1
|
Zhao Y, Xiong M, Ho K, Rao Y, Huang Y, Cao J, Yue Y, Wang J, Wen G, Li J. Bioaerosol emission and exposure risk from a wastewater treatment plant in winter and spring. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 287:117294. [PMID: 39504877 DOI: 10.1016/j.ecoenv.2024.117294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/02/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
The potential health risks posed by bioaerosols containing pathogens originating from wastewater treatment plants (WWTPs) have gaining intensive attention. This study designated sampling locations within a WWTP situated in Xi'an, a major city in northwest China. The airborne bacterial concentration, taxonomic composition, and the associated health risks were analyzed in the aeration tanks with bottom microporous aeration system. The Anaerobic-Anoxic-Oxic (AAO) tank emitted significantly higher culturable bacteria (1.58×104 CFU m-3 in spring, 6.69×103 CFU m-3 in winter) compared to Double-ditch (DE) oxidation ditch and aerated grit chamber (AGC) chamber, aligning with 16S rDNA quantification results. The bacterial concentrations are higher in spring than that in winter, with the AAO tank posing the highest exposure risk during the spring season. The dominant genera in the air samples include Cutibacterium, Lawsonella, Acinetobacter, Pseudomonas, and Aeromonas. Among the identified genus, 139 bacterial genera were identified as potential human pathogens like Neisseria, Moraxella, Haemophilus, Escherichia-Shigella and Streptococcus. These pathogens further elevate exposure risks from WWTP bioaerosols. This study provides relevant information on the seasonal health risk variations tied to bioaerosol emissions from diverse aeration tanks with bottom microporous aeration system in the mega city of northwest China, emphasizing the imperative to enhance the management and control measures for bioaerosols originating from the AAO tank.
Collapse
Affiliation(s)
- Yulei Zhao
- Key Laboratory of Aerosol Chemistry & Physics, State Key Laboratory of Loess and Quaternary Geology (SKLLQG), Institute of Earth Environment, Chinese Academy of Sciences (CAS), Xi'an 710061, PR China; Xi'an Institute for Innovative Earth Environment Research, Xi'an 710061, PR China
| | - Mingyu Xiong
- Key Laboratory of Aerosol Chemistry & Physics, State Key Laboratory of Loess and Quaternary Geology (SKLLQG), Institute of Earth Environment, Chinese Academy of Sciences (CAS), Xi'an 710061, PR China; Xi'an Institute for Innovative Earth Environment Research, Xi'an 710061, PR China
| | - Kinfai Ho
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong 999077, PR China
| | - Yongfang Rao
- Department of Environmental Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yu Huang
- Key Laboratory of Aerosol Chemistry & Physics, State Key Laboratory of Loess and Quaternary Geology (SKLLQG), Institute of Earth Environment, Chinese Academy of Sciences (CAS), Xi'an 710061, PR China.
| | - Junji Cao
- Institute of Atmospheric Physics, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Yang Yue
- Institute of Environmental Engineering, ETH Zurich, Zürich, Switzerland
| | - Jing Wang
- Institute of Environmental Engineering, ETH Zurich, Zürich, Switzerland; Laboratory of Advanced Analytical Technologies, Empa, Dübendorf, Switzerland
| | - Gang Wen
- Key Laboratory of Northwest Water Resource, Environment and Ecology, MOE, Xi'an University of Architecture and Technology, Xi'an 710055, PR China
| | - Juntang Li
- Research Centre for Occupation and Environment Medicine, Collaborative Innovation Centre for Medical Equipment, Key Laboratory of Biological Damage Effect and Protection, Luoyang 471031, PR China
| |
Collapse
|
2
|
Trusculescu AA, Ancusa VM, Pescaru CC, Wellmann N, Fira-Mladinescu C, Oancea CI, Fira-Mladinescu O. A Multifaceted Exploration of Status Asthmaticus: A Retrospective Analysis in a Romanian Hospital. J Clin Med 2024; 13:6615. [PMID: 39518753 PMCID: PMC11546779 DOI: 10.3390/jcm13216615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Status asthmaticus is a severe, life-threatening asthma exacerbation requiring urgent medical intervention. This study aims to examine its epidemiology in Timis County, Romania, over 11 years. Methods: A retrospective analysis was conducted using hospital records from 2013 to 2023, focusing on demographic, geospatial, and temporal distributions. Network analysis of the recorded comorbidities was used to identify phenotypic clusters among patients. Results: Females and older adults were disproportionately affected. Several triggers and geospatial patterns were identified. Five phenotypic clusters were determined: two in the T2-high endotype, two in T2-low, and a mixed one. Conclusions: The findings highlight the need for personalized asthma management strategies and public healthcare interventions in Timiș County, addressing specific demographic and geospatial factors. This study also provides a valuable reference for similar regions.
Collapse
Affiliation(s)
- Adriana Ana Trusculescu
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases (CRIPMRD), Pulmology University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy, Eftimie Murgu Square no. 2, 300041 Timisoara, Romania; (A.A.T.); (C.C.P.); (N.W.); (C.I.O.); (O.F.-M.)
- Pulmology University Clinic, Clinical Hospital of Infectious Diseases and Pneumophysiology Dr. Victor Babeș Timișoara, Gheorghe Adam Street, no. 13, 300310 Timisoara, Romania
| | - Versavia Maria Ancusa
- Department of Computer and Information Technology, Automation and Computers Faculty, “Politehnica” University of Timis, Vasile Pârvan Blvd, no. 2, 300223 Timisoara, Romania
| | - Camelia Corina Pescaru
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases (CRIPMRD), Pulmology University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy, Eftimie Murgu Square no. 2, 300041 Timisoara, Romania; (A.A.T.); (C.C.P.); (N.W.); (C.I.O.); (O.F.-M.)
- Pulmology University Clinic, Clinical Hospital of Infectious Diseases and Pneumophysiology Dr. Victor Babeș Timișoara, Gheorghe Adam Street, no. 13, 300310 Timisoara, Romania
| | - Norbert Wellmann
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases (CRIPMRD), Pulmology University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy, Eftimie Murgu Square no. 2, 300041 Timisoara, Romania; (A.A.T.); (C.C.P.); (N.W.); (C.I.O.); (O.F.-M.)
- Pulmology University Clinic, Clinical Hospital of Infectious Diseases and Pneumophysiology Dr. Victor Babeș Timișoara, Gheorghe Adam Street, no. 13, 300310 Timisoara, Romania
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Corneluta Fira-Mladinescu
- Hygiene Division, Department of Microbiology, “Victor Babes” University of Medicine and Pharmacy Timisoara, Victor Babes Street, no. 16, 300226 Timisoara, Romania;
- Center for Study in Preventive Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square no. 2, 300041 Timisoara, Romania
| | - Cristian Iulian Oancea
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases (CRIPMRD), Pulmology University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy, Eftimie Murgu Square no. 2, 300041 Timisoara, Romania; (A.A.T.); (C.C.P.); (N.W.); (C.I.O.); (O.F.-M.)
- Pulmology University Clinic, Clinical Hospital of Infectious Diseases and Pneumophysiology Dr. Victor Babeș Timișoara, Gheorghe Adam Street, no. 13, 300310 Timisoara, Romania
| | - Ovidiu Fira-Mladinescu
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases (CRIPMRD), Pulmology University Clinic, ‘Victor Babes’ University of Medicine and Pharmacy, Eftimie Murgu Square no. 2, 300041 Timisoara, Romania; (A.A.T.); (C.C.P.); (N.W.); (C.I.O.); (O.F.-M.)
- Pulmology University Clinic, Clinical Hospital of Infectious Diseases and Pneumophysiology Dr. Victor Babeș Timișoara, Gheorghe Adam Street, no. 13, 300310 Timisoara, Romania
| |
Collapse
|
3
|
Alqasmi M. Therapeutic Interventions for Pseudomonas Infections in Cystic Fibrosis Patients: A Review of Phase IV Trials. J Clin Med 2024; 13:6530. [PMID: 39518670 PMCID: PMC11547045 DOI: 10.3390/jcm13216530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Pseudomonas aeruginosa (Pa) poses a significant threat to individuals with cystic fibrosis (CF), as this bacterium is highly adaptable and resistant to antibiotics. While early-stage Pa infections can often be eradicated with aggressive antibiotic therapy, chronic infections are nearly impossible to eliminate and require treatments that focus on long-term bacterial suppression. Without such suppression, these persistent infections can severely damage the lungs, leading to serious complications and a reduced life expectancy for CF patients. Evidence for a specific treatment regimen for managing Pa infections in CF patients remains limited. This narrative review provides a detailed analysis of antimicrobial therapies assessed in completed phase IV trials, focusing on their safety and efficacy, especially with prolonged use. Key antibiotics, including tobramycin, colistin, meropenem, aztreonam, ceftolozane/tazobactam, ciprofloxacin, and azithromycin, are discussed, emphasizing their use, side effects, and delivery methods. Inhaled antibiotics are preferred for their targeted action and minimal side effects, while systemic antibiotics offer potency but carry risks like nephrotoxicity. The review also explores emerging treatments, such as phage therapy and antibiofilm agents, which show promise in managing chronic infections. Nonetheless, further research is necessary to enhance the safety and effectiveness of existing therapies while investigating new approaches for better long-term outcomes.
Collapse
Affiliation(s)
- Mohammed Alqasmi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
4
|
Higgs MG, Greenwald MA, Roca C, Macdonald JK, Sidders AE, Conlon BP, Wolfgang MC. Flagellar motility and the mucus environment influence aggregation mediated antibiotic tolerance of Pseudomonas aeruginosa in chronic lung infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620240. [PMID: 39484600 PMCID: PMC11527127 DOI: 10.1101/2024.10.25.620240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Pseudomonas aeruginosa frequently causes chronic lung infection in individuals with muco-obstructive airway diseases (MADs). Chronic P. aeruginosa infections are difficult to treat, primarily owing to antibiotic treatment failure, which is often observed in the absence of antimicrobial resistance. In MADs, P. aeruginosa forms biofilm-like aggregates within the luminal mucus. While the contribution of mucin hyperconcentration towards antibiotic tolerance has been described, the mechanism for mucin driven antibiotic tolerance and the influence of aggregates have not been fully elucidated. In this study, we investigated the contribution of flagellar motility towards aggregate formation as it relates to the diseased mucus environment. We found that loss of flagellar motility resulted in increased P. aeruginosa aggregation and tolerance to multiple classes of antibiotics. Further, we observed differential roles in antimicrobial tolerance of the motAB and motCD stators, which power the flagellum. Additionally, we found that control of fliC expression was important for aggregate formation and antibiotic tolerance as a strain constitutively expressing fliC was unable to form aggregates and was highly susceptible to treatment. Lastly, we demonstrate that neutrophil elastase, an abundant immune mediator and biomarker of chronic lung infection, promotes aggregation and antibiotic tolerance by impairing flagellar motility. Collectively, these results highlight the key role of flagellar motility in aggregate formation and antibiotic tolerance and deepens our understanding of how the MADs lung environment promotes antibiotic tolerance of P. aeruginosa.
Collapse
Affiliation(s)
- Matthew G. Higgs
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Matthew A. Greenwald
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Cristian Roca
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Jade K. Macdonald
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Ashelyn E. Sidders
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Brian P. Conlon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Matthew C. Wolfgang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
5
|
Guan J, Yao W, Zhang L, Xie H, Li L, Wen Y, Chen H, Huang Y, Wen J, Ou C, Liang C, Wang J, Zhang Q, Tao A, Yan J. Contribution of Pseudomonas aeruginosa - mediated club cell necroptosis to the bias of type 17 inflammation and steroid insensitivity in asthma. J Adv Res 2024:S2090-1232(24)00475-2. [PMID: 39442871 DOI: 10.1016/j.jare.2024.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION Opportunistic pathogen infection is one of the important inducements for asthma exacerbation. Pseudomonas aeruginosa (PA) is a kind of dominant pathogenic bacteria in the respiratory tract that is associated with severe asthma, but the underlying mechanisms still remains unclear. OBJECTIVES To examine the role of PA infection in the bias of the inflammatory endotype in asthma and its effect on the sensitivity to steroid therapy. METHODS An adjusted HDM (House Dust Mite) -induced asthma model with PA inoculation in the airway was utilized to mimic the process of opportunistic PA infection in asthma, focusing on the interaction between bacteria and epithelium. Dexamethasone administration in vivo was used to test the sensitivity to steroid therapy. RESULTS It was uncovered that PA could promote the loss of club cells in the necroptosis pattern through cellular CYP450 activation, leading to an imbalance of inflammatory response and steroid insensitivity. Club cell loss results in the activation of cellular E-cadherin/β-catenin axis in the rest of club cells for goblet metaplasia and mucus hypersecretion, as well as epithelial damage and GR downregulation for steroid resistance. For clinical applications, the necroptosis inhibitor Nec-1 can effectively relieve the pathological symptoms of asthma in vivo. Meanwhile, CCSP administration in the airway can regulate the pulmonary inflammation and restore the steroid sensitivity in asthma. CONCLUSION These experiments provide a novel mechanism of concurrent PA infection in asthma through club cell necroptosis and the pathological consequences. Nec-1 treatment and CCSP supplementation may be possible therapeutic strategies for asthma treatment.
Collapse
Affiliation(s)
- Jieying Guan
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China; Department of Obstetrics and Gynecology, Center for Reproductive Medicine, The First People's Hospital of Zhaoqing, Guangdong province, China
| | - Wenruo Yao
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Le Zhang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Huancheng Xie
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Linmei Li
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Yuhuan Wen
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Honglv Chen
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Yuyi Huang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Junjie Wen
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Changxing Ou
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Canyang Liang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Jing Wang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Qingling Zhang
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Ailin Tao
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China.
| | - Jie Yan
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Canning JS, Laucirica DR, Ling KM, Nicol MP, Stick SM, Kicic A. Phage therapy to treat cystic fibrosis Burkholderia cepacia complex lung infections: perspectives and challenges. Front Microbiol 2024; 15:1476041. [PMID: 39493847 PMCID: PMC11527634 DOI: 10.3389/fmicb.2024.1476041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024] Open
Abstract
Burkholderia cepacia complex is a cause of serious lung infections in people with cystic fibrosis, exhibiting extremely high levels of antimicrobial resistance. These infections are difficult to treat and are associated with high morbidity and mortality. With a notable lack of new antibiotic classes currently in development, exploring alternative antimicrobial strategies for Burkholderia cepacia complex is crucial. One potential alternative seeing renewed interest is the use of bacteriophage (phage) therapy. This review summarises what is currently known about Burkholderia cepacia complex in cystic fibrosis, as well as challenges and insights for using phages to treat Burkholderia cepacia complex lung infections.
