151
|
Shen L, Gao L, Yang M, Zhang J, Wang Y, Feng Y, Wang L, Wang S. Deletion of the PA4427-PA4431 Operon of Pseudomonas aeruginosa PAO1 Increased Antibiotics Resistance and Reduced Virulence and Pathogenicity by Affecting Quorum Sensing and Iron Uptake. Microorganisms 2021; 9:microorganisms9051065. [PMID: 34069209 PMCID: PMC8156433 DOI: 10.3390/microorganisms9051065] [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: 04/13/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/06/2022] Open
Abstract
The respiratory chain is very important for bacterial survival and pathogenicity, yet the roles of the respiratory chain in P. aeruginosa remain to be fully elucidated. Here, we not only proved experimentally that the operon PA4427-PA4431 of Pseudomonas aeruginosa PAO1 encodes respiratory chain complex III (cytobc1), but also found that it played important roles in virulence and pathogenicity. PA4429–31 deletion reduced the production of the virulence factors, including pyocyanin, rhamnolipids, elastase, and extracellular polysaccharides, and it resulted in a remarkable decrease in pathogenicity, as demonstrated in the cabbage and Drosophila melanogaster infection models. Furthermore, RNA-seq analysis showed that PA4429–31 deletion affected the expression levels of the genes related to quorum-sensing systems and the transport of iron ions, and the iron content was also reduced in the mutant strain. Taken together, we comprehensively illustrated the function of the operon PA4427–31 and its application potential as a treatment target in P. aeruginosa infection.
Collapse
|
152
|
Rybenkov VV, Zgurskaya HI, Ganguly C, Leus IV, Zhang Z, Moniruzzaman M. The Whole Is Bigger than the Sum of Its Parts: Drug Transport in the Context of Two Membranes with Active Efflux. Chem Rev 2021; 121:5597-5631. [PMID: 33596653 PMCID: PMC8369882 DOI: 10.1021/acs.chemrev.0c01137] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cell envelope plays a dual role in the life of bacteria by simultaneously protecting it from a hostile environment and facilitating access to beneficial molecules. At the heart of this ability lie the restrictive properties of the cellular membrane augmented by efflux transporters, which preclude intracellular penetration of most molecules except with the help of specialized uptake mediators. Recently, kinetic properties of the cell envelope came into focus driven on one hand by the urgent need in new antibiotics and, on the other hand, by experimental and theoretical advances in studies of transmembrane transport. A notable result from these studies is the development of a kinetic formalism that integrates the Michaelis-Menten behavior of individual transporters with transmembrane diffusion and offers a quantitative basis for the analysis of intracellular penetration of bioactive compounds. This review surveys key experimental and computational approaches to the investigation of transport by individual translocators and in whole cells, summarizes key findings from these studies and outlines implications for antibiotic discovery. Special emphasis is placed on Gram-negative bacteria, whose envelope contains two separate membranes. This feature sets these organisms apart from Gram-positive bacteria and eukaryotic cells by providing them with full benefits of the synergy between slow transmembrane diffusion and active efflux.
Collapse
Affiliation(s)
- Valentin V Rybenkov
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Helen I Zgurskaya
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Chhandosee Ganguly
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Inga V Leus
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Zhen Zhang
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Mohammad Moniruzzaman
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| |
Collapse
|
153
|
Mayeux G, Gayet L, Liguori L, Odier M, Martin DK, Cortès S, Schaack B, Lenormand JL. Cell-free expression of the outer membrane protein OprF of Pseudomonas aeruginosa for vaccine purposes. Life Sci Alliance 2021; 4:4/6/e202000958. [PMID: 33972378 PMCID: PMC8127326 DOI: 10.26508/lsa.202000958] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 11/24/2022] Open
Abstract
Production of recombinant proteoliposomes containing OprF from P. aeruginosa promotes the active open conformation of the porin exposing native epitopes. These OprF proteoliposomes were used as vaccines to protect mice against a P. aeruginosa acute pulmonary infection model. Pseudomonas aeruginosa is the second-leading cause of nosocomial infections and pneumonia in hospitals. Because of its extraordinary capacity for developing resistance to antibiotics, treating infections by Pseudomonas is becoming a challenge, lengthening hospital stays, and increasing medical costs and mortality. The outer membrane protein OprF is a well-conserved and immunogenic porin playing an important role in quorum sensing and in biofilm formation. Here, we used a bacterial cell-free expression system to reconstitute OprF under its native forms in liposomes and we demonstrated that the resulting OprF proteoliposomes can be used as a fully functional recombinant vaccine against P. aeruginosa. Remarkably, we showed that our system promotes the folding of OprF into its active open oligomerized state as well as the formation of mega-pores. Our approach thus represents an easy and efficient way for producing bacterial membrane antigens exposing native epitopes for vaccine purposes.
Collapse
Affiliation(s)
- Géraldine Mayeux
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France
| | - Landry Gayet
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France
| | - Lavinia Liguori
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France.,Maison Familiale Rurale Moirans, Moirans, France
| | - Marine Odier
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France.,Catalent Pharma Solutions, Eberbach, Germany
| | - Donald K Martin
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France
| | | | - Béatrice Schaack
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France.,University Grenoble Alpes, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), CNRS, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Jean-Luc Lenormand
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France
| |
Collapse
|
154
|
Pelegrin AC, Palmieri M, Mirande C, Oliver A, Moons P, Goossens H, van Belkum A. Pseudomonas aeruginosa: a clinical and genomics update. FEMS Microbiol Rev 2021; 45:6273131. [PMID: 33970247 DOI: 10.1093/femsre/fuab026] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 05/07/2021] [Indexed: 12/13/2022] Open
Abstract
Antimicrobial resistance (AMR) has become a global medical priority that needs urgent resolution. Pseudomonas aeruginosa is a versatile, adaptable bacterial species with widespread environmental occurrence, strong medical relevance, a diverse set of virulence genes and a multitude of intrinsic and possibly acquired antibiotic resistance traits. P. aeruginosa causes a wide variety of infections and has an epidemic-clonal population structure. Several of its dominant global clones have collected a wide variety of resistance genes rendering them multi-drug resistant (MDR) and particularly threatening groups of vulnerable individuals including surgical patients, immunocompromised patients, Caucasians suffering from cystic fibrosis (CF) and more. AMR and MDR especially are particularly problematic in P. aeruginosa significantly complicating successful antibiotic treatment. In addition, antimicrobial susceptibility testing (AST) of P. aeruginosa can be cumbersome due to its slow growth or the massive production of exopolysaccharides and other extracellular compounds. For that reason, phenotypic AST is progressively challenged by genotypic methods using whole genome sequences (WGS) and large-scale phenotype databases as a framework of reference. We here summarize the state of affairs and the quality level of WGS-based AST for P. aeruginosa mostly from clinical origin.
