchoprovocation. This is not the same as retained phlegm or sputum. Unlike smooth muscle contraction, which is rapidly reversible using a bronchodilator, mucus hypersecretion produces airflow limitation that reverses more slowly and depends upon secretion clearance from the airway. Certain groups of patients appear to have greater mucus secretory response, including those with middle lobe syndrome, cough-dominant ("cough-variant") asthma, and severe asthma. Secretory hyperresponsiveness also is a component of forms of lung cancer associated with bronchorrhea. An extreme form of secretory hyperresponsiveness may lead to plastic bronchitis, a disease characterized by rigid branching mucus casts that obstruct the airway. Secretory hyperresponsiveness and mucus hypersecretion appear to be related to activation of the extracellular-regulated kinase 1/2, signaling through the epidermal growth factor receptor, or secretory phospholipases A2. Recognizing secretory hyperresponsiveness as a distinct clinical entity may lead to more effective and targeted therapy for these diseases.
Collapse
Affiliation(s)
- Bruce K Rubin
- Department of Pediatrics, School of Medicine, Virginia Commonwealth University, Richmond, VA.
| | - Kostas N Priftis
- Pediatric Pulmonary Unit, Third Department of Paediatrics, University of Athens School of Medicine, University General Hospital Attikon, Athens, Greece
| | - H Joel Schmidt
- Department of Pediatrics, School of Medicine, Virginia Commonwealth University, Richmond, VA
| | - Markus O Henke
- Department of Pulmonary Medicine, Philipps-Universität Marburg, Marburg, Germany
| |
Collapse
|
35
|
Neutrophil elastase causes tissue damage that decreases host tolerance to lung infection with burkholderia species. PLoS Pathog 2014; 10:e1004327. [PMID: 25166912 PMCID: PMC4148436 DOI: 10.1371/journal.ppat.1004327] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 07/11/2014] [Indexed: 02/07/2023] Open
Abstract
Two distinct defense strategies can protect the host from infection: resistance is the ability to destroy the infectious agent, and tolerance is the ability to withstand infection by minimizing the negative impact it has on the host's health without directly affecting pathogen burden. Burkholderia pseudomallei is a Gram-negative bacterium that infects macrophages and causes melioidosis. We have recently shown that inflammasome-triggered pyroptosis and IL-18 are equally important for resistance to B. pseudomallei, whereas IL-1β is deleterious. Here we show that the detrimental role of IL-1β during infection with B. pseudomallei (and closely related B. thailandensis) is due to excessive recruitment of neutrophils to the lung and consequent tissue damage. Mice deficient in the potentially damaging enzyme neutrophil elastase were less susceptible than the wild type C57BL/6J mice to infection, although the bacterial burdens in organs and the extent of inflammation were comparable between C57BL/6J and elastase-deficient mice. In contrast, lung tissue damage and vascular leakage were drastically reduced in elastase-deficient mice compared to controls. Bradykinin levels were higher in C57BL/6 than in elastase-deficient mice; administration of a bradykinin antagonist protected mice from infection, suggesting that increased vascular permeability mediated by bradykinin is one of the mechanisms through which elastase decreases host tolerance to melioidosis. Collectively, these results demonstrate that absence of neutrophil elastase increases host tolerance, rather than resistance, to infection by minimizing host tissue damage.