Collapse
Affiliation(s)
- Jack S. Canning
- Division of Infection and Immunity, School of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Western Australia, Nedlands, WA, Australia
- Wal-Yan Respiratory Research Centre, The Kids Research Institute Australia, The University of Western Australia, Nedlands, WA, Australia
| | - Daniel R. Laucirica
- Wal-Yan Respiratory Research Centre, The Kids Research Institute Australia, The University of Western Australia, Nedlands, WA, Australia
| | - Kak-Ming Ling
- Wal-Yan Respiratory Research Centre, The Kids Research Institute Australia, The University of Western Australia, Nedlands, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
| | - Mark P. Nicol
- Division of Infection and Immunity, School of Biomedical Sciences, Marshall Centre, University of Western Australia, Perth, WA, Australia
| | - Stephen M. Stick
- Wal-Yan Respiratory Research Centre, The Kids Research Institute Australia, The University of Western Australia, Nedlands, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
- School of Medicine and Pharmacology, Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| | - Anthony Kicic
- Wal-Yan Respiratory Research Centre, The Kids Research Institute Australia, The University of Western Australia, Nedlands, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
- School of Medicine and Pharmacology, Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| |
Collapse
|
7
|
Vaillancourt M, Aguilar D, Fernandes SE, Jorth PA. A chronic Pseudomonas aeruginosa mouse lung infection modeling the pathophysiology and inflammation of human cystic fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617039. [PMID: 39416002 PMCID: PMC11482824 DOI: 10.1101/2024.10.07.617039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Investigation of chronic cystic fibrosis (CF) lung infections has been limited by a lack of murine models that reproduce obstructive lung pathology, chronicity of bacterial infections, and complex inflammation in human CF lung pathology. Three different approaches have been used separately to address these limitations, including using transgenic Scnn1b-Tg mice overexpressing a lung epithelial sodium channel to mimic the mucus-rich and hyperinflammatory CF lung environment, using synthetic CF sputum medium (SCFM) in an acute infection to induce bacterial phenotypes consistent with human CF, or using agar beads to promote chronic infections. Here, we combine these three models to establish a chronic Pseudomonas aeruginosa lung infection model using SCFM agar beads and Scnn1b-Tg mice (SCFM-Tg-mice) to recapitulate nutrients, mucus, and inflammation characteristic of the human CF lung environment. Like people with CF, SCFM-Tg-mice failed to clear bacterial infections. Lung function measurements showed that infected SCFM-Tg-mice had decreased inspiratory capacity and compliance, elevated airway resistance, and significantly reduced FVC and FEV0.1. Using spectral flow cytometry and multiplex cytokine arrays we show that, like people with CF, SCFM-Tg-mice developed inflammation characterized by eosinophil infiltration and Th2 lymphocytic cytokine responses. Chronically infected SCFM-Tg-mice developed an exacerbated mix of innate and Th1, Th2, and Th17-mediated inflammation, causing higher lung cellular damage, and elevated numbers of unusual Siglec F+ neutrophils. Thus, SCFM-Tg-mice represents a powerful tool to investigate bacterial pathogenesis and potential treatments for chronic CF lung infections and reveal a potential role for Siglec F+ neutrophils in CF inflammation.
Collapse
Affiliation(s)
- Mylene Vaillancourt
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Diane Aguilar
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sheryl E. Fernandes
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Peter A. Jorth
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
8
|
Chen L, A Hoefel G, Pathinayake PS, Reid A, Pillar AL, Kelly C, Tan H, Ali A, Kim RY, Hansbro PM, Brody SL, Foster PS, Horvat JC, Riveros C, Awatade N, Wark PAB, Kaiko GE. Inflammation-induced loss of CFTR-expressing airway ionocytes in non-eosinophilic asthma. Respirology 2024. [PMID: 39358991 DOI: 10.1111/resp.14833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND AND OBJECTIVE Severe asthma is a heterogeneous disease with subtype classification according to dominant airway infiltrates, including eosinophilic (Type 2 high), or non-eosinophilic asthma. Non-eosinophilic asthma is further divided into paucigranulocytic or neutrophilic asthma characterized by elevated neutrophils, and mixed Type 1 and Type 17 cytokines in the airways. Severe non-eosinophilic asthma has few effective treatments and many patients do not qualify for biologic therapies. The cystic fibrosis transmembrane conductance regulator (CFTR) is dysregulated in multiple respiratory diseases including cystic fibrosis and chronic obstructive pulmonary disease and has proven a valuable therapeutic target. We hypothesized that the CFTR may also play a role in non-eosinophilic asthma. METHODS Patient-derived human bronchial epithelial cells (hBECs) were isolated and differentiated at the air-liquid interface. Single cell RNA-sequencing (scRNAseq) was used to identify epithelial cell subtypes and transcriptional activity. Ion transport was investigated with Ussing chambers and immunofluorescent quantification of ionocyte abundance in human airway epithelial cells and murine models of asthma. RESULTS We identified that hBECs from patients with non-eosinophilic asthma had reduced CFTR function, and did not differentiate into CFTR-expressing ionocytes compared to those from eosinophilic asthma or healthy donors. Similarly, ionocytes were also diminished in the airways of a murine model of neutrophilic-dominant but not eosinophilic asthma. Treatment of hBECs from healthy donors with a neutrophilic asthma-like inflammatory cytokine mixture led to a reduction in ionocytes. CONCLUSION Inflammation-induced loss of CFTR-expressing ionocytes in airway cells from non-eosinophilic asthma may represent a key feature of disease pathogenesis and a novel drug target.
Collapse
Affiliation(s)
- Ling Chen
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Gabriela A Hoefel
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Prabuddha S Pathinayake
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Andrew Reid
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Amber L Pillar
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Coady Kelly
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - HuiYing Tan
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Ayesha Ali
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Richard Y Kim
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Philip M Hansbro
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, New South Wales, Australia
| | - Steven L Brody
- Department of Medicine, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - Paul S Foster
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Jay C Horvat
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Carlos Riveros
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Nikhil Awatade
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Peter A B Wark
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton, New South Wales, Australia
- Department of Respiratory Medicine, Alfred Health, Melbourne, Victoria, Australia
| | - Gerard E Kaiko
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
9
|
Glen KA, Lamont IL. Penicillin-binding protein 3 sequence variations reduce susceptibility of Pseudomonas aeruginosa to β-lactams but inhibit cell division. J Antimicrob Chemother 2024; 79:2170-2178. [PMID: 39001778 PMCID: PMC11368433 DOI: 10.1093/jac/dkae203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 06/03/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND β-lactam antibiotics, which inhibit penicillin-binding protein 3 (PBP3) that is required for cell division, play a key role in treating P. aeruginosa infections. Some sequence variations in PBP3 have been associated with β-lactam resistance but the effects of variations on antibiotic susceptibility and on cell division have not been quantified. Antibiotic efflux can also reduce susceptibility. OBJECTIVES To quantify the effects of PBP3 variations on β-lactam susceptibility and cell morphology in P. aeruginosa. METHODS Nineteen PBP3 variants were expressed from a plasmid in the reference strain P. aeruginosa PAO1 and genome engineering was used to construct five mutants expressing PBP3 variants from the chromosome. The effects of the variations on β-lactam minimum inhibitory concentration (MIC) and cell morphology were measured. RESULTS Some PBP3 variations reduced susceptibility to a variety of β-lactam antibiotics including meropenem, ceftazidime, cefepime and ticarcillin with different variations affecting different antibiotics. None of the tested variations reduced susceptibility to imipenem or piperacillin. Antibiotic susceptibility was further reduced when PBP3 variants were expressed in mutant bacteria overexpressing the MexAB-OprM efflux pump, with some variations conferring clinical levels of resistance. Some PBP3 variations, and sub-MIC levels of β-lactams, reduced bacterial growth rates and inhibited cell division, causing elongated cells. CONCLUSIONS PBP3 variations in P. aeruginosa can increase the MIC of multiple β-lactam antibiotics, although not imipenem or piperacillin. PBP3 variations, or the presence of sub-lethal levels of β-lactams, result in elongated cells indicating that variations reduce the activity of PBP3 and may reduce bacterial fitness.
Collapse
Affiliation(s)
- Karl A Glen
- Department of Biochemistry, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Iain L Lamont
- Department of Biochemistry, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| |
Collapse
|
10
|
Frigoli M, Lowdon JW, Donetti N, Crapnell RD, Banks CE, Cleij TJ, Diliën H, Eersels K, van Grinsven B. Electrochemical Detection of Pseudomonas aeruginosa Quorum Sensing Molecule ( S)- N-Butyryl Homoserine Lactone Using Molecularly Imprinted Polymers. ACS OMEGA 2024; 9:36411-36420. [PMID: 39220512 PMCID: PMC11359617 DOI: 10.1021/acsomega.4c03970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/09/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Pseudomonas aeruginosa is a multidrug-resistant Gram-negative bacterium that poses a significant threat to public health, necessitating rapid and on-site detection methods for rapid recognition. The goal of the project is therefore to indirectly detect the presence of P. aeruginosa in environmental water samples targeting one of its quorum-sensing molecules, namely, (S)-N-butyryl homoserine lactone (BHL). To this aim, molecularly imprinted polymers (MIPs) were synthesized via bulk free-radical polymerization using BHL as a template molecule. The obtained MIP particles were immobilized onto screen-printed electrodes (MIP-SPEs), and the BHL rebinding was analyzed via electrochemical impedance spectroscopy (EIS). To study the specificity of the synthesized MIPs, isotherm curves were built after on-point rebinding analysis performed via LC-MS measurements for both MIPs and NIPs (nonimprinted polymers, used as a negative control), obtaining an imprinting factor (IF) of 2.8 (at C f = 0.4 mM). The MIP-SPEs were integrated into an electrochemical biosensor with a linear range of 1 × 101-1 × 103 nM and a limit of detection (LoD) of 31.78 ± 4.08 nM. Selectivity measurements were also performed after choosing specific interferent molecules, such as structural analogs and potential interferents, followed by on-point analysis performed in spiked tap water to prove the sensor's potential to detect the presence of the quorum-sensing molecule in environmentally related real-life samples.
Collapse
Affiliation(s)
- Margaux Frigoli
- Sensor
Engineering Department, Faculty of Science and Engineering, Maastricht University, P.O. Box 616, Maastricht 6200 MD, The Netherlands
| | - Joseph W. Lowdon
- Sensor
Engineering Department, Faculty of Science and Engineering, Maastricht University, P.O. Box 616, Maastricht 6200 MD, The Netherlands
| | - Nicolas Donetti
- Sensor
Engineering Department, Faculty of Science and Engineering, Maastricht University, P.O. Box 616, Maastricht 6200 MD, The Netherlands
| | - Robert D. Crapnell
- John
Dalton Building, Faculty of Science and Engineering, Manchester Metropolitan University, Chester Street, Manchester M1 5GD, U.K.
| | - Craig E. Banks
- John
Dalton Building, Faculty of Science and Engineering, Manchester Metropolitan University, Chester Street, Manchester M1 5GD, U.K.
| | - Thomas J. Cleij
- Sensor
Engineering Department, Faculty of Science and Engineering, Maastricht University, P.O. Box 616, Maastricht 6200 MD, The Netherlands
| | - Hanne Diliën
- Sensor
Engineering Department, Faculty of Science and Engineering, Maastricht University, P.O. Box 616, Maastricht 6200 MD, The Netherlands
| | - Kasper Eersels
- Sensor
Engineering Department, Faculty of Science and Engineering, Maastricht University, P.O. Box 616, Maastricht 6200 MD, The Netherlands
| | - Bart van Grinsven
- Sensor
Engineering Department, Faculty of Science and Engineering, Maastricht University, P.O. Box 616, Maastricht 6200 MD, The Netherlands
| |
Collapse
|
11
|
Panickar A, Manoharan A, Anbarasu A, Ramaiah S. Respiratory tract infections: an update on the complexity of bacterial diversity, therapeutic interventions and breakthroughs. Arch Microbiol 2024; 206:382. [PMID: 39153075 DOI: 10.1007/s00203-024-04107-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
Respiratory tract infections (RTIs) have a significant impact on global health, especially among children and the elderly. The key bacterial pathogens Streptococcus pneumoniae, Haemophilus influenzae, Klebsiella pneumoniae, Staphylococcus aureus and non-fermenting Gram Negative bacteria such as Acinetobacter baumannii and Pseudomonas aeruginosa are most commonly associated with RTIs. These bacterial pathogens have evolved a diverse array of resistance mechanisms through horizontal gene transfer, often mediated by mobile genetic elements and environmental acquisition. Treatment failures are primarily due to antimicrobial resistance and inadequate bacterial engagement, which necessitates the development of alternative treatment strategies. To overcome this, our review mainly focuses on different virulence mechanisms and their resulting pathogenicity, highlighting different therapeutic interventions to combat resistance. To prevent the antimicrobial resistance crisis, we also focused on leveraging the application of artificial intelligence and machine learning to manage RTIs. Integrative approaches combining mechanistic insights are crucial for addressing the global challenge of antimicrobial resistance in respiratory infections.
Collapse
Affiliation(s)
- Avani Panickar
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Anand Manoharan
- Infectious Diseases Medical and Scientific Affairs, GlaxoSmithKline (GSK), Worli, Maharashtra, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
12
|
Jantaruk P, Teerapo K, Charoenwutthikun S, Roytrakul S, Kunthalert D. Anti-Biofilm and Anti-Inflammatory Properties of the Truncated Analogs of the Scorpion Venom-Derived Peptide IsCT against Pseudomonas aeruginosa. Antibiotics (Basel) 2024; 13:775. [PMID: 39200075 PMCID: PMC11352108 DOI: 10.3390/antibiotics13080775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen in humans and a frequent cause of severe nosocomial infections and fatal infections in immunocompromised individuals. Its ability to form biofilms has been the main driving force behind its resistance to almost all conventional antibiotics, thereby limiting treatment efficacy. In an effort to discover novel therapeutic agents to fight P. aeruginosa-associated biofilm infections, the truncated analogs of scorpion venom-derived peptide IsCT were synthesized and their anti-biofilm properties were examined. Among the investigated peptides, the IsCT-Δ6-8 peptide evidently showed the most potential anti-P. aeruginosa biofilm activity and the effect was not due to bacterial growth inhibition. The IsCT-Δ6-8 peptide also exhibited inhibitory activity against the production of pyocyanin, an important virulence factor of P. aeruginosa. Furthermore, the IsCT-Δ6-8 peptide significantly suppressed the production of inflammatory mediators nitric oxide and interleukin-6 in P. aeruginosa LPS-induced macrophages. Due to its low cytotoxicity to mammalian cells, the IsCT-Δ6-8 peptide emerges as a promising candidate with significant anti-biofilm and anti-inflammatory properties. These findings highlight its potential application in treating P. aeruginosa-related biofilm infections.