Collapse
Affiliation(s)
- Andreu Coello Pelegrin
- bioMérieux, Data Analytics Unit, 3 Route du Port Michaud, 38390 La Balme les Grottes, France
| | - Mattia Palmieri
- bioMérieux, Data Analytics Unit, 3 Route du Port Michaud, 38390 La Balme les Grottes, France
| | - Caroline Mirande
- bioMérieux, R&D Microbiology, Route du Port Michaud, 38390 La Balme-les-Grottes, France
| | - Antonio Oliver
- Servicio de Microbiología, Módulo J, segundo piso, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria Illes Balears (IdISBa), Ctra. Valldemossa, 79, 07120 Palma de Mallorca, Spain
| | - Pieter Moons
- Laboratory of Medical Microbiology, University of Antwerp, Universiteitsplein 1, building S, 2610 Wilrijk, Antwerp, Belgium
| | - Herman Goossens
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Alex van Belkum
- bioMérieux, Open Innovation and Partnerships, 3 Route du Port Michaud, 38390 La Balme Les Grottes, France
| |
Collapse
|
155
|
Vázquez-Ucha JC, Rodríguez D, Lasarte-Monterrubio C, Lence E, Arca-Suarez J, Maneiro M, Gato E, Perez A, Martínez-Guitián M, Juan C, Oliver A, Bou G, González-Bello C, Beceiro A. 6-Halopyridylmethylidene Penicillin-Based Sulfones Efficiently Inactivate the Natural Resistance of Pseudomonas aeruginosa to β-Lactam Antibiotics. J Med Chem 2021; 64:6310-6328. [PMID: 33913328 DOI: 10.1021/acs.jmedchem.1c00369] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pseudomonas aeruginosa, a major cause of nosocomial infections, is considered a paradigm of antimicrobial resistance, largely due to hyperproduction of chromosomal cephalosporinase AmpC. Here, we explore the ability of 6-pyridylmethylidene penicillin-based sulfones 1-3 to inactivate the AmpC β-lactamase and thus rescue the activity of the antipseudomonal ceftazidime. These compounds increased the susceptibility to ceftazidime in a collection of clinical isolates and PAO1 mutant strains with different ampC expression levels and also improved the inhibition kinetics relative to avibactam, displaying a slow deacylation rate and involving the formation of an indolizine adduct. Bromide 2 was the inhibitor with the lowest KI (15.6 nM) and the highest inhibitory efficiency (kinact/KI). Computational studies using diverse AmpC enzymes revealed that the aromatic moiety in 1-3 targets a tunnel-like site adjacent to the catalytic serine and induces the folding of the H10 helix, indicating the potential value of this not-always-evident pocket in drug design.
Collapse
Affiliation(s)
- Juan C Vázquez-Ucha
- Servicio de Microbiología do Complejo Hospitalario Universitario da Coruña (CHUAC), Instituto de Investigación Biomédica da Coruña (CICA-INIBIC), Xubias de Arriba, 84, A Coruña 15006, Spain
| | - Diana Rodríguez
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, Santiago de Compostela 15782, Spain
| | - Cristina Lasarte-Monterrubio
- Servicio de Microbiología do Complejo Hospitalario Universitario da Coruña (CHUAC), Instituto de Investigación Biomédica da Coruña (CICA-INIBIC), Xubias de Arriba, 84, A Coruña 15006, Spain
| | - Emilio Lence
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, Santiago de Compostela 15782, Spain
| | - Jorge Arca-Suarez
- Servicio de Microbiología do Complejo Hospitalario Universitario da Coruña (CHUAC), Instituto de Investigación Biomédica da Coruña (CICA-INIBIC), Xubias de Arriba, 84, A Coruña 15006, Spain
| | - María Maneiro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, Santiago de Compostela 15782, Spain
| | - Eva Gato
- Servicio de Microbiología do Complejo Hospitalario Universitario da Coruña (CHUAC), Instituto de Investigación Biomédica da Coruña (CICA-INIBIC), Xubias de Arriba, 84, A Coruña 15006, Spain
| | - Astrid Perez
- Servicio de Microbiología do Complejo Hospitalario Universitario da Coruña (CHUAC), Instituto de Investigación Biomédica da Coruña (CICA-INIBIC), Xubias de Arriba, 84, A Coruña 15006, Spain
| | - Marta Martínez-Guitián
- Servicio de Microbiología do Complejo Hospitalario Universitario da Coruña (CHUAC), Instituto de Investigación Biomédica da Coruña (CICA-INIBIC), Xubias de Arriba, 84, A Coruña 15006, Spain
| | - Carlos Juan
- Servicio de Microbiología y Unidad de Investigación, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria Illes Balears (IdiSBA), Carretera de Valldemossa, 79, Palma de Mallorca 07120, Spain
| | - Antonio Oliver
- Servicio de Microbiología y Unidad de Investigación, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria Illes Balears (IdiSBA), Carretera de Valldemossa, 79, Palma de Mallorca 07120, Spain
| | - German Bou
- Servicio de Microbiología do Complejo Hospitalario Universitario da Coruña (CHUAC), Instituto de Investigación Biomédica da Coruña (CICA-INIBIC), Xubias de Arriba, 84, A Coruña 15006, Spain
| | - Concepción González-Bello
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, Santiago de Compostela 15782, Spain
| | - Alejandro Beceiro
- Servicio de Microbiología do Complejo Hospitalario Universitario da Coruña (CHUAC), Instituto de Investigación Biomédica da Coruña (CICA-INIBIC), Xubias de Arriba, 84, A Coruña 15006, Spain
| |
Collapse
|
156
|
Langendonk RF, Neill DR, Fothergill JL. The Building Blocks of Antimicrobial Resistance in Pseudomonas aeruginosa: Implications for Current Resistance-Breaking Therapies. Front Cell Infect Microbiol 2021; 11:665759. [PMID: 33937104 PMCID: PMC8085337 DOI: 10.3389/fcimb.2021.665759] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
P. aeruginosa is classified as a priority one pathogen by the World Health Organisation, and new drugs are urgently needed, due to the emergence of multidrug-resistant (MDR) strains. Antimicrobial-resistant nosocomial pathogens such as P. aeruginosa pose unwavering and increasing threats. Antimicrobial stewardship has been a challenge during the COVID-19 pandemic, with a majority of those hospitalized with SARS-CoV2 infection given antibiotics as a safeguard against secondary bacterial infection. This increased usage, along with increased handling of sanitizers and disinfectants globally, may further accelerate the development and spread of cross-resistance to antibiotics. In addition, P. aeruginosa is the primary causative agent of morbidity and mortality in people with the life-shortening genetic disease cystic fibrosis (CF). Prolonged periods of selective pressure, associated with extended antibiotic treatment and the actions of host immune effectors, results in widespread adaptive and acquired resistance in P. aeruginosa found colonizing the lungs of people with CF. This review discusses the arsenal of resistance mechanisms utilized by P. aeruginosa, how these operate under high-stress environments such as the CF lung and how their interconnectedness can result in resistance to multiple antibiotic classes. Intrinsic, adaptive and acquired resistance mechanisms will be described, with a focus on how each layer of resistance can serve as a building block, contributing to multi-tiered resistance to antimicrobial activity. Recent progress in the development of anti-resistance adjuvant therapies, targeting one or more of these building blocks, should lead to novel strategies for combatting multidrug resistant P. aeruginosa. Anti-resistance adjuvant therapy holds great promise, not least because resistance against such therapeutics is predicted to be rare. The non-bactericidal nature of anti-resistance adjuvants reduce the selective pressures that drive resistance. Anti-resistance adjuvant therapy may also be advantageous in facilitating efficacious use of traditional antimicrobials, through enhanced penetration of the antibiotic into the bacterial cell. Promising anti-resistance adjuvant therapeutics and targets will be described, and key remaining challenges highlighted. As antimicrobial stewardship becomes more challenging in an era of emerging and re-emerging infectious diseases and global conflict, innovation in antibiotic adjuvant therapy can play an important role in extending the shelf-life of our existing antimicrobial therapeutic agents.