Collapse
|
36
|
Henderson AG, Ehre C, Button B, Abdullah LH, Cai LH, Leigh MW, DeMaria GC, Matsui H, Donaldson SH, Davis CW, Sheehan JK, Boucher RC, Kesimer M. Cystic fibrosis airway secretions exhibit mucin hyperconcentration and increased osmotic pressure. J Clin Invest 2014; 124:3047-60. [PMID: 24892808 DOI: 10.1172/jci73469] [Citation(s) in RCA: 258] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The pathogenesis of mucoinfective lung disease in cystic fibrosis (CF) patients likely involves poor mucus clearance. A recent model of mucus clearance predicts that mucus flow depends on the relative mucin concentration of the mucus layer compared with that of the periciliary layer; however, mucin concentrations have been difficult to measure in CF secretions. Here, we have shown that the concentration of mucin in CF sputum is low when measured by immunologically based techniques, and mass spectrometric analyses of CF mucins revealed mucin cleavage at antibody recognition sites. Using physical size exclusion chromatography/differential refractometry (SEC/dRI) techniques, we determined that mucin concentrations in CF secretions were higher than those in normal secretions. Measurements of partial osmotic pressures revealed that the partial osmotic pressure of CF sputum and the retained mucus in excised CF lungs were substantially greater than the partial osmotic pressure of normal secretions. Our data reveal that mucin concentration cannot be accurately measured immunologically in proteolytically active CF secretions; mucins are hyperconcentrated in CF secretions; and CF secretion osmotic pressures predict mucus layer-dependent osmotic compression of the periciliary liquid layer in CF lungs. Consequently, mucin hypersecretion likely produces mucus stasis, which contributes to key infectious and inflammatory components of CF lung disease.
Collapse
|
37
|
Ehre C, Ridley C, Thornton DJ. Cystic fibrosis: an inherited disease affecting mucin-producing organs. Int J Biochem Cell Biol 2014; 52:136-45. [PMID: 24685676 DOI: 10.1016/j.biocel.2014.03.011] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 02/28/2014] [Accepted: 03/17/2014] [Indexed: 02/01/2023]
Abstract
Our current understanding of cystic fibrosis (CF) has revealed that the biophysical properties of mucus play a considerable role in the pathogenesis of the disease in view of the fact that most mucus-producing organs are affected in CF patients. In this review, we discuss the potential causal relationship between altered cystic fibrosis transmembrane conductance regulator (CFTR) function and the production of mucus with abnormal biophysical properties in the intestine and lungs, highlighting what has been learned from cell cultures and animal models that mimic CF pathogenesis. A similar cascade of events, including mucus obstruction, infection and inflammation, is common to all epithelia affected by impaired surface hydration. Hence, the main structural components of mucus, namely the polymeric, gel-forming mucins, are critical to the onset of the disease. Defective CFTR leads to epithelial surface dehydration, altered pH/electrolyte composition and mucin concentration. Further, it can influence mucin transition from the intracellular to extracellular environment, potentially resulting in aberrant mucus gel formation. While defective HCO3(-) production has long been identified as a feature of CF, it has only recently been considered as a key player in the transition phase of mucins. We conclude by examining the influence of mucins on the biophysical properties of CF sputum and discuss existing and novel therapies aimed at removing mucus from the lungs. This article is part of a Directed Issue entitled: Cystic Fibrosis: From o-mics to cell biology, physiology, and therapeutic advances.
Collapse
Affiliation(s)
- Camille Ehre
- CF/Pulmonary Research & Treatment Centre, The University of North Carolina at Chapel Hill, USA.
| | - Caroline Ridley
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, UK
| | - David J Thornton
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, UK
| |
Collapse
|
38
|
Biogeochemical forces shape the composition and physiology of polymicrobial communities in the cystic fibrosis lung. mBio 2014; 5:e00956-13. [PMID: 24643867 PMCID: PMC3967525 DOI: 10.1128/mbio.00956-13] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The cystic fibrosis (CF) lung contains thick mucus colonized by opportunistic pathogens which adapt to the CF lung environment over decades. The difficulty associated with sampling airways has impeded a thorough examination of the biochemical microhabitats these pathogens are exposed to. An indirect approach is to study the responses of microbial communities to these microhabitats, facilitated by high-throughput sequencing of microbial DNA and RNA from sputum samples. Microbial metagenomes and metatranscriptomes were sequenced from multiple CF patients, and the reads were assigned taxonomy and function through sequence homology to NCBI and the Kyoto Encyclopedia of Genes and Genomes (KEGG) database hierarchies. For a comparison, saliva microbial metagenomes from the Human Microbiome Project (HMP) were also analyzed. These analyses identified that functions encoded and expressed by CF microbes were significantly enriched for amino acid catabolism, folate biosynthesis, and lipoic acid biosynthesis. The data indicate that the community