Collapse
Affiliation(s)
- Pornpimon Jantaruk
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (P.J.); (K.T.); (S.C.)
| | - Kittitat Teerapo
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (P.J.); (K.T.); (S.C.)
| | - Supattra Charoenwutthikun
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (P.J.); (K.T.); (S.C.)
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Thailand Science Park, Pathumthani 12120, Thailand;
| | - Duangkamol Kunthalert
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (P.J.); (K.T.); (S.C.)
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| |
Collapse
|
13
|
Radhouani M, Starkl P. Adjuvant-independent airway sensitization and infection mouse models leading to allergic asthma. FRONTIERS IN ALLERGY 2024; 5:1423938. [PMID: 39157265 PMCID: PMC11327155 DOI: 10.3389/falgy.2024.1423938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/05/2024] [Indexed: 08/20/2024] Open
Abstract
Asthma is a chronic respiratory disease of global importance. Mouse models of allergic asthma have been instrumental in advancing research and novel therapeutic strategies for patients. The application of relevant allergens and physiological routes of exposure in such models has led to valuable insights into the complexities of asthma onset and development as well as key disease mechanisms. Furthermore, environmental microbial exposures and infections have been shown to play a fundamental part in asthma pathogenesis and alter disease outcome. In this review, we delve into physiological mouse models of allergic asthma and explore literature reports on most significant interplays between microbial infections and asthma development with relevance to human disease.
Collapse
Affiliation(s)
- Mariem Radhouani
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Philipp Starkl
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
14
|
Mudgil U, Khullar L, Chadha J, Prerna, Harjai K. Beyond antibiotics: Emerging antivirulence strategies to combat Pseudomonas aeruginosa in cystic fibrosis. Microb Pathog 2024; 193:106730. [PMID: 38851361 DOI: 10.1016/j.micpath.2024.106730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that poses a significant threat to individuals suffering from cystic fibrosis (CF). The pathogen is highly prevalent in CF individuals and is responsible for chronic infection, resulting in severe tissue damage and poor patient outcome. Prolonged antibiotic administration has led to the emergence of multidrug resistance in P. aeruginosa. In this direction, antivirulence strategies achieving targeted inhibition of bacterial virulence pathways, including quorum sensing, efflux pumps, lectins, and iron chelators, have been explored against CF isolates of P. aeruginosa. Hence, this review article presents a bird's eye view on the pulmonary infections involving P. aeruginosa in CF patients by laying emphasis on factors contributing to bacterial colonization, persistence, and disease progression along with the current line of therapeutics against P. aeruginosa in CF. We further collate scientific literature and discusses various antivirulence strategies that have been tested against P. aeruginosa isolates from CF patients.
Collapse
Affiliation(s)
- Umang Mudgil
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Lavanya Khullar
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Jatin Chadha
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Prerna
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Kusum Harjai
- Department of Microbiology, Panjab University, Chandigarh, India.
| |
Collapse
|
15
|
Hajdú G, Szathmári C, Sőti C. Modeling Host-Pathogen Interactions in C. elegans: Lessons Learned from Pseudomonas aeruginosa Infection. Int J Mol Sci 2024; 25:7034. [PMID: 39000143 PMCID: PMC11241598 DOI: 10.3390/ijms25137034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/17/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Infections, such as that by the multiresistant opportunistic bacterial pathogen Pseudomonas aeruginosa, may pose a serious health risk, especially on vulnerable patient populations. The nematode Caenorhabditis elegans provides a simple organismal model to investigate both pathogenic mechanisms and the emerging role of innate immunity in host protection. Here, we review the virulence and infection strategies of P. aeruginosa and host defenses of C. elegans. We summarize the recognition mechanisms of patterns of pathogenesis, including novel pathogen-associated molecular patterns and surveillance immunity of translation, mitochondria, and lysosome-related organelles. We also review the regulation of antimicrobial and behavioral defenses by the worm's neuroendocrine system. We focus on how discoveries in this rich field align with well-characterized evolutionary conserved protective pathways, as well as on potential crossovers to human pathogenesis and innate immune responses.
Collapse
Affiliation(s)
- Gábor Hajdú
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Csenge Szathmári
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Csaba Sőti
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| |
Collapse
|
16
|
Eklöf J, Alispahic IA, Armbruster K, Lapperre TS, Browatzki A, Overgaard RH, Harboe ZB, Janner J, Moberg M, Ulrik CS, Andreassen HF, Weinreich UM, Kjærgaard JL, Villadsen J, Fenlev CS, Jensen TT, Christensen CW, Bangsborg J, Ostergaard C, Ghathian KSA, Jordan A, Klausen TW, Nielsen TL, Wilcke T, Seersholm N, Sivapalan P, Jensen JUS. Systemic antibiotics for Pseudomonas aeruginosa infection in outpatients with non-hospitalised exacerbations of pre-existing lung diseases: a randomised clinical trial. Respir Res 2024; 25:236. [PMID: 38844921 PMCID: PMC11157704 DOI: 10.1186/s12931-024-02860-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND The effect of dual systemic antibiotic therapy against Pseudomonas aeruginosa in patients with pre-existing lung disease is unknown. To assess whether dual systemic antibiotics against P. aeruginosa in outpatients with COPD, non-cystic fibrosis (non-CF) bronchiectasis, or asthma can improve outcomes. METHODS Multicenter, randomised, open-label trial conducted at seven respiratory outpatient clinics in Denmark. Outpatients with COPD, non-CF bronchiectasis, or asthma with a current P. aeruginosa-positive lower respiratory tract culture (clinical routine samples obtained based on symptoms of exacerbation not requiring hospitalisation), regardless of prior P. aeruginosa-status, no current need for hospitalisation, and at least two moderate or one hospitalisation-requiring exacerbation within the last year were eligible. Patients were assigned 1:1 to 14 days of dual systemic anti-pseudomonal antibiotics or no antibiotic treatment. Primary outcome was time to prednisolone or antibiotic-requiring exacerbation or death from day 20 to day 365. RESULTS The trial was stopped prematurely based in lack of recruitment during the COVID-19 pandemic, this decision was endorsed by the Data and Safety Monitoring Board. Forty-nine outpatients were included in the study. There was a reduction in risk of the primary outcome in the antibiotic group compared to the control group (HR 0.51 (95%CI 0.27-0.96), p = 0.037). The incidence of admissions with exacerbation within one year was 1.1 (95%CI 0.6-1.7) in the dual antibiotic group vs. 2.9 (95%CI 1.3-4.5) in the control group, p = 0.037. CONCLUSIONS Use of dual systemic antibiotics for 14 days against P. aeruginosa in outpatients with chronic lung diseases and no judged need for hospitalisation, improved clinical outcomes markedly. The main limitation was the premature closure of the trial. TRIAL REGISTRATION ClinicalTrials.gov, NCT03262142, registration date 2017-08-25.
Collapse
Affiliation(s)
- Josefin Eklöf
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark.
| | - Imane Achir Alispahic
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Karin Armbruster
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Therese Sophie Lapperre
- Department of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital, Bispebjerg Frederiksberg, Denmark
- Department of Respiratory Medicine, Antwerp University Hospital, Antwerp, Belgium
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Andrea Browatzki
- Department of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital, North Zealand, Denmark
| | - Rikke Holmen Overgaard
- Department of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital, North Zealand, Denmark
| | - Zitta Barrella Harboe
- Department of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital, North Zealand, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Julie Janner
- Department of Respiratory Medicine, Copenhagen University Hospital, Hvidovre, Denmark
| | - Mia Moberg
- Department of Respiratory Medicine, Copenhagen University Hospital, Hvidovre, Denmark
| | | | - Helle Frost Andreassen
- Department of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital, Bispebjerg Frederiksberg, Denmark
| | - Ulla Møller Weinreich
- Department of Respiratory Medicine, Aalborg University Hospital and Department of Clinical Medicine, Aalborg, Denmark
| | - Jakob Lyngby Kjærgaard
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Jenny Villadsen
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Camilla Sund Fenlev
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | | | | | - Jette Bangsborg
- Department of Clinical Microbiology, Copenhagen University Hospital, Herlev, Denmark
| | - Christian Ostergaard
- Department of Clinical Microbiology, Copenhagen University Hospital, Hvidovre, Denmark
| | | | - Alexander Jordan
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Tobias Wirenfeldt Klausen
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Thyge Lynghøj Nielsen
- Department of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital, North Zealand, Denmark
| | - Torgny Wilcke
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Niels Seersholm
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
| | - Pradeesh Sivapalan
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jens-Ulrik Stæhr Jensen
- Department of Internal Medicine, Herlev Gentofte University Hospital, Section of Respiratory Medicine, Copenhagen University Hospital, Herlev Gentofte, Hellerup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
Ammazzalorso A, Granese A, De Filippis B. Recent trends and challenges to overcome Pseudomonas aeruginosa infections. Expert Opin Ther Pat 2024; 34:493-509. [PMID: 38683024 DOI: 10.1080/13543776.2024.2348602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
INTRODUCTION Pseudomonas aeruginosa (PA) is a Gram-negative bacterium that can cause a wide range of severe infections in immunocompromised patients. The most difficult challenge is due to its ability to rapidly develop multi drug-resistance. New strategies are urgently required to improve the outcome of patients with PA infections. The present patent review highlights the new molecules acting on different targets involved in the antibiotic resistance. AREA COVERED This review offers an insight into new potential PA treatment disclosed in patent literature. From a broad search of documents claiming new PA inhibitors, we selected and summarized molecules that showed in vitro and in vivo activity against PA spp. in the period 2020 and 2023. We collected the search results basing on the targets explored. EXPERT OPINION This review examined the main patented compounds published in the last three years, with regard to the structural novelty and the identification of innovative targets. The main areas of antibiotic resistance have been explored. The compounds are structurally unrelated to earlier antibiotics, characterized by a medium-high molecular weight and the presence of heterocycle rings. Peptides and antibodies have also been reported as potential alternatives to chemical treatment, hereby expanding the therapeutic possibilities in this field.
Collapse
Affiliation(s)
| | - Arianna Granese
- Department of Drug Chemistry and Technology, "Sapienza" University of Rome, Rome, Italy
| | | |
Collapse
|
18
|
García-Villada L, Degtyareva NP, Brooks AM, Goldberg JB, Doetsch PW. A role for the stringent response in ciprofloxacin resistance in Pseudomonas aeruginosa. Sci Rep 2024; 14:8598. [PMID: 38615146 PMCID: PMC11016087 DOI: 10.1038/s41598-024-59188-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/08/2024] [Indexed: 04/15/2024] Open
Abstract
Pseudomonas aeruginosa is a major cause of nosocomial infections and the leading cause of chronic lung infections in cystic fibrosis and chronic obstructive pulmonary disease patients. Antibiotic treatment remains challenging because P. aeruginosa is resistant to high concentrations of antibiotics and has a remarkable ability to acquire mutations conferring resistance to multiple groups of antimicrobial agents. Here we report that when P. aeruginosa is plated on ciprofloxacin (cipro) plates, the majority of cipro-resistant (ciproR) colonies observed at and after 48 h of incubation carry mutations in genes related to the Stringent Response (SR). Mutations in one of the major SR components, spoT, were present in approximately 40% of the ciproR isolates. Compared to the wild-type strain, most of these isolates had decreased growth rate, longer lag phase and altered intracellular ppGpp content. Also, 75% of all sequenced mutations were insertions and deletions, with short deletions being the most frequently occurring mutation type. We present evidence that most of the observed mutations are induced on the selective plates in a subpopulation of cells that are not instantly killed by cipro. Our results suggests that the SR may be an important contributor to antibiotic resistance acquisition in P. aeruginosa.
Collapse
Affiliation(s)
| | | | - Ashley M Brooks
- Integrative Bioinformatics, Biostatistics and Computational Biology Branch, NIEHS, Durham, NC, USA
| | - Joanna B Goldberg
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Paul W Doetsch
- Genomic Integrity and Structural Biology Laboratory, NIEHS, Durham, NC, USA.
| |
Collapse
|
19
|
Elfadadny A, Ragab RF, AlHarbi M, Badshah F, Ibáñez-Arancibia E, Farag A, Hendawy AO, De los Ríos-Escalante PR, Aboubakr M, Zakai SA, Nageeb WM. Antimicrobial resistance of Pseudomonas aeruginosa: navigating clinical impacts, current resistance trends, and innovations in breaking therapies. Front Microbiol 2024; 15:1374466. [PMID: 38646632 PMCID: PMC11026690 DOI: 10.3389/fmicb.2024.1374466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/05/2024] [Indexed: 04/23/2024] Open
Abstract
Pseudomonas aeruginosa, a Gram-negative bacterium, is recognized for its adaptability and opportunistic nature. It poses a substantial challenge in clinical settings due to its complicated antibiotic resistance mechanisms, biofilm formation, and capacity for persistent infections in both animal and human hosts. Recent studies revealed a potential zoonotic transmission of P. aeruginosa between animals, the environment, and human populations which highlights awareness of this microbe. Implementation of the One Health approach, which underscores the connection between human, animal, and environmental health, we aim to offer a comprehensive perspective on the current landscape of P. aeruginosa management. This review presents innovative strategies designed to counteract P. aeruginosa infections. Traditional antibiotics, while effective in many cases, are increasingly compromised by the development of multidrug-resistant strains. Non-antibiotic avenues, such as quorum sensing inhibition, phage therapy, and nanoparticle-based treatments, are emerging as promising alternatives. However, their clinical application encounters obstacles like cost, side effects, and safety concerns. Effectively addressing P. aeruginosa infections necessitates persistent research efforts, advancements in clinical development, and a comprehension of host-pathogen interactions to deal with this resilient pathogen.