Collapse
Affiliation(s)
- R. Frèdi Langendonk
- Institute of Infection, Veterinary and Ecological Science, University of Liverpool, Liverpool, United Kingdom
| | | | | |
Collapse
|
157
|
A Novel Infection Protocol in Zebrafish Embryo to Assess Pseudomonas aeruginosa Virulence and Validate Efficacy of a Quorum Sensing Inhibitor In Vivo. Pathogens 2021; 10:pathogens10040401. [PMID: 33805384 PMCID: PMC8065929 DOI: 10.3390/pathogens10040401] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/23/2022] Open
Abstract
The opportunistic human pathogen Pseudomonas aeruginosa is responsible for a variety of acute infections and is a major cause of mortality in chronically infected cystic fibrosis patients. Due to increased resistance to antibiotics, new therapeutic strategies against P. aeruginosa are urgently needed. In this context, we aimed to develop a simple vertebrate animal model to rapidly assess in vivo drug efficacy against P. aeruginosa. Zebrafish are increasingly considered for modeling human infections caused by bacterial pathogens, which are commonly microinjected in embryos. In the present study, we established a novel protocol for zebrafish infection by P. aeruginosa based on bath immersion in 96-well plates of tail-injured embryos. The immersion method, followed by a 48-hour survey of embryo viability, was first validated to assess the virulence of P. aeruginosa wild-type PAO1 and a known attenuated mutant. We then validated its relevance for antipseudomonal drug testing by first using a clinically used antibiotic, ciprofloxacin. Secondly, we used a novel quorum sensing (QS) inhibitory molecule, N-(2-pyrimidyl)butanamide (C11), the activity of which had been validated in vitro but not previously tested in any animal model. A significant protective effect of C11 was observed on infected embryos, supporting the ability of C11 to attenuate in vivo P. aeruginosa pathogenicity. In conclusion, we present here a new and reliable method to compare the virulence of P. aeruginosa strains in vivo and to rapidly assess the efficacy of clinically relevant drugs against P. aeruginosa, including new antivirulence compounds.
Collapse
|
158
|
Jurado-Martín I, Sainz-Mejías M, McClean S. Pseudomonas aeruginosa: An Audacious Pathogen with an Adaptable Arsenal of Virulence Factors. Int J Mol Sci 2021; 22:3128. [PMID: 33803907 PMCID: PMC8003266 DOI: 10.3390/ijms22063128] [Citation(s) in RCA: 265] [Impact Index Per Article: 88.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
Pseudomonas aeruginosa is a dominant pathogen in people with cystic fibrosis (CF) contributing to morbidity and mortality. Its tremendous ability to adapt greatly facilitates its capacity to cause chronic infections. The adaptability and flexibility of the pathogen are afforded by the extensive number of virulence factors it has at its disposal, providing P. aeruginosa with the facility to tailor its response against the different stressors in the environment. A deep understanding of these virulence mechanisms is crucial for the design of therapeutic strategies and vaccines against this multi-resistant pathogen. Therefore, this review describes the main virulence factors of P. aeruginosa and the adaptations it undergoes to persist in hostile environments such as the CF respiratory tract. The very large P. aeruginosa genome (5 to 7 MB) contributes considerably to its adaptive capacity; consequently, genomic studies have provided significant insights into elucidating P. aeruginosa evolution and its interactions with the host throughout the course of infection.
Collapse
Affiliation(s)
| | | | - Siobhán McClean
- School of Biomolecular and Biomedical Sciences, University College Dublin, Belfield, Dublin 4 D04 V1W8, Ireland; (I.J.-M.); (M.S.-M.)
| |
Collapse
|
159
|
Mojsoska B, Ghoul M, Perron GG, Jenssen H, Alatraktchi FA. Changes in toxin production of environmental Pseudomonas aeruginosa isolates exposed to sub-inhibitory concentrations of three common antibiotics. PLoS One 2021; 16:e0248014. [PMID: 33662048 PMCID: PMC7932067 DOI: 10.1371/journal.pone.0248014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/17/2021] [Indexed: 12/02/2022] Open
Abstract
Pseudomonas aeruginosa is an environmental pathogen that can cause severe infections in immunocompromised patients. P. aeruginosa infections are typically treated with multiple antibiotics including tobramycin, ciprofloxacin, and meropenem. However, antibiotics do not always entirely clear the bacteria from the infection site, where they may remain virulent. This is because the effective antibiotic concentration and diffusion in vitro may differ from the in vivo environment in patients. Therefore, it is important to understand the effect of non-lethal sub-inhibitory antibiotic concentrations on bacterial phenotype. Here, we investigate if sub-inhibitory antimicrobial concentrations cause alterations in bacterial virulence factor production using pyocyanin as a model toxin. We tested this using the aforementioned antibiotics on 10 environmental P. aeruginosa strains. Using on-the-spot electrochemical screening, we were able to directly quantify changes in production of pyocyanin in a measurement time of 17 seconds. Upon selecting 3 representative strains to further test the effects of sub-minimum inhibitory concentration (MICs), we found that pyocyanin production changed significantly when the bacteria were exposed to 10-fold MIC of the 3 antibiotics tested, and this was strain specific. A series of biologically relevant measured pyocyanin concentrations were also used to assess the effects of increased virulence on a culture of epithelial cells. We found a decreased viability of the epithelial cells when incubated with biologically relevant pyocyanin concentrations. This suggests that the antibiotic-induced virulence also is a value worth being enclosed in regular testing of pathogens.
Collapse
Affiliation(s)
- Biljana Mojsoska
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
- PreDiagnose, Karlslunde, Denmark
| | - Melanie Ghoul
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Gabriel G. Perron
- Department of Biology, Bard College, Annandale-On-Hudson, NY, United States of America
| | - Håvard Jenssen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | | |
Collapse
|
160
|
Paulsson M, Kragh KN, Su YC, Sandblad L, Singh B, Bjarnsholt T, Riesbeck K. Peptidoglycan-Binding Anchor Is a Pseudomonas aeruginosa OmpA Family Lipoprotein With Importance for Outer Membrane Vesicles, Biofilms, and the Periplasmic Shape. Front Microbiol 2021; 12:639582. [PMID: 33717034 PMCID: PMC7947798 DOI: 10.3389/fmicb.2021.639582] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/28/2021] [Indexed: 01/02/2023] Open
Abstract
The outer membrane protein A (OmpA) family contains an evolutionary conserved domain that links the outer membrane in Gram-negative bacteria to the semi-rigid peptidoglycan (PG) layer. The clinically significant pathogen Pseudomonas aeruginosa carries several OmpA family proteins (OprF, OprL, PA0833, and PA1048) that share the PG-binding domain. These proteins are important for cell morphology, membrane stability, and biofilm and outer membrane vesicle (OMV) formation. In addition to other OmpAs, in silico analysis revealed that the putative outer membrane protein (OMP) with gene locus PA1041 is a lipoprotein with an OmpA domain and, hence, is a potential virulence factor. This study aimed to evaluate PA1041 as a PG-binding protein and describe its effect on the phenotype. Clinical strains were confirmed to contain the lipoprotein resulting from PA1041 expression with Western blot, and PG binding was verified in enzyme-linked immunosorbent assay (ELISA). By using a Sepharose bead-based ELISA, we found that the lipoprotein binds to meso-diaminopimelic acid (mDAP), an amino acid in the pentapeptide portion of PGs. The reference strain PAO1 and the corresponding transposon mutant PW2884 devoid of the lipoprotein were examined for phenotypic changes. Transmission electron microscopy revealed enlarged periplasm spaces near the cellular poles in the mutant. In addition, we observed an increased release of OMV, which could be confirmed by nanoparticle tracking analysis. Importantly, mutants without the lipoprotein produced a thick, but loose and unorganized, biofilm in flow cells. In conclusion, the lipoprotein from gene locus PA1041 tethers the outer membrane to the PG layer, and mutants are viable, but display severe phenotypic changes including disordered biofilm formation. Based upon the phenotype of the P. aeruginosa PW2884 mutant and the function of the protein, we designate the lipoprotein with locus tag PA1041 as “peptidoglycan-binding anchor” (Pba).