uses oxidative phosphorylation as a major energy source but that terminal electron acceptors were diverse. Nitrate reduction was the most abundant anaerobic respiratory pathway, and genes for nitrate reductase were largely assigned to Pseudomonas and Rothia. Although many reductive pathways of the nitrogen cycle were present, the cycle was incomplete, because the oxidative pathways were absent. Due to the abundant amino acid catabolism and incomplete nitrogen cycle, the CF microbial community appears to accumulate ammonia. This finding was verified experimentally using a CF bronchiole culture model system. The data also revealed abundant sensing and transport of iron, ammonium, zinc, and other metals along with a low-oxygen environment. This study reveals the core biochemistry and physiology of the CF microbiome. The cystic fibrosis (CF) microbial community is complex and adapts to the environmental conditions of the lung over the lifetime of a CF patient. This analysis illustrates the core functions of the CF microbial community in the context of CF lung biochemistry. There are many studies of the metabolism and physiology of individual microbes within the CF lung, but none that collectively analyze data from the whole microbiome. Understanding the core metabolism of microbes that inhabit the CF lung can provide new targets for novel therapies. The fundamental processes that CF pathogens rely on for survival may represent an Achilles heel for this pathogenic community. Novel therapies that are designed to disrupt understudied survival strategies of the CF microbial community may succeed against otherwise untreatable or antibiotic-resistant microbes.
Collapse
|
39
|
Secretion properties, clearance, and therapy in airway disease. TRANSLATIONAL RESPIRATORY MEDICINE 2014; 2:6. [PMID: 25505698 PMCID: PMC4215824 DOI: 10.1186/2213-0802-2-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 02/19/2014] [Indexed: 01/26/2023]
Abstract
Chronic airway diseases like cystic fibrosis, chronic bronchitis, asthma, diffuse panbronchiolitis, and bronchiectasis are all associated with chronic inflammation. The airway mucosa responds to infection and inflammation in part by surface mucous (goblet) cell and submucosal gland hyperplasia and hypertrophy with mucus hypersecretion. Products of inflammation including neutrophil derived DNA and filamentous actin, effete cells, bacteria, and cell debris all contribute to mucus purulence and, when this is expectorated it is called sputum. Mucus is usually cleared by ciliary movement, and sputum is cleared by cough. These airway diseases each are associated with the production of mucus and sputum with characteristic composition, polymer structure, and biophysical properties. These properties change with the progress of the disease making it possible to use sputum analysis to identify the potential cause and severity of airway diseases. This information has also been important for the development of effective mucoactive therapy to promote airway hygiene.
Collapse
|
40
|
Abstract
As a student I recall being told that half of what we would learn in medical school would be proven to be wrong. The challenges were to identify the incorrect half and, often more challenging, be willing to give up our entrenched ideas. Myths have been defined as traditional concepts or practice with no basis in fact. A misunderstanding is a mistaken approach or incomplete knowledge that can be resolved with better evidence, while firmly established misunderstandings can become dogma; a point of view put forth as authoritative without basis in fact. In this paper, I explore a number of myths, mistakes, and dogma related to cystic fibrosis disease and care. Many of these are myths that have long been vanquished and even forgotten, while others are controversial. In the future, many things taken as either fact or "clinical experience" today will be proven wrong. Let us examine these myths with an open mind and willingness to change our beliefs when justified.
Collapse
Affiliation(s)
- Bruce K Rubin
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, U.S.A..
| |
Collapse
|
41
|
Reassessment of the importance of mucins in determining sputum properties in cystic fibrosis. J Cyst Fibros 2013; 13:260-6. [PMID: 24332705 PMCID: PMC3994278 DOI: 10.1016/j.jcf.2013.11.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/11/2013] [Accepted: 11/12/2013] [Indexed: 12/21/2022]
Abstract
Background There is conflicting evidence about the importance of airway mucins (MUC5AC and MUC5B) in determining physical properties of sputum in cystic fibrosis (CF). We studied the effects of endogenous degradation of mucins on CF sputum elasticity and apparent mucin concentrations. Methods Elastic shear moduli (G′) and mucin concentrations in sputum of 12 CF patients were measured before and after incubation at 37 °C for 60 min. Results G′ fell from a median of 5.98 to 4.70 Pa (p = 0.01). There were significant falls in MUC5AC (8.2 to 5.2 μg/ml, p = 0.02) and MUC5B (17.3 to 12.5 μg/ml, p = 0.02) over the same period, and associated decrease in molecular weight and size. Conclusions Sputum is not inert and degradation reduces apparent mucin concentrations and sputum elasticity. Even if care is taken to process samples rapidly, sputum may therefore differ from secretions retained in airways. Previous studies may have underestimated the role of mucins in CF sputum.