Collapse
Affiliation(s)
- Ahmed Elfadadny
- Laboratory of Internal Medicine, Cooperative Division of Veterinary Sciences, Tokyo University of Agriculture and Technology, Fuchu, Japan
- Department of Internal Medicine, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Rokaia F. Ragab
- Laboratory of Internal Medicine, Cooperative Division of Veterinary Sciences, Tokyo University of Agriculture and Technology, Fuchu, Japan
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Maha AlHarbi
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Farhad Badshah
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Science, Beijing, China
| | - Eliana Ibáñez-Arancibia
- PhD Program in Sciences Mentioning Applied Molecular and Cell Biology, La Frontera University, Temuco, Chile
- Laboratory of Engineering, Biotechnology and Applied Biochemistry – LIBBA, Department of Chemical Engineering, Faculty of Engineering and Science, La Frontera University, Temuco, Chile
- Department of Biological and Chemical Sciences, Faculty of Natural Resources, Catholic University of Temuco, Temuco, Chile
| | - Ahmed Farag
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Amin Omar Hendawy
- Department of Animal and Poultry Production, Faculty of Agriculture, Damanhour University, Damanhour, Egypt
| | - Patricio R. De los Ríos-Escalante
- Department of Biological and Chemical Sciences, Faculty of Natural Resources, Catholic University of Temuco, Temuco, Chile
- Nucleus of Environmental Sciences, Faculty of Natural Resources, Catholic University of Temuco, Temuco, Chile
| | - Mohamed Aboubakr
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Qaliobiya, Egypt
| | - Shadi A. Zakai
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Wedad M. Nageeb
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
20
|
Correnti S, Preianò M, Gamboni F, Stephenson D, Pelaia C, Pelaia G, Savino R, D'Alessandro A, Terracciano R. An integrated metabo-lipidomics profile of induced sputum for the identification of novel biomarkers in the differential diagnosis of asthma and COPD. J Transl Med 2024; 22:301. [PMID: 38521955 PMCID: PMC10960495 DOI: 10.1186/s12967-024-05100-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/15/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Due to their complexity and to the presence of common clinical features, differentiation between asthma and chronic obstructive pulmonary disease (COPD) can be a challenging task, complicated in such cases also by asthma-COPD overlap syndrome. The distinct immune/inflammatory and structural substrates of COPD and asthma are responsible for significant differences in the responses to standard pharmacologic treatments. Therefore, an accurate diagnosis is of central relevance to assure the appropriate therapeutic intervention in order to achieve safe and effective patient care. Induced sputum (IS) accurately mirrors inflammation in the airways, providing a more direct picture of lung cell metabolism in comparison to those specimen that reflect analytes in the systemic circulation. METHODS An integrated untargeted metabolomics and lipidomics analysis was performed in IS of asthmatic (n = 15) and COPD (n = 22) patients based on Ultra-High-Pressure Liquid Chromatography-Mass Spectrometry (UHPLC-MS) and UHPLC-tandem MS (UHPLC-MS/MS). Partial Least Squares-Discriminant Analysis (PLS-DA) was applied to resulting dataset. The analysis of main enriched metabolic pathways and the association of the preliminary metabolites/lipids pattern identified to clinical parameters of asthma/COPD differentiation were explored. Multivariate ROC analysis was performed in order to determine the discriminatory power and the reliability of the putative biomarkers for diagnosis between COPD and asthma. RESULTS PLS-DA indicated a clear separation between COPD and asthmatic patients. Among the 15 selected candidate biomarkers based on Variable Importance in Projection scores, putrescine showed the highest score. A differential IS bio-signature of 22 metabolites and lipids was found, which showed statistically significant variations between asthma and COPD. Of these 22 compounds, 18 were decreased and 4 increased in COPD compared to asthmatic patients. The IS levels of Phosphatidylethanolamine (PE) (34:1), Phosphatidylglycerol (PG) (18:1;18:2) and spermine were significantly higher in asthmatic subjects compared to COPD. CONCLUSIONS This is the first pilot study to analyse the IS metabolomics/lipidomics signatures relevant in discriminating asthma vs COPD. The role of polyamines, of 6-Hydroxykynurenic acid and of D-rhamnose as well as of other important players related to the alteration of glycerophospholipid, aminoacid/biotin and energy metabolism provided the construction of a diagnostic model that, if validated on a larger prospective cohort, might be used to rapidly and accurately discriminate asthma from COPD.
Collapse
Affiliation(s)
- Serena Correnti
- Department of Health Sciences, Magna Græcia University, 88100, Catanzaro, Italy.
| | | | - Fabia Gamboni
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Corrado Pelaia
- Department of Medical and Surgical Sciences, Magna Græcia University, 88100, Catanzaro, Italy
| | - Girolamo Pelaia
- Department of Health Sciences, Magna Græcia University, 88100, Catanzaro, Italy
| | - Rocco Savino
- Department of Medical and Surgical Sciences, Magna Græcia University, 88100, Catanzaro, Italy
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Rosa Terracciano
- Department of Experimental and Clinical Medicine, Magna Græcia University, 88100, Catanzaro, Italy.
| |
Collapse
|
21
|
Fu T, Gifford DR, Knight CG, Brockhurst MA. Eco-evolutionary dynamics of experimental Pseudomonas aeruginosa populations under oxidative stress. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001396. [PMID: 37943284 PMCID: PMC10710836 DOI: 10.1099/mic.0.001396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/26/2023] [Indexed: 11/10/2023]
Abstract
Within-host environments are likely to present a challenging and stressful environment for opportunistic pathogenic bacteria colonizing from the external environment. How populations of pathogenic bacteria respond to such environmental challenges and how this varies between strains is not well understood. Oxidative stress is one of the defences adopted by the human immune system to confront invading bacteria. In this study, we show that strains of the opportunistic pathogenic bacterium Pseudomonas aeruginosa vary in their eco-evolutionary responses to hydrogen peroxide stress. By quantifying their 24 h growth kinetics across hydrogen peroxide gradients we show that a transmissible epidemic strain isolated from a chronic airway infection of a cystic fibrosis patient, LESB58, is much more susceptible to hydrogen peroxide than either of the reference strains, PA14 or PAO1, with PAO1 showing the lowest susceptibility. Using a 12 day serial passaging experiment combined with a mathematical model, we then show that short-term susceptibility controls the longer-term survival of populations exposed to subinhibitory levels of hydrogen peroxide, but that phenotypic evolutionary responses can delay population extinction. Our model further suggests that hydrogen peroxide driven extinctions are more likely with higher rates of population turnover. Together, these findings suggest that hydrogen peroxide is likely to be an effective defence in host niches where there is high population turnover, which may explain the counter-intuitively high susceptibility of a strain isolated from chronic lung infection, where such ecological dynamics may be slower.
Collapse
Affiliation(s)
- Taoran Fu
- Division of Evolution, Infection and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Danna R. Gifford
- Division of Evolution, Infection and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Christopher G. Knight
- Department of Earth and Environmental Sciences, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PT, UK
| | - Michael A. Brockhurst
- Division of Evolution, Infection and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
22
|
Hirsch MJ, Matthews EL, Bollenbecker S, Easter M, Kiedrowski MR, Barnes JW, Krick S. Fibroblast Growth Factor 23 Signaling Does Not Increase Inflammation from Pseudomonas aeruginosa Infection in the Cystic Fibrosis Bronchial Epithelium. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1635. [PMID: 37763754 PMCID: PMC10538042 DOI: 10.3390/medicina59091635] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023]
Abstract
Background and Objectives: Chronic inflammation due to Pseudomonas aeruginosa (PA) infection in people with cystic fibrosis (CF) remains a concerning issue in the wake of modulator therapy initiation. Given the perpetuating cycle of colonization, infection, chronic inflammation, and recurrent injury to the lung, there are increases in the risk for mortality in the CF population. We have previously shown that fibroblast growth factor (FGF) 23 can exaggerate transforming growth factor (TGF) beta-mediated bronchial inflammation in CF. Our study aims to shed light on whether FGF23 signaling also plays a role in PA infection of the CF bronchial epithelium. Materials and Methods: CF bronchial epithelial cells were pretreated with FGF23 or inhibitors for FGF receptors (FGFR) and then infected with different PA isolates. After infection, immunoblot analyses were performed on these samples to assess the levels of phosphorylated phospholipase C gamma (PLCγ), total PLCγ, phosphorylated extracellular signal-regulated kinase (ERK), and total ERK. Additionally, the expression of FGFRs and interleukins at the transcript level (RT-qPCR), as well as production of interleukin (IL)-6 and IL-8 at the protein level (ELISA) were determined. Results: Although there were decreases in isoform-specific FGFRs with increases in interleukins at the mRNA level as well as phosphorylated PLCγ and the production of IL-8 protein with PA infection, treatment with FGF23 or FGFR blockade did not alter downstream targets such as IL-6 and IL-8. Conclusions: FGF23 signaling does not seem to modulate the PA-mediated inflammatory response of the CF bronchial epithelium.
Collapse
Affiliation(s)
- Meghan June Hirsch
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35233, USA; (M.J.H.)
- Gregory Fleming James Cystic Fibrosis Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Emma Lea Matthews
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35233, USA; (M.J.H.)
- Gregory Fleming James Cystic Fibrosis Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Seth Bollenbecker
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35233, USA; (M.J.H.)
| | - Molly Easter
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35233, USA; (M.J.H.)
- Gregory Fleming James Cystic Fibrosis Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Megan R. Kiedrowski
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35233, USA; (M.J.H.)
- Gregory Fleming James Cystic Fibrosis Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jarrod W. Barnes
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35233, USA; (M.J.H.)
| | - Stefanie Krick
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35233, USA; (M.J.H.)
- Gregory Fleming James Cystic Fibrosis Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
23
|
Grayton QE, Nguyen HK, Broberg CA, Ocampo J, Nagy SG, Schoenfisch MH. Biofilm Dispersal, Reduced Viscoelasticity, and Antibiotic Sensitization via Nitric Oxide-Releasing Biopolymers. ACS Infect Dis 2023; 9:1730-1741. [PMID: 37566512 DOI: 10.1021/acsinfecdis.3c00198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2023]
Abstract
Compared to planktonic bacteria, biofilms are notoriously difficult to eradicate due to their inherent protection against the immune response and antimicrobial agents. Inducing biofilm dispersal to improve susceptibility to antibiotics is an attractive therapeutic avenue for eradicating biofilms. Nitric oxide (NO), an endogenous antibacterial agent, has previously been shown to induce biofilm dispersal, but with limited understanding of the effects of NO-release properties. Herein, the antibiofilm effects of five promising NO-releasing biopolymer candidates were studied by assessing dispersal, changes in biofilm viscoelasticity, and increased sensitization to tobramycin after treatment with NO. A threshold level of NO was needed to achieve biofilm dispersal, with longer-releasing systems requiring lower concentrations. The most positively charged NO-release systems (from the presence of primary amines) led to the greatest reduction in viscoelasticity of Pseudomonas aeruginosa biofilms. Co-treatment of tobramycin with the NO-releasing biopolymer greatly decreased the dose of tobramycin required to eradicate tobramycin-susceptible and -resistant biofilms in both cellular and tissue models.
Collapse
|
24
|
Dagne A, Degu S, Abebe A, Bisrat D. Antibacterial Activity of a Phenylpropanoid from the Root Extract of Carduus leptacanthus Fresen. J Trop Med 2023; 2023:4983608. [PMID: 37711213 PMCID: PMC10499531 DOI: 10.1155/2023/4983608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/16/2023] Open
Abstract
Background The emergence and rapid spread of antimicrobial drug-resistance microorganisms exacerbate the treatment of infectious diseases, underscoring the importance of finding new, safe, and effective drugs. In Ethiopia, the roots of Carduus leptacanthus have traditionally been employed to treat microbial infectious diseases The aim of this study was to evaluate the antibacterial activity of the root extract and its primary components against six bacterial strains (Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus agalactiae, Escherichia coli, Pseudomonas aeruginosa, and Klebsiella pneumonia). Methods The extraction involved maceration of air-dried and powdered roots of C. leptacanthus with 80% methanol. The compound was isolated from the root extract using silica gel column chromatography and recrystallization in CHCl3/MeOH (9 : 1) and was characterized using ESI-MS and 1D-NMR spectroscopy. Antibacterial activity of the extract was assessed using agar well diffusion and broth microdilution methods. Results Syringin, a phenylpropanoid, was isolated and characterized from the extract of C. leptacanthus. The extract showed the most substantial efficacy against S. epidermidis (MIC = 5.33 mg/ml and inhibition zone diameter of 24 mm at 200 mg/m). Syringin also elicited antibacterial activity against S. aureus (MIC = 13.33 mg/ml), S. epidermidis (MIC = 16 mg/ml), and S. agalactiae (MIC = 16 mg/ml). Despite being tested up to a maximum concentration of 16 mg/ml, syringin did not exhibit antibacterial activity against the Gram-negative bacteria (P. aeruginosa, E. coli, and K. pneumonia). Conclusions In conclusion, the findings suggest that syringin exhibits partial involvement in the root extract's antibacterial activity, thereby potentially supporting the traditional medicinal use of the plant.
Collapse
Affiliation(s)
- Abebe Dagne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Sileshi Degu
- Traditional and Modern Medicine Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Abiy Abebe
- Traditional and Modern Medicine Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Daniel Bisrat
- Department of Pharmaceutical Chemistry and Pharmacognosy, School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
25
|
Flint R, Laucirica DR, Chan HK, Chang BJ, Stick SM, Kicic A. Stability Considerations for Bacteriophages in Liquid Formulations Designed for Nebulization. Cells 2023; 12:2057. [PMID: 37626867 PMCID: PMC10453214 DOI: 10.3390/cells12162057] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Pulmonary bacterial infections present a significant health risk to those with chronic respiratory diseases (CRDs) including cystic fibrosis (CF) and chronic-obstructive pulmonary disease (COPD). With the emergence of antimicrobial resistance (AMR), novel therapeutics are desperately needed to combat the emergence of resistant superbugs. Phage therapy is one possible alternative or adjunct to current antibiotics with activity against antimicrobial-resistant pathogens. How phages are administered will depend on the site of infection. For respiratory infections, a number of factors must be considered to deliver active phages to sites deep within the lung. The inhalation of phages via nebulization is a promising method of delivery to distal lung sites; however, it has been shown to result in a loss of phage viability. Although preliminary studies have assessed the use of nebulization for phage therapy both in vitro and in vivo, the factors that determine phage stability during nebulized delivery have yet to be characterized. This review summarizes current findings on the formulation and stability of liquid phage formulations designed for nebulization, providing insights to maximize phage stability and bactericidal activity via this delivery method.
Collapse
Affiliation(s)
- Rohan Flint
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia;
- Wal-yan Respiratory Research Center, Telethon Kids Institute, The University of Western Australia, Perth, WA 6009, Australia; (D.R.L.); (S.M.S.)
| | - Daniel R. Laucirica
- Wal-yan Respiratory Research Center, Telethon Kids Institute, The University of Western Australia, Perth, WA 6009, Australia; (D.R.L.); (S.M.S.)
| | - Hak-Kim Chan
- Advanced Drug Delivery Group, School of Pharmacy, University of Sydney, Sydney, NSW 2050, Australia;
| | - Barbara J. Chang
- The Marshall Center for Infectious Diseases Research and Training, School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia;
| | - Stephen M. Stick
- Wal-yan Respiratory Research Center, Telethon Kids Institute, The University of Western Australia, Perth, WA 6009, Australia; (D.R.L.); (S.M.S.)