Collapse
Affiliation(s)
- Magnus Paulsson
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden.,Division for Infectious Diseases, Skåne University Hospital, Lund, Sweden
| | - Kasper Nørskov Kragh
- Faculty of Health and Medical Sciences, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Yu-Ching Su
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Linda Sandblad
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Birendra Singh
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Thomas Bjarnsholt
- Faculty of Health and Medical Sciences, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kristian Riesbeck
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
161
|
Askarian F, Uchiyama S, Masson H, Sørensen HV, Golten O, Bunæs AC, Mekasha S, Røhr ÅK, Kommedal E, Ludviksen JA, Arntzen MØ, Schmidt B, Zurich RH, van Sorge NM, Eijsink VGH, Krengel U, Mollnes TE, Lewis NE, Nizet V, Vaaje-Kolstad G. The lytic polysaccharide monooxygenase CbpD promotes Pseudomonas aeruginosa virulence in systemic infection. Nat Commun 2021; 12:1230. [PMID: 33623002 PMCID: PMC7902821 DOI: 10.1038/s41467-021-21473-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 01/29/2021] [Indexed: 12/20/2022] Open
Abstract
The recently discovered lytic polysaccharide monooxygenases (LPMOs), which cleave polysaccharides by oxidation, have been associated with bacterial virulence, but supporting functional data is scarce. Here we show that CbpD, the LPMO of Pseudomonas aeruginosa, is a chitin-oxidizing virulence factor that promotes survival of the bacterium in human blood. The catalytic activity of CbpD was promoted by azurin and pyocyanin, two redox-active virulence factors also secreted by P. aeruginosa. Homology modeling, molecular dynamics simulations, and small angle X-ray scattering indicated that CbpD is a monomeric tri-modular enzyme with flexible linkers. Deletion of cbpD rendered P. aeruginosa unable to establish a lethal systemic infection, associated with enhanced bacterial clearance in vivo. CbpD-dependent survival of the wild-type bacterium was not attributable to dampening of pro-inflammatory responses by CbpD ex vivo or in vivo. Rather, we found that CbpD attenuates the terminal complement cascade in human serum. Studies with an active site mutant of CbpD indicated that catalytic activity is crucial for virulence function. Finally, profiling of the bacterial and splenic proteomes showed that the lack of this single enzyme resulted in substantial re-organization of the bacterial and host proteomes. LPMOs similar to CbpD occur in other pathogens and may have similar immune evasive functions.
Collapse
Affiliation(s)
- Fatemeh Askarian
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway.
| | - Satoshi Uchiyama
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego, La Jolla, CA, USA
| | - Helen Masson
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | | | - Ole Golten
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Anne Cathrine Bunæs
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Sophanit Mekasha
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Åsmund Kjendseth Røhr
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Eirik Kommedal
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | | | - Magnus Ø Arntzen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Benjamin Schmidt
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego, La Jolla, CA, USA
| | - Raymond H Zurich
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego, La Jolla, CA, USA
| | - Nina M van Sorge
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Netherlands Reference Laboratory for Bacterial Meningitis, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Vincent G H Eijsink
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Ute Krengel
- Department of Chemistry, University of Oslo, Oslo, Norway
| | - Tom Eirik Mollnes
- Research Laboratory, Nordland Hospital, Bodø, Norway
- K.G. Jebsen TREC, Faculty of Health Sciences, UiT- The Arctic University of Norway, Tromsø, Norway
- Department of Immunology, Oslo University Hospital, and K.G. Jebsen IRC, University of Oslo, Oslo, Norway
- Center of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nathan E Lewis
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA, USA
- Novo Nordisk Foundation Center for Biosustainability at UC San Diego, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Victor Nizet
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego, La Jolla, CA, USA.
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA, USA.
| | - Gustav Vaaje-Kolstad
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway.
| |
Collapse
|
162
|
Xu C, Cao Q, Lan L. Glucose-Binding of Periplasmic Protein GltB Activates GtrS-GltR Two-Component System in Pseudomonas aeruginosa. Microorganisms 2021; 9:447. [PMID: 33670077 PMCID: PMC7927077 DOI: 10.3390/microorganisms9020447] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 12/14/2022] Open
Abstract
A two-component system GtrS-GltR is required for glucose transport activity in P. aeruginosa and plays a key role during P. aeruginosa-host interactions. However, the mechanism of action of GtrS-GltR has not been definitively established. Here, we show that gltB, which encodes a periplasmic glucose binding protein, is essential for the glucose-induced activation of GtrS-GltR in P. aeruginosa. We determined that GltB is capable of binding to membrane regulatory proteins including GtrS, the sensor kinase of the GtrS-GltR TCS. We observed that alanine substitution of glucose-binding residues abolishes the ability of GltB to promote the activation of GtrS-GltR. Importantly, like the gtrS deletion mutant, gltB deletion mutant showed attenuated virulence in both Drosophila melanogaster and mouse models of infection. In addition, using CHIP-seq experiments, we showed that the promoter of gltB is the major in vivo target of GltR. Collectively, these data suggest that periplasmic binding protein GltB and GtrS-GltR TCS form a complex regulatory circuit that regulates the virulence of P. aeruginosa in response to glucose.
Collapse
Affiliation(s)
- Chenchen Xu
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China;
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China;
| | - Qiao Cao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China;
| | - Lefu Lan
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China;
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China;
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- NMPA Key Laboratory for Testing Technology of Pharmaceutical Microbiology, Shanghai Institute for Food and Drug Control, Shanghai 201203, China
| |
Collapse
|
163
|
van Haren MJ, Tehrani KHME, Kotsogianni I, Wade N, Brüchle NC, Mashayekhi V, Martin NI. Cephalosporin Prodrug Inhibitors Overcome Metallo-β-Lactamase Driven Antibiotic Resistance. Chemistry 2021; 27:3806-3811. [PMID: 33237604 PMCID: PMC7986777 DOI: 10.1002/chem.202004694] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/24/2020] [Indexed: 12/21/2022]
Abstract
The increasing prevalence of metallo-β-lactamase (MBL)-expressing bacteria presents a worrying trend in antibiotic resistance. MBLs rely on active site zinc ions for their hydrolytic activity and the pursuit of MBL-inhibitors has therefore involved the investigation of zinc chelators. To ensure that such chelators specifically target MBLs, a series of cephalosporin prodrugs of two potent zinc-binders: dipicolinic acid (DPA) and 8-thioquinoline (8-TQ) was prepared. Although both DPA and 8-TQ bind free zinc very tightly (Kd values in the low nm range), the corresponding cephalosporin conjugates do not. The cephalosporin conjugates are efficiently hydrolyzed by MBLs to release DPA or 8-TQ, as confirmed by using both NMR and LC-MS studies. Notably, the cephalosporin prodrugs of DPA and 8-TQ show potent inhibitory activity against NDM, VIM, and IMP classes of MBLs and display potent synergy with meropenem against MBL-expressing clinical isolates of K. pneumoniae and E. coli.