Collapse
|
42
|
|
43
|
Button B, Okada SF, Frederick CB, Thelin WR, Boucher RC. Mechanosensitive ATP release maintains proper mucus hydration of airways. Sci Signal 2013; 6:ra46. [PMID: 23757023 DOI: 10.1126/scisignal.2003755] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The clearance of mucus from the airways protects the lungs from inhaled noxious and infectious materials. Proper hydration of the mucus layer enables efficient mucus clearance through beating of cilia on airway epithelial cells, and reduced clearance of excessively concentrated mucus occurs in patients with chronic obstructive pulmonary disease and cystic fibrosis. Key steps in the mucus transport process are airway epithelia sensing and responding to changes in mucus hydration. We reported that extracellular adenosine triphosphate (ATP) and adenosine were important luminal autocrine and paracrine signals that regulated the hydration of the surface of human airway epithelial cultures through their action on apical membrane purinoceptors. Mucus hydration in human airway epithelial cultures was sensed by an interaction between cilia and the overlying mucus layer: Changes in mechanical strain, proportional to mucus hydration, regulated ATP release rates, adjusting fluid secretion to optimize mucus layer hydration. This system provided a feedback mechanism by which airways maintained mucus hydration in an optimum range for cilia propulsion. Understanding how airway epithelia can sense and respond to changes in mucus properties helps us to understand how the mucus clearance system protects the airways in health and how it fails in lung diseases such as cystic fibrosis.
Collapse
Affiliation(s)
- Brian Button
- Cystic Fibrosis Research and Treatment Center, University of North Carolina, Chapel Hill, NC 27599-7248, USA.
| | | | | | | | | |
Collapse
|
44
|
Hayashi N, Matsukawa M, Horinishi Y, Nakai K, Shoji A, Yoneko Y, Yoshida N, Minagawa S, Gotoh N. Interplay of flagellar motility and mucin degradation stimulates the uassociation of Pseudomonas aeruginosa with human epithelial colorectal adenocarcinoma (Caco-2) cells. J Infect Chemother 2013; 19:305-15. [DOI: 10.1007/s10156-013-0554-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 01/07/2013] [Indexed: 01/02/2023]
|
45
|
Hasnain SZ, Gallagher AL, Grencis RK, Thornton DJ. A new role for mucins in immunity: insights from gastrointestinal nematode infection. Int J Biochem Cell Biol 2012; 45:364-74. [PMID: 23107603 DOI: 10.1016/j.biocel.2012.10.011] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 10/21/2012] [Accepted: 10/23/2012] [Indexed: 12/26/2022]
Abstract
The body's mucosal surfaces are protected from pathogens and physical and chemical attack by the gel-like extracellular matrix, mucus. The framework of this barrier is provided by polymeric, gel-forming mucins. These enormous O-linked glycoproteins are synthesised, stored and secreted by goblet cells that are also the source of other protective factors. Immune regulation of goblet cells during the course of infection impacts on mucin production and properties and ultimately upon barrier function. The barrier function of mucins in protection of the host is well accepted as an important aspect of innate defence. However, it is becoming increasingly clear that mucins have a much more direct role in combating pathogens and parasites and are an important part of the coordinated immune response to infection. Of particular relevance to this review is the finding that mucins are essential anti-parasitic effector molecules. The current understanding of the roles of these multifunctional glycoproteins, and other goblet cell products, in mucosal defence against intestinal dwelling nematodes is discussed.