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Perth, WA 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, Harry Perkins Institute of Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - Anthony Kicic
- Wal-yan Respiratory Research Center, Telethon Kids Institute, The University of Western Australia, Perth, WA 6009, Australia; (D.R.L.); (S.M.S.)
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Perth, WA 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, Harry Perkins Institute of Medical Research, The University of Western Australia, Perth, WA 6009, Australia
- School of Population Health, Curtin University, Perth, WA 6102, Australia
| |
Collapse
|
26
|
Gomez-Olivas JD, Oscullo G, Martinez-Garcia MA. Isolation of Pseudomonas aeruginosa in Stable Chronic Obstructive Pulmonary Disease Patients-Should We Treat It? J Clin Med 2023; 12:5054. [PMID: 37568456 PMCID: PMC10419574 DOI: 10.3390/jcm12155054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most frequent inflammatory diseases of the airways [...].
Collapse
Affiliation(s)
- Jose Daniel Gomez-Olivas
- Servicio de Neumología, Hospital Universitario y Politécnico La Fe, Avenida Fernando Abril Martorell 2026, 46026 Valencia, Spain; (J.D.G.-O.); (G.O.)
| | - Grace Oscullo
- Servicio de Neumología, Hospital Universitario y Politécnico La Fe, Avenida Fernando Abril Martorell 2026, 46026 Valencia, Spain; (J.D.G.-O.); (G.O.)
| | - Miguel Angel Martinez-Garcia
- Servicio de Neumología, Hospital Universitario y Politécnico La Fe, Avenida Fernando Abril Martorell 2026, 46026 Valencia, Spain; (J.D.G.-O.); (G.O.)
- CIBERES de Enfermedades Respiratorias, ISCIII, 28029 Madrid, Spain
| |
Collapse
|
27
|
Hirsch MJ, Hughes EM, Easter MM, Bollenbecker SE, Howze IV PH, Birket SE, Barnes JW, Kiedrowski MR, Krick S. A novel in vitro model to study prolonged Pseudomonas aeruginosa infection in the cystic fibrosis bronchial epithelium. PLoS One 2023; 18:e0288002. [PMID: 37432929 PMCID: PMC10335692 DOI: 10.1371/journal.pone.0288002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 06/16/2023] [Indexed: 07/13/2023] Open
Abstract
Pseudomonas aeruginosa (PA) is known to chronically infect airways of people with cystic fibrosis (CF) by early adulthood. PA infections can lead to increased airway inflammation and lung tissue damage, ultimately contributing to decreased lung function and quality of life. Existing models of PA infection in vitro commonly utilize 1-6-hour time courses. However, these relatively early time points may not encompass downstream airway cell signaling in response to the chronic PA infections observed in people with cystic fibrosis. To fill this gap in knowledge, the aim of this study was to establish an in vitro model that allows for PA infection of CF bronchial epithelial cells, cultured at the air liquid interface, for 24 hours. Our model shows with an inoculum of 2 x 102 CFUs of PA for 24 hours pro-inflammatory markers such as interleukin 6 and interleukin 8 are upregulated with little decrease in CF bronchial epithelial cell survival or monolayer confluency. Additionally, immunoblotting for phosphorylated phospholipase C gamma, a well-known downstream protein of fibroblast growth factor receptor signaling, showed significantly elevated levels after 24 hours with PA infection that were not seen at earlier timepoints. Finally, inhibition of phospholipase C shows significant downregulation of interleukin 8. Our data suggest that this newly developed in vitro "prolonged PA infection model" recapitulates the elevated inflammatory markers observed in CF, without compromising cell survival. This extended period of PA growth on CF bronchial epithelial cells will have impact on further studies of cell signaling and microbiological studies that were not possible in previous models using shorter PA exposures.
Collapse
Affiliation(s)
- Meghan J. Hirsch
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States of America
- Gregory Fleming James Cystic Fibrosis Center, The University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Emily M. Hughes
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States of America
- Gregory Fleming James Cystic Fibrosis Center, The University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Molly M. Easter
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States of America
- Gregory Fleming James Cystic Fibrosis Center, The University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Seth E. Bollenbecker
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Patrick H. Howze IV
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Susan E. Birket
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States of America
- Gregory Fleming James Cystic Fibrosis Center, The University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Jarrod W. Barnes
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Megan R. Kiedrowski
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States of America
- Gregory Fleming James Cystic Fibrosis Center, The University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Stefanie Krick
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States of America
- Gregory Fleming James Cystic Fibrosis Center, The University of Alabama at Birmingham, Birmingham, AL, United States of America
| |
Collapse
|
28
|
Wen L, Zhang T, Chen F, Hu L, Dou C, Ding X, Altamirano A, Wei G, Yan Z. Modified Dingchuan Decoction treats cough-variant asthma by suppressing lung inflammation and regulating the lung microbiota. JOURNAL OF ETHNOPHARMACOLOGY 2023; 306:116171. [PMID: 36646156 DOI: 10.1016/j.jep.2023.116171] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Modified Dingchuan Decoction (MDD) is a Chinese medicine formula containing 11 materials with cough suppression, asthma relief, and anti-inflammatory effects. AIM OF THE STUDY This study aimed to evaluate the therapeutic effect of MDD on cough-variant asthma (CVA) and to investigate its mechanism of action. MATERIALS AND METHODS The chemical constituents of MDD were analyzed by ultra-performance liquid chromatography-quadrupole/electrostatic field orbitrap high-resolution mass spectrometry (UPLC-Q-Orbitrap HRMS). A guinea pig CVA model was established using an intramuscular injection of ovalbumin (OVA), combined with an intraperitoneal injection of aluminum hydroxide [Al(OH)3] and nebulized OVA. At the beginning of day 18, the low, medium, and high MDD groups were gavaged with 7.23 g/kg, 14.46 g/kg, and 28.92 g/kg of MDD, respectively, and the positive group was gavaged with 5 mg/kg of prednisone acetate combined with 1 mg/kg of montelukast sodium; the normal and model groups were given an equal volume of distilled water, once a day for 21 days. The cough was induced by 10-3 mol/L capsaicin solution 1 h after the last administration, and the number of coughs and the latency of coughs were evaluated. Hematoxylin and eosin staining (H&E) was used to observe pathological changes in the lungs and airways. The concentration of inflammatory factors in bronchoalveolar lavage fluid (BALF) was measured by enzyme-linked immunosorbent assay (ELISA). We analyzed the lung microbiota using 16 S ribosomal DNA (16 S rDNA) high-throughput sequencing. RESULTS The 38 chemical components were found in MDD, and MDD reduced the number of coughs in guinea pigs with CVA, prolonged cough latency, improved pathological damage to the lungs and airways, regulated inflammatory factor levels in BALF, and modulated the lung microbiota. CONCLUSIONS This study demonstrated that treating CVA with MDD may be related to inhibiting lung inflammation and regulating lung microbiota.
Collapse
Affiliation(s)
- Lingmiao Wen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Tinglan Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Fangfang Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Lin Hu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Chongyang Dou
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Xian Ding
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Alvin Altamirano
- Department of Chemistry and Biochemistry, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Guihua Wei
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Zhiyong Yan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
29
|
Carneiro J, Magalhães RP, de la Oliva Roque VM, Simões M, Pratas D, Sousa SF. TargIDe: a machine-learning workflow for target identification of molecules with antibiofilm activity against Pseudomonas aeruginosa. J Comput Aided Mol Des 2023; 37:265-278. [PMID: 37085636 DOI: 10.1007/s10822-023-00505-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/12/2023] [Indexed: 04/23/2023]
Abstract
Bacterial biofilms are a source of infectious human diseases and are heavily linked to antibiotic resistance. Pseudomonas aeruginosa is a multidrug-resistant bacterium widely present and implicated in several hospital-acquired infections. Over the last years, the development of new drugs able to inhibit Pseudomonas aeruginosa by interfering with its ability to form biofilms has become a promising strategy in drug discovery. Identifying molecules able to interfere with biofilm formation is difficult, but further developing these molecules by rationally improving their activity is particularly challenging, as it requires knowledge of the specific protein target that is inhibited. This work describes the development of a machine learning multitechnique consensus workflow to predict the protein targets of molecules with confirmed inhibitory activity against biofilm formation by Pseudomonas aeruginosa. It uses a specialized database containing all the known targets implicated in biofilm formation by Pseudomonas aeruginosa. The experimentally confirmed inhibitors available on ChEMBL, together with chemical descriptors, were used as the input features for a combination of nine different classification models, yielding a consensus method to predict the most likely target of a ligand. The implemented algorithm is freely available at https://github.com/BioSIM-Research-Group/TargIDe under licence GNU General Public Licence (GPL) version 3 and can easily be improved as more data become available.
Collapse
Affiliation(s)
- João Carneiro
- Interdisciplinary Centre of Marine and Environmental Research, CIIMAR, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, Porto, 4450-208, Portugal.
| | - Rita P Magalhães
- Faculty of Medicine, Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4200-319, Porto, Portugal
- Department of Biomedicine, Faculty of Medicine, UCIBIO-Applied Molecular Biosciences Unit, University of Porto, BioSIM, Porto, 4200-319, Portugal
| | - Victor M de la Oliva Roque
- Faculty of Medicine, Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4200-319, Porto, Portugal
- Department of Biomedicine, Faculty of Medicine, UCIBIO-Applied Molecular Biosciences Unit, University of Porto, BioSIM, Porto, 4200-319, Portugal
| | - Manuel Simões
- Faculty of Engineering, LEPABE Laboratory for Process Engineering, Environment, Biotechnology and Energy, University of Porto, Rua Dr. Roberto Frias, s/n, Porto, 4200-465, Portugal
- Faculty of Engineering, ALiCE-Associate Laboratory in Chemical Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
| | - Diogo Pratas
- Institute of Electronics and Informatics Engineering of Aveiro, IEETA, University of Aveiro, Aveiro, Portugal
- Department of Electronics, Telecommunications and Informatics, DETI, University of Aveiro, Aveiro, Portugal
- Department of Virology, DoV, University of Helsinki, Helsinki, Finland
| | - Sérgio F Sousa
- Faculty of Medicine, Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4200-319, Porto, Portugal
- Department of Biomedicine, Faculty of Medicine, UCIBIO-Applied Molecular Biosciences Unit, University of Porto, BioSIM, Porto, 4200-319, Portugal
| |
Collapse
|
30
|
Pottier M, Gravey F, Castagnet S, Auzou M, Langlois B, Guérin F, Giard JC, Léon A, Le Hello S. A 10-year microbiological study of Pseudomonas aeruginosa strains revealed the circulation of populations resistant to both carbapenems and quaternary ammonium compounds. Sci Rep 2023; 13:2639. [PMID: 36788252 PMCID: PMC9929048 DOI: 10.1038/s41598-023-29590-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Pseudomonas aeruginosa is one of the leading causes of healthcare-associated infections. For this study, the susceptibility profiles to antipseudomonal antibiotics and a quaternary ammonium compound, didecyldimethylammonium chloride (DDAC), widely used as a disinfectant, were established for 180 selected human and environmental hospital strains isolated between 2011 and 2020. Furthermore, a genomic study determined resistome and clonal putative relatedness for 77 of them. During the ten-year study period, it was estimated that 9.5% of patients' strains were resistant to carbapenems, 11.9% were multidrug-resistant (MDR), and 0.7% were extensively drug-resistant (XDR). Decreased susceptibility (DS) to DDAC was observed for 28.0% of strains, a phenotype significantly associated with MDR/XDR profiles and from hospital environmental samples (p < 0.0001). According to genomic analyses, the P. aeruginosa population unsusceptible to carbapenems and/or to DDAC was diverse but mainly belonged to top ten high-risk clones described worldwide by del Barrio-Tofiño et al. The carbapenem resistance appeared mainly due to the production of the VIM-2 carbapenemase (39.3%) and DS to DDAC mediated by MexAB-OprM pump efflux overexpression. This study highlights the diversity of MDR/XDR populations of P. aeruginosa which are unsusceptible to compounds that are widely used in medicine and hospital disinfection and are probably distributed in hospitals worldwide.
Collapse
Affiliation(s)
- Marine Pottier
- Research Department, LABÉO, 14053, Caen, France.,UNICAEN, Univ Rouen Normandie, INSERM DYNAMICURE UMR 1311, CHU Caen, department of microbiology, Normandie Univ, 14000, Caen, France
| | - François Gravey
- UNICAEN, Univ Rouen Normandie, INSERM DYNAMICURE UMR 1311, CHU Caen, department of microbiology, Normandie Univ, 14000, Caen, France.,Service de Microbiologie, CHU de Caen, Avenue de la Côte de Nacre, 14033, Caen Cedex, France
| | - Sophie Castagnet
- Research Department, LABÉO, 14053, Caen, France.,UNICAEN, Univ Rouen Normandie, INSERM DYNAMICURE UMR 1311, CHU Caen, department of microbiology, Normandie Univ, 14000, Caen, France
| | - Michel Auzou
- Service de Microbiologie, CHU de Caen, Avenue de la Côte de Nacre, 14033, Caen Cedex, France
| | - Bénédicte Langlois
- Service de Microbiologie, CHU de Caen, Avenue de la Côte de Nacre, 14033, Caen Cedex, France
| | - François Guérin
- Laboratoire de Bactériologie et Hygiène Hospitalière, CHU de Rennes, 2 Rue Henri Le Guilloux, 35033, Rennes Cedex 9, France
| | - Jean-Christophe Giard
- UNICAEN, Univ Rouen Normandie, INSERM DYNAMICURE UMR 1311, CHU Caen, department of microbiology, Normandie Univ, 14000, Caen, France
| | - Albertine Léon
- Research Department, LABÉO, 14053, Caen, France.,UNICAEN, Univ Rouen Normandie, INSERM DYNAMICURE UMR 1311, CHU Caen, department of microbiology, Normandie Univ, 14000, Caen, France
| | - Simon Le Hello
- UNICAEN, Univ Rouen Normandie, INSERM DYNAMICURE UMR 1311, CHU Caen, department of microbiology, Normandie Univ, 14000, Caen, France. .,Service de Microbiologie, CHU de Caen, Avenue de la Côte de Nacre, 14033, Caen Cedex, France. .,Service d'Hygiène Hospitalière, CHU de Caen, Avenue de la Côte de Nacre, 14033, Caen Cedex, France.
| |
Collapse
|
31
|
Salvà-Serra F, Jaén-Luchoro D, Marathe NP, Adlerberth I, Moore ERB, Karlsson R. Responses of carbapenemase-producing and non-producing carbapenem-resistant Pseudomonas aeruginosa strains to meropenem revealed by quantitative tandem mass spectrometry proteomics. Front Microbiol 2023; 13:1089140. [PMID: 36845973 PMCID: PMC9948630 DOI: 10.3389/fmicb.2022.1089140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/08/2022] [Indexed: 02/11/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen with increasing incidence of multidrug-resistant strains, including resistance to last-resort antibiotics, such as carbapenems. Resistances are often due to complex interplays of natural and acquired resistance mechanisms that are enhanced by its large regulatory network. This study describes the proteomic responses of two carbapenem-resistant P. aeruginosa strains of high-risk clones ST235 and ST395 to subminimal inhibitory concentrations (sub-MICs) of meropenem by identifying differentially regulated proteins and pathways. Strain CCUG 51971 carries a VIM-4 metallo-β-lactamase or 'classical' carbapenemase; strain CCUG 70744 carries no known acquired carbapenem-resistance genes and exhibits 'non-classical' carbapenem-resistance. Strains were cultivated with different sub-MICs of meropenem and analyzed, using quantitative shotgun proteomics based on tandem mass tag (TMT) isobaric labeling, nano-liquid chromatography tandem-mass spectrometry and complete genome sequences. Exposure of strains to sub-MICs of meropenem resulted in hundreds of differentially regulated proteins, including β-lactamases, proteins associated with transport, peptidoglycan metabolism, cell wall organization, and regulatory proteins. Strain CCUG 51971 showed upregulation of intrinsic β-lactamases and VIM-4 carbapenemase, while CCUG 70744 exhibited a combination of upregulated intrinsic β-lactamases, efflux pumps, penicillin-binding proteins and downregulation of porins. All components of the H1 type VI secretion system were upregulated in strain CCUG 51971. Multiple metabolic pathways were affected in both strains. Sub-MICs of meropenem cause marked changes in the proteomes of carbapenem-resistant strains of P. aeruginosa exhibiting different resistance mechanisms, involving a wide range of proteins, many uncharacterized, which might play a role in the susceptibility of P. aeruginosa to meropenem.