Collapse
Affiliation(s)
- Matthijs J. van Haren
- Biological Chemistry GroupInstitute of Biology LeidenLeiden UniversitySylviusweg 722333 BELeidenThe Netherlands
| | - Kamaleddin H. M. E. Tehrani
- Biological Chemistry GroupInstitute of Biology LeidenLeiden UniversitySylviusweg 722333 BELeidenThe Netherlands
| | - Ioli Kotsogianni
- Biological Chemistry GroupInstitute of Biology LeidenLeiden UniversitySylviusweg 722333 BELeidenThe Netherlands
| | - Nicola Wade
- Biological Chemistry GroupInstitute of Biology LeidenLeiden UniversitySylviusweg 722333 BELeidenThe Netherlands
| | - Nora C. Brüchle
- Biological Chemistry GroupInstitute of Biology LeidenLeiden UniversitySylviusweg 722333 BELeidenThe Netherlands
| | - Vida Mashayekhi
- Department of BiologyUtrecht UniversityUniversiteitsweg 993584 CGUtrechtThe Netherlands
| | - Nathaniel I. Martin
- Biological Chemistry GroupInstitute of Biology LeidenLeiden UniversitySylviusweg 722333 BELeidenThe Netherlands
| |
Collapse
|
164
|
Piselli C, Benz R. Fosmidomycin transport through the phosphate-specific porins OprO and OprP of Pseudomonas aeruginosa. Mol Microbiol 2021; 116:97-108. [PMID: 33561903 DOI: 10.1111/mmi.14693] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 01/08/2023]
Abstract
The Gram-negative bacterium Pseudomonas aeruginosa is an opportunistic pathogen, responsible for many hospital-acquired infections. The bacterium is quite resistant toward many antibiotics, in particular because of the fine-tuned permeability of its outer membrane (OM). General diffusion outer membrane pores are quite rare in this organism. Instead, its OM contains many substrate-specific porins. Their expression is varying according to growth conditions and virulence. Phosphate limitations, as well as pathogenicity factors, result in the induction of the two mono- and polyphosphate-specific porins, OprP and OprO, respectively, together with an inner membrane uptake mechanism and a periplasmic binding protein. These outer membrane channels could serve as outer membrane pathways for the uptake of phosphonates. Among them are not only herbicides, but also potent antibiotics, such as fosfomycin and fosmidomycin. In this study, we investigated the interaction between OprP and OprO and fosmidomycin in detail. We could demonstrate that fosmidomycin is able to bind to the phosphate-specific binding site inside the two porins. The inhibition of chloride conductance of OprP and OprO by fosmidomycin is considerably less than that of phosphate or diphosphate, but it can be measured in titration experiments of chloride conductance and also in single-channel experiments. The results suggest that fosmidomycin transport across the OM of P. aeruginosa occurs through OprP and OprO. Our data with the ones already known in the literature show that phosphonic acid-containing antibiotics are in general good candidates to treat the infections of P. aeruginosa at the very beginning through a favorable OM transport system.
Collapse
Affiliation(s)
- Claudio Piselli
- Department of Life Sciences and Chemistry, Focus Health, Jacobs University Bremen, Bremen, Germany
| | - Roland Benz
- Rudolf-Virchow-Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| |
Collapse
|
165
|
González-Vázquez MC, Rocha-Gracia RDC, Carabarín-Lima A, Bello-López E, Huerta-Romano F, Martínez-Laguna Y, Lozano-Zarain P. Location of OprD porin in Pseudomonas aeruginosa clinical isolates. APMIS 2021; 129:213-224. [PMID: 33471435 DOI: 10.1111/apm.13118] [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: 05/11/2020] [Accepted: 01/12/2021] [Indexed: 11/28/2022]
Abstract
Multidrug-resistant Pseudomonas aeruginosa is one of the main opportunistic pathogens causing severe infection. One of the mechanisms involved in the resistance to imipenem in clinical isolates is the loss of the OprD porin. Changes like substitutions, deletions, insertions, or mutations in the oprD gene can modify the conformation of OprD porin or inhibit its presence and generate resistance to carbapenems. The aim of this work was to obtain anti-OprD polyclonal antibodies and to determine by both immunofluorescence microscopy (IFI) and Western blot assays, the presence of the OprD porin in resistant-carbapenem P. aeruginosa strains with different changes in the oprD gene. Changes in the gene oprD were identified in clinical isolates of P. aeruginosa. When proteins were translated, several polymorphisms were found; however, these did not affect the presence of OprD porin (PCM25, PCM36, and PCM78). Also it was detected an insertion sequence ISPa1328 (PCM52) and a premature stop codon (PCM91), which inhibited the presence of the OprD porin. This study shows how changes in the oprD gene of P. aeruginosa clinical isolates affect the presence of the OprD porin detected by Western blot and indirect immunofluorescence assays using specific polyclonal anti-OprD antibodies generated in this work.
Collapse
Affiliation(s)
- María Cristina González-Vázquez
- Instituto de Ciencias, Centro de Investigaciones de Ciencias Microbiológicas, Posgrado en Microbiología, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Rosa Del Carmen Rocha-Gracia
- Instituto de Ciencias, Centro de Investigaciones de Ciencias Microbiológicas, Posgrado en Microbiología, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Alejandro Carabarín-Lima
- Instituto de Ciencias, Centro de Investigaciones de Ciencias Microbiológicas, Posgrado en Microbiología, Benemérita Universidad Autónoma de Puebla, Puebla, México.,Instituto de Ciencias, Licenciatura en Biotecnología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Elena Bello-López
- Instituto de Ciencias, Centro de Investigaciones de Ciencias Microbiológicas, Posgrado en Microbiología, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | | | - Ygnacio Martínez-Laguna
- Instituto de Ciencias, Centro de Investigaciones de Ciencias Microbiológicas, Posgrado en Microbiología, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Patricia Lozano-Zarain
- Instituto de Ciencias, Centro de Investigaciones de Ciencias Microbiológicas, Posgrado en Microbiología, Benemérita Universidad Autónoma de Puebla, Puebla, México
| |
Collapse
|
166
|
Diversity and Distribution of Resistance Markers in Pseudomonas aeruginosa International High-Risk Clones. Microorganisms 2021; 9:microorganisms9020359. [PMID: 33673029 PMCID: PMC7918723 DOI: 10.3390/microorganisms9020359] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Pseudomonas aeruginosa high-risk clones are disseminated worldwide and they are common causative agents of hospital-acquired infections. In this review, we will summarize available data of high-risk P. aeruginosa clones from confirmed outbreaks and based on whole-genome sequence data. Common feature of high-risk clones is the production of beta-lactamases and among metallo-beta-lactamases NDM, VIM and IMP types are widely disseminated in different sequence types (STs), by contrast FIM type has been reported in ST235 in Italy, whereas GIM type in ST111 in Germany. In the case of ST277, it is most frequently detected in Brazil and it carries a resistome linked to blaSPM. Colistin resistance develops among P. aeruginosa clones in a lesser extent compared to other resistance mechanisms, as ST235 strains remain mainly susceptible to colistin however, some reports described mcr positive P. aeurigonsa ST235. Transferable quinolone resistance determinants are detected in P. aeruginosa high-risk clones and aac(6′)-Ib-cr variant is the most frequently reported as this determinant is incorporated in integrons. Additionally, qnrVC1 was recently detected in ST773 in Hungary and in ST175 in Spain. Continuous monitoring and surveillance programs are mandatory to track high-risk clones and to analyze emergence of novel clones as well as novel resistance determinants.