Collapse
Affiliation(s)
- Sumaira Z Hasnain
- Immunity, Infection and Inflammation Program, Mater Medical Research Institute, Mater Health Services and the University of Queensland, Brisbane, QLD 4029, Australia
| | | | | | | |
Collapse
|
46
|
Abstract
Mucus pathology in cystic fibrosis (CF) has been known for as long as the disease has been recognized and is sometimes called mucoviscidosis. The disease is marked by mucus hyperproduction and plugging in many organs, which are usually most fatal in the airways of CF patients, once the problem of meconium ileus at birth is resolved. After the CF gene, CFTR, was cloned and its protein product identified as a cAMP-regulated Cl(-) channel, causal mechanisms underlying the strong mucus phenotype of the disease became obscure. Here we focus on mucin genes and polymeric mucin glycoproteins, examining their regulation and potential relationships to a dysfunctional cystic fibrosis transmembrane conductance regulator (CFTR). Detailed examination of CFTR expression in organs and different cell types indicates that changes in CFTR expression do not always correlate with the severity of CF disease or mucus accumulation. Thus, the mucus hyperproduction that typifies CF does not appear to be a direct cause of a defective CFTR but, rather, to be a downstream consequence. In organs like the lung, up-regulation of mucin gene expression by inflammation results from chronic infection; however, in other instances and organs, the inflammation may have a non-infectious origin. The mucus plugging phenotype of the β-subunit of the epithelial Na(+) channel (βENaC)-overexpressing mouse is proving to be an archetypal example of this kind of inflammation, with a dehydrated airway surface/concentrated mucus gel apparently providing the inflammatory stimulus. Data indicate that the luminal HCO(3)(-) deficiency recently described for CF epithelia may also provide such a stimulus, perhaps by causing a mal-maturation of mucins as they are released onto luminal surfaces. In any event, the path between CFTR dysfunction and mucus hyperproduction has proven tortuous, and its unraveling continues to offer its own twists and turns, along with fascinating glimpses into biology.
Collapse
Affiliation(s)
- Silvia M Kreda
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina, Chapel Hill, NC 27517-7248, USA
| | | | | |
Collapse
|
47
|
Haley CL, Colmer-Hamood JA, Hamood AN. Characterization of biofilm-like structures formed by Pseudomonas aeruginosa in a synthetic mucus medium. BMC Microbiol 2012; 12:181. [PMID: 22900764 PMCID: PMC3494610 DOI: 10.1186/1471-2180-12-181] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 08/06/2012] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The accumulation of thick stagnant mucus provides a suitable environment for the growth of Pseudomonas aeruginosa and Staphylococcus aureus within the lung alveoli of cystic fibrosis (CF) patients. These infections cause significant lung damage, leading to respiratory failure and death. In an artificial mucin containing medium ASM+, P. aeruginosa forms structures that resemble typical biofilms but are not attached to any surface. We refer to these structures as biofilm like structures (BLS). Using ASM+ in a static microtiter plate culture system, we examined the roles of mucin, extracellular DNA, environmental oxygen (EO2), and quorum sensing (QS) in the development of biofilm-like structures (BLS) by P. aeruginosa; and the effect of EO2 and P. aeruginosa on S. aureus BLS. RESULTS Under 20% EO2, P. aeruginosa strain PAO1 produced BLS that resemble typical biofilms but are confined to the ASM+ and not attached to the surface. Levels of mucin and extracellular DNA within the ASM+ were optimized to produce robust well developed BLS. At 10% EO2, PAO1 produced thicker, more developed BLS, while under 0% EO2, BLS production was diminished. In contrast, the S. aureus strain AH133 produced well-developed BLS only under 20% EO2. In PAO1, loss of the QS system genes rhlI and rhlR affected the formation of BLS in ASM+ in terms of both structure and architecture. Whether co-inoculated into ASM+ with AH133, or added to established AH133 BLS, PAO1 eliminated AH133 within 48-56 h. CONCLUSIONS The thick, viscous ASM+, which contains mucin and extracellular DNA levels similar to those found in the CF lung, supports the formation of biofilm-like structures similar to the aggregates described within CF airways. Alterations in environmental conditions or in the QS genes of P. aeruginosa, as occurs naturally during the progression of CF lung infection, affect the architecture and quantitative structural features of these BLS. Thus, ASM+ provides an in vitro medium in which the effect of changing levels of substances produced by the host and the bacteria can be analyzed to determine the effect on such structures and on the susceptibility of the bacteria within the BLS to various treatments.