Collapse
Affiliation(s)
- Francisco Salvà-Serra
- Department of Infectious Diseases, Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden,Culture Collection University of Gothenburg (CCUG), Department of Clinical Microbiology, Sahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden,Microbiology, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain,*Correspondence: Francisco Salvà-Serra, ✉
| | - Daniel Jaén-Luchoro
- Department of Infectious Diseases, Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Culture Collection University of Gothenburg (CCUG), Department of Clinical Microbiology, Sahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden
| | | | - Ingegerd Adlerberth
- Department of Infectious Diseases, Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden,Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden
| | - Edward R. B. Moore
- Department of Infectious Diseases, Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden,Culture Collection University of Gothenburg (CCUG), Department of Clinical Microbiology, Sahlgrenska University Hospital and Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden
| | - Roger Karlsson
- Department of Infectious Diseases, Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden,Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden,Nanoxis Consulting AB, Gothenburg, Sweden,Roger Karlsson, ✉
| |
Collapse
|
32
|
Martin LW, Gray AR, Brockway B, Lamont IL. Pseudomonas aeruginosa is oxygen-deprived during infection in cystic fibrosis lungs, reducing the effectiveness of antibiotics. FEMS Microbiol Lett 2023; 370:fnad076. [PMID: 37516450 PMCID: PMC10408701 DOI: 10.1093/femsle/fnad076] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/04/2023] [Accepted: 07/20/2023] [Indexed: 07/31/2023] Open
Abstract
Pseudomonas aeruginosa infects the lungs of patients with cystic fibrosis. Sputum expectorated from the lungs of patients contains low levels of oxygen, indicating that P. aeruginosa may be oxygen-deprived during infection. During in vitro growth under oxygen-limiting conditions, a P. aeruginosa reference strain increases expression of a cytochrome oxidase with a high affinity for oxygen, and of nitrate and nitrite reductases that enable it to use nitrate instead of oxygen during respiration. Here, we quantified transcription of the genes encoding these three enzymes in sputum samples from 18 infected patients, and in bacteria isolated from the sputum samples and grown in aerobic and anaerobic culture. In culture, expression of all three genes was increased by averages of 20- to 500-fold in anaerobically grown bacteria compared with those grown aerobically, although expression levels varied greatly between isolates. Expression of the same genes in sputum was similar to that of the corresponding bacteria in anaerobic culture. The isolated bacteria were less susceptible to tobramycin and ciprofloxacin, two widely used anti-pseudomonal antibiotics, when grown anaerobically than when grown aerobically. Our findings show that P. aeruginosa experiences oxygen starvation during infection in cystic fibrosis, reducing the effectiveness of antibiotic treatment.
Collapse
Affiliation(s)
- Lois W Martin
- Department of Biochemistry, University of Otago, Dunedin, 9016, New Zealand
| | - Andrew R Gray
- Biostatistics Centre, University of Otago, Dunedin 9016, New Zealand
| | - Ben Brockway
- Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Iain L Lamont
- Department of Biochemistry, University of Otago, Dunedin, 9016, New Zealand
| |
Collapse
|
33
|
Gene-Gene Interactions Reduce Aminoglycoside Susceptibility of Pseudomonas aeruginosa through Efflux Pump-Dependent and -Independent Mechanisms. Antibiotics (Basel) 2023; 12:antibiotics12010152. [PMID: 36671353 PMCID: PMC9854422 DOI: 10.3390/antibiotics12010152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/10/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Pseudomonas aeruginosa causes a wide range of acute and chronic infections. Aminoglycosides are a cornerstone of treatment, but isolates are often resistant. The purpose of this research was to better understand the genetic basis of aminoglycoside resistance in P. aeruginosa. Bioinformatic approaches identified mutations in resistance-associated genes in the clinical isolates of P. aeruginosa. The common mutations were then engineered into the genome of P. aeruginosa reference strain PAO1. Mutations in the elongation factor gene fusA1 caused the biggest reduction in aminoglycoside susceptibility, with mutations in the two-component regulator gene amgS and the efflux pump regulator gene mexZ having less impact. This susceptibility was further reduced by combinations of mutations. Mutations in fusA1, amgS and mexZ all increased the expression of the mexXY efflux pump that is strongly associated with aminoglycoside resistance. Furthermore, the fusA1 amgS mexZ triple mutant had the highest efflux pump gene expression. Engineering fusA1 and amgS mutants lacking this efflux pump showed that fusA1 and amgS also reduce aminoglycoside susceptibility through additional mechanisms. The fusA1 and amgS mutations reduced bacterial growth, showing that these mutations have a fitness cost. Our findings demonstrate the complex interplay between mutations, efflux pump expression and other mechanisms for reducing the susceptibility of P. aeruginosa to aminoglycosides.
Collapse
|
34
|
Liu Y, Teo SM, Méric G, Tang HHF, Zhu Q, Sanders JG, Vázquez-Baeza Y, Verspoor K, Vartiainen VA, Jousilahti P, Lahti L, Niiranen T, Havulinna AS, Knight R, Salomaa V, Inouye M. The gut microbiome is a significant risk factor for future chronic lung disease. J Allergy Clin Immunol 2022; 151:943-952. [PMID: 36587850 PMCID: PMC10109092 DOI: 10.1016/j.jaci.2022.12.810] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/21/2022] [Accepted: 12/05/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND The gut-lung axis is generally recognized, but there are few large studies of the gut microbiome and incident respiratory disease in adults. OBJECTIVE We sought to investigate the association and predictive capacity of the gut microbiome for incident asthma and chronic obstructive pulmonary disease (COPD). METHODS Shallow metagenomic sequencing was performed for stool samples from a prospective, population-based cohort (FINRISK02; N = 7115 adults) with linked national administrative health register-derived classifications for incident asthma and COPD up to 15 years after baseline. Generalized linear models and Cox regressions were used to assess associations of microbial taxa and diversity with disease occurrence. Predictive models were constructed using machine learning with extreme gradient boosting. Models considered taxa abundances individually and in combination with other risk factors, including sex, age, body mass index, and smoking status. RESULTS A total of 695 and 392 statistically significant associations were found between baseline taxonomic groups and incident asthma and COPD, respectively. Gradient boosting decision trees of baseline gut microbiome abundance predicted incident asthma and COPD in the validation data sets with mean area under the curves of 0.608 and 0.780, respectively. Cox analysis showed that the baseline gut microbiome achieved higher predictive performance than individual conventional risk factors, with C-indices of 0.623 for asthma and 0.817 for COPD. The integration of the gut microbiome and conventional risk factors further improved prediction capacities. CONCLUSIONS The gut microbiome is a significant risk factor for incident asthma and incident COPD and is largely independent of conventional risk factors.
Collapse
Affiliation(s)
- Yang Liu
- Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Melbourne, Australia; Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - Shu Mei Teo
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia; Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom; Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Guillaume Méric
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Howard H F Tang
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia; Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Qiyun Zhu
- School of Life Sciences, Arizona State University, Tempe, Ariz; Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, Ariz
| | - Jon G Sanders
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY
| | - Yoshiki Vázquez-Baeza
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California San Diego, La Jolla, Calif
| | - Karin Verspoor
- School of Computing Technologies, RMIT University, Melbourne, Australia; School of Computing and Information Systems, The University of Melbourne, Melbourne, Australia
| | - Ville A Vartiainen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Pulmonary Medicine, Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Pekka Jousilahti
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Teemu Niiranen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Division of Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Aki S Havulinna
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Institute for Molecular Medicine Finland, FIMM-HiLIFE, University of Helsinki, Helsinki, Finland
| | - Rob Knight
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California San Diego, La Jolla, Calif; Department of Computer Science and Engineering, University of California San Diego, La Jolla, Calif; Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, Calif
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Michael Inouye
- Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Melbourne, Australia; Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia; Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom; British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom; British Heart Foundation Cambridge Centre of Research Excellence, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom; Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom; The Alan Turing Institute, London, United Kingdom; Heart and Lung Research Institute, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
35
|
Sarkar PK, Akand N, Tahura S, Kamruzzaman M, Akter J, Zaman KA, Farhana T, Rima SS, Alam MJ, Hassan MK, Fardous J. Antimicrobial sensitivity pattern of children with cystic fibrosis in Bangladesh: a lesson from a specialized Sishu (Children) Hospital. EGYPTIAN PEDIATRIC ASSOCIATION GAZETTE 2022. [DOI: 10.1186/s43054-022-00127-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Abstract
Background
Infection control in cystic fibrosis (CF) patients plays a crucial role in improving the survival of patients with CF. Antimicrobial sensitivity patterns in these patient groups in our country are currently lacking. Therefore, the purpose of the study was to evaluate the microbiological cultures and antimicrobial susceptibility pattern of pediatric CF patients.
Method
A total of 50 respiratory samples were prospectively collected from the period between February 2021 and October 2021. Sputum and oropharyngeal swabs were processed for culture and microbiological testing. Sample collection and evaluation were performed according to the Good Laboratory Practice guidelines (GLP). Informed written consent was ensured before participation. Statistical analysis was performed with SPSS v 26.
Result
The median age of the children was 30 months (6–120) months, with a male predominance (66% vs 34%). Single and two organisms were isolated in 72% (n = 36) and 12% (n = 6) of cases, respectively. During the study period, 36% of the patients harbored Pseudomonas aeruginosa, 18% harbored Klebsiella pneumoniae, and both Staphylococcus aureus and Escherichia coli were detected in 16% of cases. Levofloxacin was found to be the most active antibiotic agent with 100% susceptibility. In contrast, nearly all isolates were resistant to amoxicillin, erythromycin and rifampicin.
Conclusion
Levofloxacin is the most effective agent to treat CF patients. Active surveillance of the resistance pattern should always continue to be promoted.
Collapse
|
36
|
Laucirica DR, Stick SM, Garratt LW, Kicic A. Bacteriophage: A new therapeutic player to combat neutrophilic inflammation in chronic airway diseases. Front Med (Lausanne) 2022; 9:1069929. [PMID: 36590945 PMCID: PMC9794625 DOI: 10.3389/fmed.2022.1069929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
Persistent respiratory bacterial infections are a clinical burden in several chronic inflammatory airway diseases and are often associated with neutrophil infiltration into the lungs. Following recruitment, dysregulated neutrophil effector functions such as increased granule release and formation of neutrophil extracellular traps (NETs) result in damage to airway tissue, contributing to the progression of lung disease. Bacterial pathogens are a major driver of airway neutrophilic inflammation, but traditional management of infections with antibiotic therapy is becoming less effective as rates of antimicrobial resistance rise. Bacteriophages (phages) are now frequently identified as antimicrobial alternatives for antimicrobial resistant (AMR) airway infections. Despite growing recognition of their bactericidal function, less is known about how phages influence activity of neutrophils recruited to sites of bacterial infection in the lungs. In this review, we summarize current in vitro and in vivo findings on the effects of phage therapy on neutrophils and their inflammatory mediators, as well as mechanisms of phage-neutrophil interactions. Understanding these effects provides further validation of their safe use in humans, but also identifies phages as a targeted neutrophil-modulating therapeutic for inflammatory airway conditions.
Collapse
Affiliation(s)
- Daniel R. Laucirica
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
| | - Stephen M. Stick
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| | - Luke W. Garratt
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
| | - Anthony Kicic
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
| |
Collapse
|
37
|
van Dijk MD, Voor In 't Holt AF, Alp E, Hell M, Petrosillo N, Presterl E, Tsakris A, Severin JA, Vos MC. Infection prevention and control policies in hospitals and prevalence of highly resistant microorganisms: an international comparative study. Antimicrob Resist Infect Control 2022; 11:152. [PMID: 36474304 PMCID: PMC9727845 DOI: 10.1186/s13756-022-01165-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/20/2022] [Accepted: 10/08/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND There are differences in infection prevention and control (IPC) policies to prevent transmission of highly resistant microorganisms (HRMO). The aim of this study is to give an overview of the IPC policy of six European hospitals and their HRMO prevalence, to compare the IPC policies of these hospitals with international guidelines, and to investigate the hospitals' adherence to their own IPC policy. METHODS The participating hospitals were located in Salzburg (Austria), Vienna (Austria), Kayseri (Turkey), Piraeus (Greece), Rome (Italy) and Rotterdam (The Netherlands). Data were collected via an online survey. Questions were aimed at prevalence rates in the years 2014, 2015, 2016 of carbapenemase-producing Klebsiella pneumoniae (CPK), carbapenemase-producing Pseudomonas aeruginosa (CPPA), vancomycin-resistant Enterococcus faecium (VRE) and hospitals' IPC policies of 2017. Implemented IPC measures (i.e. with a self-reported adherence of > 90%) were counted (26 points maximal). RESULTS The self-reported prevalence of CPK per year was low in the Austrian and Dutch hospitals and high in the Turkish and Greek hospitals. CPPA was highly prevalent in the Turkish hospital only, while the prevalence of VRE in four hospitals, except the Austrian hospitals which reported lower prevalence numbers, was more evenly distributed. The Dutch hospital had implemented the most IPC measures (n = 21), the Turkish and Greek hospitals the least (n = 14 and 7, respectively). CONCLUSION Hospitals with the highest self-reported prevalence of CPK and CPPA reported the least implemented IPC measures. Also, hospitals with a higher prevalence often reported a lower adherence to own IPC policy.