Collapse
|
167
|
Zhang P, Zou B, Liou YC, Huang C. The pathogenesis and diagnosis of sepsis post burn injury. BURNS & TRAUMA 2021; 9:tkaa047. [PMID: 33654698 PMCID: PMC7901709 DOI: 10.1093/burnst/tkaa047] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/20/2020] [Indexed: 02/05/2023]
Abstract
Burn is an under-appreciated trauma that is associated with unacceptably high morbidity and mortality. Although the survival rate after devastating burn injuries has continued to increase in previous decades due to medical advances in burn wound care, nutritional and fluid resuscitation and improved infection control practices, there are still large numbers of patients at a high risk of death. One of the most common complications of burn is sepsis, which is defined as “severe organ dysfunction attributed to host's disordered response to infection” and is the primary cause of death in burn patients. Indeed, burn injuries are accompanied by a series of events that lead to sepsis and multiple organ dysfunction syndrome, such as a hypovolaemic state, immune and inflammatory responses and metabolic changes. Therefore, clear diagnostic criteria and predictive biomarkers are especially important in the prevention and treatment of sepsis and septic shock. In this review, we focus on the pathogenesis of burn wound infection and the post-burn events leading to sepsis. Moreover, the clinical and promising biomarkers of burn sepsis will also be summarized.
Collapse
Affiliation(s)
- Pengju Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17 People's South Road, Chengdu, 610041, China
| | - Bingwen Zou
- Department of Thoracic Oncology and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu, 610041, China
| | - Yih-Cherng Liou
- Department of Biological Sciences, Faculty of Science, National University of Singapore, 14 Science Drive 4, 117543, Singapore
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17 People's South Road, Chengdu, 610041, China
| |
Collapse
|
168
|
Interplay between ESKAPE Pathogens and Immunity in Skin Infections: An Overview of the Major Determinants of Virulence and Antibiotic Resistance. Pathogens 2021; 10:pathogens10020148. [PMID: 33540588 PMCID: PMC7912840 DOI: 10.3390/pathogens10020148] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/16/2022] Open
Abstract
The skin is the largest organ in the human body, acting as a physical and immunological barrier against pathogenic microorganisms. The cutaneous lesions constitute a gateway for microbial contamination that can lead to chronic wounds and other invasive infections. Chronic wounds are considered as serious public health problems due the related social, psychological and economic consequences. The group of bacteria known as ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter sp.) are among the most prevalent bacteria in cutaneous infections. These pathogens have a high level of incidence in hospital environments and several strains present phenotypes of multidrug resistance. In this review, we discuss some important aspects of skin immunology and the involvement of ESKAPE in wound infections. First, we introduce some fundamental aspects of skin physiology and immunology related to cutaneous infections. Following this, the major virulence factors involved in colonization and tissue damage are highlighted, as well as the most frequently detected antimicrobial resistance genes. ESKAPE pathogens express several virulence determinants that overcome the skin's physical and immunological barriers, enabling them to cause severe wound infections. The high ability these bacteria to acquire resistance is alarming, particularly in the hospital settings where immunocompromised individuals are exposed to these pathogens. Knowledge about the virulence and resistance markers of these species is important in order to develop new strategies to detect and treat their associated infections.
Collapse
|
169
|
Moussouni M, Berry L, Sipka T, Nguyen-Chi M, Blanc-Potard AB. Pseudomonas aeruginosa OprF plays a role in resistance to macrophage clearance during acute infection. Sci Rep 2021; 11:359. [PMID: 33432030 PMCID: PMC7801371 DOI: 10.1038/s41598-020-79678-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022] Open
Abstract
While considered an extracellular pathogen, Pseudomonas aeruginosa has been reported to be engulfed by macrophages in cellular and animal models. However, the role of macrophages in P. aeruginosa clearance in vivo remains poorly studied. The major outer membrane porin OprF has been recently shown to be involved in P. aeruginosa fate within cultured macrophages and analysis of an oprF mutant may thus provide insights to better understand the relevance of this intramacrophage stage during infection. In the present study, we investigated for the first time the virulence of a P. aeruginosa oprF mutant in a vertebrate model that harbors functional macrophages, the zebrafish (Danio rerio) embryo, which offers powerful tools to address macrophage–pathogen interactions. We established that P. aeruginosa oprF mutant is attenuated in zebrafish embryos in a macrophage-dependent manner. Visualization and quantification of P. aeruginosa bacteria phagocytosed by macrophages after injection into closed cavities suggested that the attenuated phenotype of oprF mutant is not linked to higher macrophage recruitment nor better phagocytosis than wild-type strain. Using cultured macrophages, we showed an intramacrophage survival defect of P. aeruginosa oprF mutant, which is correlated with elevated association of bacteria with acidic compartments. Notably, treatment of embryos with bafilomycin, an inhibitor of acidification, increased the sensibility of embryos towards both wild-type and oprF mutant, and partially suppressed the attenuation of oprF mutant. Taken together, this work supports zebrafish embryo as state-of-the-art model to address in vivo the relevance of P. aeruginosa intramacrophage stage. Our results highlight the contribution of macrophages in the clearance of P. aeruginosa during acute infection and suggest that OprF protects P. aeruginosa against macrophage clearance by avoiding bacterial elimination in acidified phagosomes.
Collapse
Affiliation(s)
- Malika Moussouni
- Laboratory of Pathogen-Host Interactions (LPHI), CNRS-UMR5235, Université de Montpellier, Montpellier, France
| | - Laurence Berry
- Laboratory of Pathogen-Host Interactions (LPHI), CNRS-UMR5235, Université de Montpellier, Montpellier, France
| | - Tamara Sipka
- Laboratory of Pathogen-Host Interactions (LPHI), CNRS-UMR5235, Université de Montpellier, Montpellier, France
| | - Mai Nguyen-Chi
- Laboratory of Pathogen-Host Interactions (LPHI), CNRS-UMR5235, Université de Montpellier, Montpellier, France
| | - Anne-Béatrice Blanc-Potard
- Laboratory of Pathogen-Host Interactions (LPHI), CNRS-UMR5235, Université de Montpellier, Montpellier, France.
| |
Collapse
|
170
|
New Textile for Personal Protective Equipment—Plasma Chitosan/Silver Nanoparticles Nylon Fabric. FIBERS 2021. [DOI: 10.3390/fib9010003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fabric structures are prone to contamination with microorganisms, as their morphology and ability to retain moisture creates a proper environment for their growth. In this work, a novel, easily processed and cheap coating for a nylon fabric with antimicrobial characteristics was developed. After plasma treatment, made to render the fabric surface more reactive sites, the fabric was impregnated with chitosan and silver nanoparticles by simply dipping it into a mixture of different concentrations of both components. Silver nanoparticles were previously synthesized using the Lee–Meisel method, and their successful obtention was proven by UV–Vis, showing the presence of the surface plasmon resonance band at 410 nm. Nanoparticles with 25 nm average diameter observed by STEM were stable, mainly in the presence of chitosan, which acted as a surfactant for silver nanoparticles, avoiding their aggregation. The impregnated fabric possessed bactericidal activity higher for Gram-positive Staphylococcus aureus than for Gram-negative Pseudomonas aeruginosa bacteria for all combinations. The percentage of live S. aureus and P. aeruginosa CFU was reduced to less than 20% and 60%, respectively, when exposed to each of the coating combinations. The effect was more pronounced when both chitosan and silver were present in the coating, suggesting an effective synergy between these components. After a washing process, the antimicrobial effect was highly reduced, suggesting that the coating is unstable after washing, being almost completely removed from the fabric. Nevertheless, the new-coated fabric can be successfully used in single-use face masks. To our knowledge, the coating of nylon fabrics intended for face-mask material with both agents has never been reported.