Collapse
Affiliation(s)
- Cecily L Haley
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, School of Medicine, Lubbock TX, USA
| | | | | |
Collapse
|
48
|
Callaghan M, McClean S. Bacterial host interactions in cystic fibrosis. Curr Opin Microbiol 2012; 15:71-7. [DOI: 10.1016/j.mib.2011.11.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 11/02/2011] [Accepted: 11/02/2011] [Indexed: 01/01/2023]
|
49
|
Papayannopoulos V, Staab D, Zychlinsky A. Neutrophil elastase enhances sputum solubilization in cystic fibrosis patients receiving DNase therapy. PLoS One 2011; 6:e28526. [PMID: 22174830 PMCID: PMC3235130 DOI: 10.1371/journal.pone.0028526] [Citation(s) in RCA: 187] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 11/09/2011] [Indexed: 01/27/2023] Open
Abstract
Cystic fibrosis patients suffer from chronic lung infection and inflammation due to the secretion of viscous sputum. Sputum viscosity is caused by extracellular DNA, some of which originates from the release of neutrophil extracellular traps (NETs). During NET formation neutrophil elastase (NE) partially processes histones to decondense chromatin. NE is abundant in CF sputum and is thought to contribute to tissue damage. Exogenous nucleases are a palliative treatment in CF as they promote sputum solubilization. We show that in a process reminiscent of NET formation, NE enhances sputum solubilization by cleaving histones to enhance the access of exogenous nucleases to DNA. In addition, we find that in Cf sputum NE is predominantly bound to DNA, which is known to downregulate its proteolytic activity and may restrict host tissue damage. The beneficial role of NE in CF sputum solubilization may have important implications for the development of CF therapies targeting NE.
Collapse
Affiliation(s)
- Venizelos Papayannopoulos
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Berlin, Germany
- * E-mail:
| | - Doris Staab
- Clinic for Pediatric Pneumology and Immunology, Christiane Herzog Cystic Fibrosis Center, Charité Medical School, Berlin, Germany
| | - Arturo Zychlinsky
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
50
|
KynR, a Lrp/AsnC-type transcriptional regulator, directly controls the kynurenine pathway in Pseudomonas aeruginosa. J Bacteriol 2011; 193:6567-75. [PMID: 21965577 DOI: 10.1128/jb.05803-11] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The opportunistic pathogen Pseudomonas aeruginosa can utilize a variety of carbon sources and produces many secondary metabolites to help survive harsh environments. P. aeruginosa is part of a small group of bacteria that use the kynurenine pathway to catabolize tryptophan. Through the kynurenine pathway, tryptophan is broken down into anthranilate, which is further degraded into tricarboxylic acid cycle intermediates or utilized to make numerous aromatic compounds, including the Pseudomonas quinolone signal (PQS). We have previously shown that the kynurenine pathway is a critical source of anthranilate for PQS synthesis and that the kynurenine pathway genes (kynA and kynBU) are upregulated in the presence of kynurenine. A putative Lrp/AsnC-type transcriptional regulator (gene PA2082, here called kynR), is divergently transcribed from the kynBU operon and is highly conserved in gram-negative bacteria that harbor the kynurenine pathway. We show that a mutation in kynR renders P. aeruginosa unable to utilize L-tryptophan as a sole carbon source and decreases PQS production. In addition, we found that the increase of kynA and kynB transcriptional activity in response to kynurenine was completely abolished in a kynR mutant, further indicating that KynR mediates the kynurenine-dependent expression of the kynurenine pathway genes. Finally, we found that purified KynR specifically bound the kynA promoter in the presence of kynurenine and bound the kynB promoter in the absence or presence of kynurenine. Taken together, our data show that KynR directly regulates the kynurenine pathway genes.
Collapse
|