Collapse
Affiliation(s)
- Manon D van Dijk
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Centre Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Anne F Voor In 't Holt
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Centre Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Emine Alp
- Department of Infectious Diseases and Clinical Microbiology, Medical Faculty, Erciyes University, Kayseri, Turkey
- Department of Infectious Diseases and Clinical Microbiology, Medical Faculty, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Markus Hell
- Department of Clinical Microbiology and Infection Control, MEDILAB-Academic Teaching Laboratories, Paracelsus Medical University, Salzburg, Austria
- Teaching Hospital, Kardinal Schwarzenberg Klinikum, Paracelsus Medical University, Schwarzach, Austria
| | - Nicola Petrosillo
- Clinical and Research Department for Infectious Diseases, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
- Department of Infection Control, University Hospital Campus Bio-Medico, Rome, Italy
| | - Elisabeth Presterl
- Department of Infection Control and Hospital Epidemiology, Medical University of Vienna, Vienna, Austria
| | - Athanasios Tsakris
- Department of Microbiology, Medical School, University of Athens, Athens, Greece
| | - Juliëtte A Severin
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Centre Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Margreet C Vos
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Centre Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | | |
Collapse
|
38
|
Effect of Flagellin Pre-Exposure on the Inflammatory and Antifungal Response of Bronchial Epithelial Cells to Fungal Pathogens. J Fungi (Basel) 2022; 8:jof8121268. [PMID: 36547601 PMCID: PMC9782670 DOI: 10.3390/jof8121268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Bronchial epithelial cells (BEC) play a crucial role in innate immunity against inhaled fungi. Indeed, in response to microorganisms, BEC synthesize proinflammatory cytokines involved in the recruitment of neutrophils. We have recently shown that BEC exert antifungal activity against Aspergillus fumigatus by inhibiting filament growth. In the present study, we first analyzed the inflammatory and antifungal responses of BEC infected by several fungal species such as Aspergillus spp., Scedosporium apiospermum and Candida albicans, which are frequently isolated from the sputum of people with chronic pulmonary diseases. The airways of these patients, such as people with cystic fibrosis (pwCF), are mainly colonized by P. aeruginosa and secondary by fungal pathogens. We have previously demonstrated that BEC are capable of innate immune memory, allowing them to increase their inflammatory response against A. fumigatus following a previous contact with Pseudomonas aeruginosa flagellin. To identify the impact of bacteria exposure on BEC responses to other fungal infections, we extended the analysis of BEC innate immune memory to Aspergillus spp., Scedosporium apiospermum and Candida albicans infection. Our results show that BEC are able to recognize and respond to Aspergillus spp., S. apiospermum and C. albicans infection and that the modulation of BEC responses by pre-exposure to flagellin varies according to the fungal species encountered. Deepening our knowledge of the innate immune memory of BEC should open new therapeutic avenues to modulate the inflammatory response against polymicrobial infections observed in chronic pulmonary diseases such as CF.
Collapse
|
39
|
Clinical Course of Children with Chronic Suppurative Lung Disease or Bronchiectasis Infected with Pseudomonas aeruginosa. CHILDREN (BASEL, SWITZERLAND) 2022; 9:children9121822. [PMID: 36553266 PMCID: PMC9776566 DOI: 10.3390/children9121822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
Children with chronic wet cough and without cystic fibrosis (non-CF) may suffer from chronic suppurative lung disease (CSLD) or bronchiectasis. Pseudomonas aeruginosa (Pa) can be one of the offending microbes in these children. The present study aimed to describe the clinical course of children with the above two conditions who were infected with Pa. Data of 54 children with CSLD/bronchiectasis who were diagnosed and attended in our department were retrospectively analysed through a Cox proportional hazard model, with age, presence of bronchiectasis, use of inhaled colistin, azithromycin, inhaled hypertonic saline as the covariates. In 42 of the 54 patients, there was no identifiable cause or underlying chronic disorder. Microbiological clearance was defined as the absence of daily wet cough for four months along with four negative cultures taken during the last four consecutive follow-up visits. Multivariate analysis was performed with a Cox proportional hazard model with time to microbiological clearance as the outcome. Results are described as Hazard Ratios (HR) with 95% Confidence Intervals (95%CI). Nebulised antibiotics and the presence of bronchiectasis were statistically significant predictors of remission (HR: 3.99; 95%CI: 1.12-14.14; p = 0.032, and HR: 0.24; 95%CI: 0.08-0.71; p = 0.010). In conclusion, the rate of microbiological clearance increases with the use of inhaled colistin and decreases when there is established bronchiectasis.
Collapse
|
40
|
Phage phiZ98: a novel tri-segmented dsRNA cystovirus for controlling Pseudomonas strains with defective lipopolysaccharides in foods. Food Res Int 2022; 162:112197. [DOI: 10.1016/j.foodres.2022.112197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022]
|
41
|
Bermeo R, Lal K, Ruggeri D, Lanaro D, Mazzotta S, Vasile F, Imberty A, Belvisi L, Varrot A, Bernardi A. Targeting a Multidrug-Resistant Pathogen: First Generation Antagonists of Burkholderia cenocepacia's BC2L-C Lectin. ACS Chem Biol 2022; 17:2899-2910. [PMID: 36174276 PMCID: PMC9594048 DOI: 10.1021/acschembio.2c00532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Multidrug-resistant pathogens such as Burkholderia cenocepacia have become a hazard in the context of healthcare-associated infections, especially for patients admitted with cystic fibrosis or immuno-compromising conditions. Like other opportunistic Gram-negative bacteria, this pathogen establishes virulence and biofilms through lectin-mediated adhesion. In particular, the superlectin BC2L-C is believed to cross-link human epithelial cells to B. cenocepacia during pulmonary infections. We aimed to obtain glycomimetic antagonists able to inhibit the interaction between the N-terminal domain of BC2L-C (BC2L-C-Nt) and its target fucosylated human oligosaccharides. In a previous study, we identified by fragment virtual screening and validated a small set of molecular fragments that bind BC2L-C-Nt in the vicinity of the fucose binding site. Here, we report the rational design and synthesis of bifunctional C- or N-fucosides, generated by connecting these fragments to a fucoside core using a panel of rationally selected linkers. A modular route starting from two key fucoside intermediates was implemented for the synthesis, followed by evaluation of the new compounds as BC2L-C-Nt ligands with a range of techniques (surface plasmon resonance, isothermal titration calorimetry, saturation transfer difference NMR, differential scanning calorimetry, and X-ray crystallography). This study resulted in a hit molecule with an order of magnitude gain over the starting methyl fucoside and in two crystal structures of antagonist/lectin complexes.
Collapse
Affiliation(s)
- Rafael Bermeo
- CNRS,
CERMAV, Univ. Grenoble Alpes, Grenoble 38000, France,Dipartimento
di Chimica, Università degli Studi
di Milano, via Golgi 19, Milano 20133, Italy
| | - Kanhaya Lal
- CNRS,
CERMAV, Univ. Grenoble Alpes, Grenoble 38000, France,Dipartimento
di Chimica, Università degli Studi
di Milano, via Golgi 19, Milano 20133, Italy
| | - Davide Ruggeri
- Dipartimento
di Chimica, Università degli Studi
di Milano, via Golgi 19, Milano 20133, Italy
| | - Daniele Lanaro
- Dipartimento
di Chimica, Università degli Studi
di Milano, via Golgi 19, Milano 20133, Italy
| | - Sarah Mazzotta
- Dipartimento
di Chimica, Università degli Studi
di Milano, via Golgi 19, Milano 20133, Italy
| | - Francesca Vasile
- Dipartimento
di Chimica, Università degli Studi
di Milano, via Golgi 19, Milano 20133, Italy
| | - Anne Imberty
- CNRS,
CERMAV, Univ. Grenoble Alpes, Grenoble 38000, France
| | - Laura Belvisi
- Dipartimento
di Chimica, Università degli Studi
di Milano, via Golgi 19, Milano 20133, Italy
| | | | - Anna Bernardi
- Dipartimento
di Chimica, Università degli Studi
di Milano, via Golgi 19, Milano 20133, Italy,
| |
Collapse
|
42
|
Lin Z, Yang X, Guan L, Qin L, Ding J, Zhou L. The link between ferroptosis and airway inflammatory diseases: A novel target for treatment. Front Mol Biosci 2022; 9:985571. [PMID: 36060261 PMCID: PMC9428508 DOI: 10.3389/fmolb.2022.985571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Ferroptosis is an iron-dependent mode of cell death characterized by intracellular lipid peroxide accumulation and a redox reaction imbalance. Compared with other modes of cell death, ferroptosis has specific biological and morphological features. The iron-dependent lipid peroxidation accumulation is manifested explicitly in the abnormal metabolism of intracellular lipid oxides catalyzed by excessive iron ions with the production of many reactive oxygen species and over-oxidization of polyunsaturated fatty acids. Recent studies have shown that various diseases, which include intestinal diseases and cancer, are associated with ferroptosis, but few studies are related to airway inflammatory diseases. This review provides a comprehensive analysis of the primary damage mechanisms of ferroptosis and summarizes the relationship between ferroptosis and airway inflammatory diseases. In addition to common acute and chronic airway inflammatory diseases, we also focus on the progress of research on COVID-19 in relation to ferroptosis. New therapeutic approaches and current issues to be addressed in the treatment of inflammatory airway diseases using ferroptosis are further proposed.
Collapse
|
43
|
Mai K, Chen X, Zhang K, Gu S, Wu X, Gu Z, Wu Z, Huang K, Liu Z, Yang Z, Chen D. A juvenile murine model with chronic lung inflammation induced by repeated intratracheal instillation of lipopolysaccharides: a versatile and replicable model. Transl Pediatr 2022; 11:1292-1300. [PMID: 36072534 PMCID: PMC9442212 DOI: 10.21037/tp-22-44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/06/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Recurrent lower respiratory tract infection or chronic pulmonary infection often occur in children with chronic lung diseases (CLDs). By continuous lung inflammation, recurrent and chronic infection could cause irreversible airway structural and lung function damage, which eventually leads to respiratory failure and death. METHODS In purpose of recapitulating persistent high-intensity lung inflammation caused by recurrent lower respiratory tract infection or chronic infection, we established a juvenile murine model with chronic lung inflammation induced by repeated intratracheal instillations of lipopolysaccharides (LPS) from Pseudomonas aeruginosa once a week for 4 weeks. Four-week-old C57BL/6N mice were divided into 4 groups, including LPS0.5 group (n=15), LPS1.0 group (n=15), Control group (n=15) and Normal group (n=15). Mice in LPS0.5 group and LPS1.0 group were instilled intratracheally with 0.5 mg/kg LPS and 1.0 mg/kg LPS respectively. Mice in control group were instilled intratracheally with LPS-free sterile 0.9% NaCl, whereas normal group received no treatment. The successful chronic lung inflammation murine model was validated via (I) pathological manifestations of chronic inflammatory mononuclear-cell infiltration and lung parenchyma damage; (II) decreased lung function. RESULTS All mice in LPS1.0 group died before the third instillation. No death after instillation was observed in Control and LPS0.5 group. Histological analysis revealed that in LPS0.5 group, 7 days after the third instillation, most bronchus and parabronchial vessels were wrapped by infiltrating monocytes and lymphocyte and alveolar cavities were compressed, which were not observed in control and normal group. Also, ratio of forced expiratory volume in 0.1 second (FEV0.1) and forced vital capacity (FVC) in LPS0.5 group was significantly lower (P<0.0001) than both control group and normal group, suggesting ventilatory dysfunction developed after repeatedly intratracheal instillation once a week for 4 weeks. CONCLUSIONS Intratracheal instillation of 0.5 mg/kg LPS once a week for 4 weeks can cause chronic lung inflammation in young mice.
Collapse
Affiliation(s)
- Kailin Mai
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaowen Chen
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kangkang Zhang
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shujun Gu
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiao Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zihao Gu
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhongji Wu
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kaiyin Huang
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhenwei Liu
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dehui Chen
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
44
|
Wang M, Deng Z, Li Y, Xu K, Ma Y, Yang ST, Wang J. Antibiofilm property and multiple action of peptide PEW300 against Pseudomonas aeruginosa. Front Microbiol 2022; 13:963292. [PMID: 35966656 PMCID: PMC9372277 DOI: 10.3389/fmicb.2022.963292] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/12/2022] [Indexed: 11/24/2022] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa), an opportunistic pathogen, is often associated with difficulties in treating hospital-acquired infections. Biofilms formed by P. aeruginosa significantly improve its resistance to antimicrobial agents, thereby, posing a great challenge to the combat of P. aeruginosa infection. Antimicrobial peptides (AMPs) have recently emerged as promising antibiofilm agents and increasingly attracting the attention of scientists worldwide. However, current knowledge of their antibiofilm behavior is limited and their underlying mechanism remains unclear. In this study, a novel AMP, named PEW300, with three-point mutations (E9H, D17K, and T33A) from Cecropin A was used to investigate its antibiofilm property and antibiofilm pathway against P. aeruginosa. PEW300 displayed strong antibacterial and antibiofilm activity against P. aeruginosa with no significant hemolysis or cytotoxicity to mouse erythrocyte and human embryonic kidney 293 cells. Besides, the antibiofilm pathway results showed that PEW300 preferentially dispersed the mature biofilm, leading to the biofilm-encapsulated bacteria exposure and death. Meanwhile, we also found that the extracellular DNA was a critical target of PEW300 against the mature biofilm of P. aeruginosa. In addition, multiple actions of PEW300 including destroying the cell membrane integrity, inducing high levels of intracellular reactive oxygen species, and interacting with genomic DNA were adopted to exert its antibacterial activity. Moreover, PEW300 could dramatically reduce the virulence of P. aeruginosa. Taken together, PEW300 might be served as a promising antibiofilm candidate to combat P. aeruginosa biofilms.