Collapse
|
171
|
Behzadi P, Baráth Z, Gajdács M. It's Not Easy Being Green: A Narrative Review on the Microbiology, Virulence and Therapeutic Prospects of Multidrug-Resistant Pseudomonas aeruginosa. Antibiotics (Basel) 2021; 10:42. [PMID: 33406652 PMCID: PMC7823828 DOI: 10.3390/antibiotics10010042] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/29/2020] [Accepted: 12/31/2020] [Indexed: 12/11/2022] Open
Abstract
Pseudomonas aeruginosa is the most frequent cause of infection among non-fermenting Gram-negative bacteria, predominantly affecting immunocompromised patients, but its pathogenic role should not be disregarded in immunocompetent patients. These pathogens present a concerning therapeutic challenge to clinicians, both in community and in hospital settings, due to their increasing prevalence of resistance, and this may lead to prolonged therapy, sequelae, and excess mortality in the affected patient population. The resistance mechanisms of P. aeruginosa may be classified into intrinsic and acquired resistance mechanisms. These mechanisms lead to occurrence of resistant strains against important antibiotics-relevant in the treatment of P. aeruginosa infections-such as β-lactams, quinolones, aminoglycosides, and colistin. The occurrence of a specific resistotype of P. aeruginosa, namely the emergence of carbapenem-resistant but cephalosporin-susceptible (Car-R/Ceph-S) strains, has received substantial attention from clinical microbiologists and infection control specialists; nevertheless, the available literature on this topic is still scarce. The aim of this present review paper is to provide a concise summary on the adaptability, virulence, and antibiotic resistance of P. aeruginosa to a readership of basic scientists and clinicians.
Collapse
Affiliation(s)
- Payam Behzadi
- Department of Microbiology, College of Basic Sciences, Shahr-e-Qods Branch, Islamic Azad University, Tehran 37541-374, Iran;
| | - Zoltán Baráth
- Department of Prosthodontics, Faculty of Dentistry, University of Szeged, Tisza Lajos körút 62-64, 6720 Szeged, Hungary;
| | - Márió Gajdács
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, 1089 Budapest, Hungary
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, 6720 Szeged, Hungary
| |
Collapse
|
172
|
Zhelev G. Bacterial resistance to antiseptics and disinfectants – minireview. BULGARIAN JOURNAL OF VETERINARY MEDICINE 2021. [DOI: 10.15547/bjvm.2019-0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The appearance of bacterial resistance to disinfectants and antiseptics is an issue of substantial health concern, resulting in low efficiency of epidemic control activities and emergence of microorganisms with cross-resistance to antibiotics and biocides. A synopsis of the main mechanisms of development of resistance to biocides is presented. The emphasis is placed to health risks and impact on medical practice. The main methods for detection of resistance, and prevention measures of key importance for its control are outlined.
Collapse
Affiliation(s)
- G. Zhelev
- Department of Veterinary Microbiology, Infectious and Parasitic Diseases, Faculty of Veterinary Medicine , Trakia University, 6000 Stara Zagora, Bulgaria
| |
Collapse
|
173
|
Dogan Guzel F, Pletzer D, Norouz Dizaji A, Al-Nahas K, Bajrai M, Winterhalter M. Towards understanding single-channel characteristics of OccK8 purified from Pseudomonas aeruginosa. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2021; 50:87-98. [PMID: 33481046 DOI: 10.1007/s00249-021-01498-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/10/2021] [Indexed: 10/22/2022]
Abstract
Antibiotic resistance in Gram-negative bacteria causes serious health issues worldwide. Bacteria employ several resistance mechanisms to cope with antimicrobials. One of their strategies is to reduce the permeability of antibiotics either through general diffusion porins or substrate-specific channels. In this study, one of the substrate-specific channels from Pseudomonas aeruginosa, OccK8 (also known as OprE), was investigated using single-channel electrophysiology. The study also includes the investigation of permeability properties of several amino acids with different charged groups (i.e. arginine, glycine and glutamic acid) through OccK8. We observed four different conformations of the same OccK8 channel when inserted in lipid bilayers. This is in contrast to previous studies where heterologous expressed OccK8 in E. coli showed only one conformation. We hypothesized that the difference in our study was due to the expression and purification of the native channel from P. aeruginosa. The single-channel uptake characteristics of the porin showed that negatively charged glutamic acid preferentially interacted with the channel while the positively charged arginine molecule showed infrequent interaction with OccK8. The neutral amino acid glycine did not show any interaction at the physiological conditions.
Collapse
Affiliation(s)
- Fatma Dogan Guzel
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Ankara Yildirim Beyazit University, 06010, Ankara, Turkey.
- School of Engineering and Science, Jacobs University Bremen, Campus Ring 1, 28759, Bremen, Germany.
| | - Daniel Pletzer
- School of Engineering and Science, Jacobs University Bremen, Campus Ring 1, 28759, Bremen, Germany
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Araz Norouz Dizaji
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Ankara Yildirim Beyazit University, 06010, Ankara, Turkey
| | - Kareem Al-Nahas
- School of Engineering and Science, Jacobs University Bremen, Campus Ring 1, 28759, Bremen, Germany
- Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, UK
| | - Mawadah Bajrai
- School of Engineering and Science, Jacobs University Bremen, Campus Ring 1, 28759, Bremen, Germany
| | - Mathias Winterhalter
- School of Engineering and Science, Jacobs University Bremen, Campus Ring 1, 28759, Bremen, Germany
| |
Collapse
|
174
|
Uchil A, Murali TS, Nayak R. Escaping ESKAPE: A chalcone perspective. RESULTS IN CHEMISTRY 2021. [DOI: 10.1016/j.rechem.2021.100229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
175
|
Perraud Q, Kuhn L, Fritsch S, Graulier G, Gasser V, Normant V, Hammann P, Schalk IJ. Opportunistic use of catecholamine neurotransmitters as siderophores to access iron by Pseudomonas aeruginosa. Environ Microbiol 2020; 24:878-893. [PMID: 33350053 DOI: 10.1111/1462-2920.15372] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 10/22/2022]
Abstract
Iron is an essential nutrient for bacterial growth and the cause of a fierce battle between the pathogen and host during infection. Bacteria have developed several strategies to access iron from the host, the most common being the production of siderophores, small iron-chelating molecules secreted into the bacterial environment. The opportunist pathogen Pseudomonas aeruginosa produces two siderophores, pyoverdine and pyochelin, and is also able to use a wide panoply of xenosiderophores, siderophores produced by other microorganisms. Here, we demonstrate that catecholamine neurotransmitters (dopamine, l-DOPA, epinephrine and norepinephrine) are able to chelate iron and efficiently bring iron into P. aeruginosa cells via TonB-dependent transporters (TBDTs). Bacterial growth assays under strong iron-restricted conditions and with numerous mutants showed that the TBDTs involved are PiuA and PirA. PiuA exhibited more pronounced specificity for dopamine uptake than for norepinephrine, epinephrine and l-DOPA, whereas PirA specificity appeared to be higher for l-DOPA and norepinephrine. Proteomic and qRT-PCR approaches showed pirA transcription and expression to be induced in the presence of all four catecholamines. Finally, the oxidative properties of catecholamines enable them to reduce iron, and we observed ferrous iron uptake via the FeoABC system in the presence of l-DOPA.