Collapse
Affiliation(s)
- Meng Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Zifeng Deng
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Yanmei Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Keyong Xu
- Kaiping Healthwise Health Food Co., Ltd, Kaiping, China
| | - Yi Ma
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, South China University of Technology, Guangzhou, China
| | - Shang-Tian Yang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States
| | - Jufang Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, South China University of Technology, Guangzhou, China
- *Correspondence: Jufang Wang,
| |
Collapse
|
45
|
Ceftazidime/Avibactam in Ventilator-Associated Pneumonia Due to Difficult-to-Treat Non-Fermenter Gram-Negative Bacteria in COVID-19 Patients: A Case Series and Review of the Literature. Antibiotics (Basel) 2022; 11:antibiotics11081007. [PMID: 35892396 PMCID: PMC9330655 DOI: 10.3390/antibiotics11081007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
Ventilator-associated pneumonia (VAP) in critically ill patients with COVID-19 represents a very huge global threat due to a higher incidence rate compared to non-COVID-19 patients and almost 50% of the 30-day mortality rate. Pseudomonas aeruginosa was the first pathogen involved but uncommon non-fermenter gram-negative organisms such as Burkholderia cepacea and Stenotrophomonas maltophilia have emerged as other potential etiological causes. Against carbapenem-resistant gram-negative microorganisms, Ceftazidime/avibactam (CZA) is considered a first-line option, even more so in case of a ceftolozane/tazobactam resistance or shortage. The aim of this report was to describe our experience with CZA in a case series of COVID-19 patients hospitalized in the ICU with VAP due to difficult-to-treat (DTT) P. aeruginosa, Burkholderia cepacea, and Stenotrophomonas maltophilia and to compare it with data published in the literature. A total of 23 patients were treated from February 2020 to March 2022: 19/23 (82%) VAPs were caused by Pseudomonas spp. (16/19 DTT), 2 by Burkholderia cepacea, and 6 by Stenotrophomonas maltophilia; 12/23 (52.1%) were polymicrobial. Septic shock was diagnosed in 65.2% of the patients and VAP occurred after a median of 29 days from ICU admission. CZA was prescribed as a combination regimen in 86% of the cases, with either fosfomycin or inhaled amikacin or cotrimoxazole. Microbiological eradication was achieved in 52.3% of the cases and the 30-day overall mortality rate was 14/23 (60.8%). Despite the high mortality of critically ill COVID-19 patients, CZA, especially in combination therapy, could represent a valid treatment option for VAP due to DTT non-fermenter gram-negative bacteria, including uncommon pathogens such as Burkholderia cepacea and Stenotrophomonas maltophilia.
Collapse
|
46
|
Han L, Huang Y, Fu Q, Xue Y, Ding F, Zhang M. Tracking the response to Pseudomonas aeruginosa infection in ozone-induced chronic obstructive pulmonary disease mouse models. Biomed Pharmacother 2022; 150:112980. [PMID: 35453008 DOI: 10.1016/j.biopha.2022.112980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/07/2022] [Accepted: 04/14/2022] [Indexed: 11/30/2022] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is commonly isolated from the sputum of COPD patients. However, the precise role of P. aeruginosa infection in the progression of COPD, especially its role in altering inflammation remains unclear. Here, we designed mice models of COPD infected with P. aeruginosa (PA) and observed dynamic changes of lung structure, lung inflammatory microenvironment, lung function. After infection, the level of mucus secretion peaked on day 3 and remained higher throughout the study period, and the airway remodeling and emphysema was starkly apparent on day 14 and 21. On day 3, interferon-γ and interleukin (IL)- 5 levels increased rapidly, accompanied by elevated T-bet mRNA expression and CD4+T-bet+ cells; at the late stage of infection (days 14 and 21), consistent with increased GATA3 mRNA expression and CD4+GATA3+ cells, IL-4 and IL-13 levels significantly increased; IL-17A level, Foxp3 mRNA expression, CD4+ROR-γt+ cells and CD4+FOXP3+ cells remained at higher levels throughout the course of the infection. Small-airway function showed a decline from day 3 to day 21; large airway function showed a decline on day 14 and 21. Overall, P. aeruginosa infection contributed to the progression of COPD. During the infection, an early Th1-related inflammation gradually shifted to a later Th2-related inflammation, and small-airway function decline occurred earlier than that of large-airway function. On the basis of infection control, the appropriate use of glucocorticoid might slow disease progression by mitigating the enhanced Th2-related inflammation, and small airways could be also an important treatment target in P. aeruginosa -infected COPD patients.
Collapse
Affiliation(s)
- Lei Han
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yuning Huang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Qiang Fu
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yishu Xue
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Fengming Ding
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Min Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| |
Collapse
|
47
|
Eklöf J, Ingebrigtsen TS, Sørensen R, Saeed MI, Alispahic IA, Sivapalan P, Boel JB, Bangsborg J, Ostergaard C, Dessau RB, Jensen US, Hansen EF, Lapperre TS, Meteran H, Wilcke T, Seersholm N, Jensen JUS. Use of inhaled corticosteroids and risk of acquiring Pseudomonas aeruginosa in patients with chronic obstructive pulmonary disease. Thorax 2022; 77:573-580. [PMID: 34446524 PMCID: PMC9120392 DOI: 10.1136/thoraxjnl-2021-217160] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/29/2021] [Indexed: 12/02/2022]
Abstract
BACKGROUND Inhaled corticosteroids (ICS) are commonly used to treat COPD and are associated with increased risk of pneumonia. The aim of this study was to assess if accumulated use of ICS is associated with a dose-dependent risk of a positive airway culture with Pseudomonas aeruginosa in patients with COPD. METHODS We conducted a multiregional epidemiological cohort study including Danish COPD patients followed in outpatient clinics during 2010-2017. ICS use was categorised based on accumulated prescriptions redeemed 365 days prior to cohort entry. Cox proportional hazard regression model was used to estimate the risk of acquiring P. aeruginosa. Propensity score matched models were used as sensitivity analyses. RESULTS A total of 21 408 patients were included in the study, of which 763 (3.6%) acquired P. aeruginosa during follow-up. ICS use was associated with a dose-dependent risk of P. aeruginosa (low ICS dose: HR 1.38, 95% CI 1.03 to 1.84, p=0.03; moderate ICS dose: HR 2.16, 95% CI 1.63 to 2.85, p<0.0001; high ICS dose: HR 3.58, 95% CI 2.75 to 4.65, p<0.0001; reference: no ICS use). A propensity matched model confirmed the results (high ICS dose compared with no/low/moderate ICS dose: HR 2.05, 95% CI 1.76 to 2.39, p p<0.0001). CONCLUSION Use of ICS in patients with COPD followed in Danish outpatient clinics was associated with a substantially increased and dose-dependent risk of acquiring P. aeruginosa. Caution should be taken when administering high doses of ICS in severely ill patients with COPD. These results should be confirmed in comparable cohorts and other settings.
Collapse
Affiliation(s)
- Josefin Eklöf
- Department of Internal Medicine, Section of Respiratory Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | | | - Rikke Sørensen
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Mohamad Isam Saeed
- Department of Internal Medicine, Section of Respiratory Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Imane Achir Alispahic
- Department of Internal Medicine, Section of Respiratory Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Pradeesh Sivapalan
- Department of Internal Medicine, Section of Respiratory Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Internal Medicine, Zealand Hospital, University of Copenhagen, Roskilde, Denmark
| | - Jonas Bredtoft Boel
- Department of Clinical Microbiology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Jette Bangsborg
- Department of Clinical Microbiology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Christian Ostergaard
- Department of Clinical Microbiology, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
| | - Ram Benny Dessau
- Department of Clinical Microbiology, Slagelse Hospital, Slagelse, Denmark
| | - Ulrich Stab Jensen
- Department of Clinical Microbiology, Slagelse Hospital, Slagelse, Denmark
| | - Ejvind Frausing Hansen
- Department of Respiratory Medicine, Amager and Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
| | - Therese Sophie Lapperre
- Department of Respiratory Medicine, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Howraman Meteran
- Department of Internal Medicine, Section of Respiratory Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Torgny Wilcke
- Department of Internal Medicine, Section of Respiratory Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Niels Seersholm
- Department of Internal Medicine, Section of Respiratory Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Jens-Ulrik Stæhr Jensen
- Department of Internal Medicine, Section of Respiratory Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
- PERSIMUNE&CHIP: Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
48
|
Zuhra K, Szabo C. The two faces of cyanide: an environmental toxin and a potential novel mammalian gasotransmitter. FEBS J 2022; 289:2481-2515. [PMID: 34297873 PMCID: PMC9291117 DOI: 10.1111/febs.16135] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/15/2021] [Accepted: 07/22/2021] [Indexed: 12/16/2022]
Abstract
Cyanide is traditionally viewed as a cytotoxic agent, with its primary mode of action being the inhibition of mitochondrial Complex IV (cytochrome c oxidase). However, recent studies demonstrate that the effect of cyanide on Complex IV in various mammalian cells is biphasic: in lower concentrations (nanomolar to low micromolar) cyanide stimulates Complex IV activity, increases ATP production and accelerates cell proliferation, while at higher concentrations (high micromolar to low millimolar) it produces the previously known ('classic') toxic effects. The first part of the article describes the cytotoxic actions of cyanide in the context of environmental toxicology, and highlights pathophysiological conditions (e.g., cystic fibrosis with Pseudomonas colonization) where bacterially produced cyanide exerts deleterious effects to the host. The second part of the article summarizes the mammalian sources of cyanide production and overviews the emerging concept that mammalian cells may produce cyanide, in low concentrations, to serve biological regulatory roles. Cyanide fulfills many of the general criteria as a 'classical' mammalian gasotransmitter and shares some common features with the current members of this class: nitric oxide, carbon monoxide, and hydrogen sulfide.
Collapse
Affiliation(s)
- Karim Zuhra
- Chair of PharmacologySection of MedicineUniversity of FribourgSwitzerland
| | - Csaba Szabo
- Chair of PharmacologySection of MedicineUniversity of FribourgSwitzerland
| |
Collapse
|
49
|
Synergistic Activity of Imipenem in Combination with Ceftazidime/Avibactam or Avibactam against Non-MBL-Producing Extensively Drug-Resistant Pseudomonas aeruginosa. Microbiol Spectr 2022; 10:e0274021. [PMID: 35315696 PMCID: PMC9045292 DOI: 10.1128/spectrum.02740-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Extensively drug-resistant Pseudomonas aeruginosa (XDRPA) infection is a significant public health threat due to a lack of effective therapeutic options. New β-lactam-β-lactamase inhibitor combinations, including ceftazidime-avibactam (CZA), have shown a high resistance rate to XDRPA. This study was therefore conducted to describe the underlying genomic mechanism of resistance for CZA nonsusceptible XDRPA strains that are non-metallo-β-lactamase (MBL) producers as well as to examine synergism of CZA and other antipseudomonal agents. Furthermore, the synergistic antibacterial activity of the most effective antimicrobial combination against non-MBL-producing XDRPA was evaluated through in vitro experiments. The resistance profiles of 15 CZA-resistant XDRPA strains isolated from clinical specimens in China-Japan Friendship Hospital between January 2017 to December 2020 were obtained by whole-genome sequencing (WGS) analysis. MBL genes blaIMP-1 and blaIMP-45 were found in 2 isolates (2/15, 13.3%); the other underlying CZA-resistance mechanisms involved the decreased OprD porin (13/13), blaAmpC overexpression (8/13) or mutation (13/13), and upregulated efflux pumps (13/13). CZA-imipenem (CZA-IPM) combination was identified to be the most effective against non-MBL-producing XDRPA according to the results of WGS analysis and combined antimicrobial susceptibility tests, with an approximately 16.62-fold reduction in MICs compared to CZA alone. Furthermore, the results of checkerboard analysis and growth curve displayed the synergistic antimicrobial activity of CZA and IPM against non-MBL-producing XDRPA. Electron microscopy also revealed that CZA-IPM combination might lead to more cellular structural alterations than CZA or IPM alone. This study suggested that the CZA-IPM combination has potential for non-MBL-producing XDRPA with blaAmpC overexpression or mutation, decreased OprD porin, and upregulated efflux pumps. IMPORTANCE Handling the infections by extensively drug-resistant Pseudomonas aeruginosa (XDRPA) strains is challenging due to their complicated antibiotic resistance mechanisms in immunosuppressed patients with pulmonary diseases (e.g., cystic fibrosis, chronic obstructive pulmonary disease, and lung transplant), ventilator-associated pneumonia, and bloodstream infections. The current study suggested the potentiality of the ceftazidime-avibactam-imipenem combination against XDRPA with blaAmpC overexpression or mutation, decreased OprD porin, and/or upregulated efflux pumps. Our findings indicate the necessity of combined drug sensitivity tests against XDRPA and also lay a foundation for the development of prevention, control, and treatment strategies in XDRPA infections.
Collapse
|
50
|
Li S, Yu C, Jie H, Han X, Zou S, Tan Q, Luo S, Chen Y, Wang J. Neutrophil side fluorescence: a new indicator for predicting the severity of patients with bronchiectasis. BMC Pulm Med 2022; 22:107. [PMID: 35346147 PMCID: PMC8962496 DOI: 10.1186/s12890-022-01893-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 03/09/2022] [Indexed: 12/01/2022] Open
Abstract
Background Neutrophilic inflammation in the airway is a hallmark of bronchiectasis. Neutrophil extracellular traps (NETs) have been reported to play an important role in the occurrence and development of bronchiectasis. Neutrophil side fluorescence is one of the characteristics of neutrophils that can reflect the activation of neutrophils and the formation of NETs. Objective To explore the relationship between the values of neutrophil side fluorescence (NEUT-SFL) in the peripheral blood of bronchiectasis patients, and the severity of the disease. Methods 82 patients with bronchiectasis from the Department of Respiratory and Critical Medicine, at the Third Affiliated Hospital of Southern Medical University and were scored with Bronchiectasis Severity Index (BSI) (2019–2021). The clinical data such as the value of NEUT-SFL, neutrophil count, C-reactive protein, and procalcitonin levels were collected and retrospectively analyzed. NEUT-SFL values neutrophil count from 28 healthy subjects were also used to ascertain cut-off values. Results Based on the BSI scores, patients were divided into three categories as mild (32%), moderate (29%), and severe (39%). Our results showed that the values of NEUT-SFL were higher in bronchiectasis patients compared to healthy controls. The levels of NEUT-SFL positively correlated with the high BSI scores in patients (P = 0.037, r = 0.23) and negatively correlated with the lung function in these patients (r = − 0.35, P = 0.001). The area under the ROC curve was 0.813, the best cut-off was 42.145, indicating that NEUT-SFL values > 42.145 can potentially predict the severity of bronchiectasis. Conclusions The values of NEUT-SFL in the peripheral blood can be used for predicting the severity of bronchiectasis.
Collapse
Affiliation(s)
- Shiqi Li
- Department of Respiration, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Chunxiao Yu
- Department of Gastroenterology, The Seventh Affiliated Hospital, Southern Medical University, Foshan, China
| | - Hongyu Jie
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Xinai Han
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Shujing Zou
- Department of Respiration, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Quanguang Tan
- Department of Respiration, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Shugeng Luo
- Department of Internal Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Youming Chen
- Department of Clinical Laboratory, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.
| | - Jinhong Wang
- Department of Respiration, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|