Collapse
Affiliation(s)
- Quentin Perraud
- Université de Strasbourg, InnoVec, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France.,CNRS, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France
| | - Lauriane Kuhn
- Plateforme Proteomique Strasbourg - Esplanade, Institut de Biologie Moléculaire et Cellulaire, CNRS, FR1589, 15 rue Descartes, Strasbourg Cedex, F-67084, France
| | - Sarah Fritsch
- Université de Strasbourg, InnoVec, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France.,CNRS, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France
| | - Gwenaëlle Graulier
- Université de Strasbourg, InnoVec, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France.,CNRS, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France
| | - Véronique Gasser
- Université de Strasbourg, InnoVec, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France.,CNRS, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France
| | - Vincent Normant
- Université de Strasbourg, InnoVec, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France.,CNRS, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France
| | - Philippe Hammann
- Plateforme Proteomique Strasbourg - Esplanade, Institut de Biologie Moléculaire et Cellulaire, CNRS, FR1589, 15 rue Descartes, Strasbourg Cedex, F-67084, France
| | - Isabelle J Schalk
- Université de Strasbourg, InnoVec, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France.,CNRS, UMR7242, ESBS, Bld Sébastien Brant, F-67413 Illkirch, Strasbourg, France
| |
Collapse
|
176
|
Sainz-Mejías M, Jurado-Martín I, McClean S. Understanding Pseudomonas aeruginosa-Host Interactions: The Ongoing Quest for an Efficacious Vaccine. Cells 2020; 9:cells9122617. [PMID: 33291484 PMCID: PMC7762141 DOI: 10.3390/cells9122617] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/18/2022] Open
Abstract
Pseudomonas aeruginosa is a leading cause of chronic respiratory infections in people with cystic fibrosis (CF), bronchiectasis or chronic obstructive pulmonary disease (COPD), and acute infections in immunocompromised individuals. The adaptability of this opportunistic pathogen has hampered the development of antimicrobial therapies, and consequently, it remains a major threat to public health. Due to its antimicrobial resistance, vaccines represent an alternative strategy to tackle the pathogen, yet despite over 50 years of research on anti-Pseudomonas vaccines, no vaccine has been licensed. Nevertheless, there have been many advances in this field, including a better understanding of the host immune response and the biology of P. aeruginosa. Multiple antigens and adjuvants have been investigated with varying results. Although the most effective protective response remains to be established, it is clear that a polarised Th2 response is sub-optimal, and a mixed Th1/Th2 or Th1/Th17 response appears beneficial. This comprehensive review collates the current understanding of the complexities of P. aeruginosa-host interactions and its implication in vaccine design, with a view to understanding the current state of Pseudomonal vaccine development and the direction of future efforts. It highlights the importance of the incorporation of appropriate adjuvants to the protective antigen to yield optimal protection.
Collapse
|
177
|
Blomquist KC, Nix DE. A Critical Evaluation of Newer β-Lactam Antibiotics for Treatment of Pseudomonas aeruginosa Infections. Ann Pharmacother 2020; 55:1010-1024. [PMID: 33228374 DOI: 10.1177/1060028020974003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE This article critically evaluates common Pseudomonas aeruginosa resistance mechanisms and the properties newer β-lactam antimicrobials possess to evade these mechanisms. DATA SOURCES An extensive PubMed, Google Scholar, and ClinicalTrials.gov search was conducted (January 1995 to July 2020) to identify relevant literature on epidemiology, resistance mechanisms, antipseudomonal agents, newer β-lactam agents, and clinical data available pertaining to P aeruginosa. STUDY SELECTION AND DATA EXTRACTION Relevant published articles and package inserts were reviewed for inclusion. DATA SYNTHESIS Therapeutic options to treat P aeruginosa infections are limited because of its intrinsic and acquired resistance mechanisms. The goal was to identify advances with newer β-lactams and characterize improvements in therapeutic potential for P aeruginosa infections. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE Multidrug-resistant (MDR) P aeruginosa isolates are increasingly encountered from a variety of infections. This review highlights potential activity gains of newer β-lactam antibacterial drugs and the current clinical data to support their use. Pharmacists will be asked to recommend or evaluate the use of these agents and need to be aware of information specific to P aeruginosa, which differs from experience derived from Enterobacterales infections. CONCLUSIONS Newer agents, including ceftazidime-avibactam, ceftolozane-tazobactam, imipenem-relebactam, and cefiderocol, are useful for the treatment of MDR P aeruginosa infections. These agents offer improved efficacy and less toxicity compared with aminoglycosides and polymyxins and can be used for pathogens that are resistant to first-line antipseudomonal β-lactams. Selection of one agent over another should consider availability, turnaround of susceptibility testing, and product cost. Efficacy data specific for pseudomonal infections are limited, and there are no direct comparisons between the newer agents.
Collapse
Affiliation(s)
- Kathleen C Blomquist
- Department of Pharmacy Practice & Science, University of Arizona, Tucson, Arizona, USA
| | - David E Nix
- Department of Pharmacy Practice & Science, University of Arizona, Tucson, Arizona, USA
- Department of Medicine, Division of Infectious Diseases, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
178
|
Kubiczek D, Raber H, Bodenberger N, Oswald T, Sahan M, Mayer D, Wiese S, Stenger S, Weil T, Rosenau F. The Diversity of a Polyclonal FluCell-SELEX Library Outperforms Individual Aptamers as Emerging Diagnostic Tools for the Identification of Carbapenem Resistant Pseudomonas aeruginosa. Chemistry 2020; 26:14536-14545. [PMID: 32515842 PMCID: PMC7756519 DOI: 10.1002/chem.202000213] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Indexed: 11/06/2022]
Abstract
Textbook procedures require the use of individual aptamers enriched in SELEX libraries which are subsequently chemically synthesized after their biochemical characterization. Here we show that this reduction of the available sequence space of large libraries and thus the diversity of binding molecules reduces the labelling efficiency and fidelity of selected single aptamers towards different strains of the human pathogen Pseudomonas aeruginosa compared to a polyclonal aptamer library enriched by a whole-cell-SELEX involving fluorescent aptamers. The library outperformed single aptamers in reliable and specific targeting of different clinically relevant strains, allowed to inhibit virulence associated cellular functions and identification of bound cell surface targets by aptamer based affinity purification and mass spectrometry. The stunning ease of this FluCell-SELEX and the convincing performance of the P. aeruginosa specific library may pave the way towards generally new and efficient diagnostic techniques based on polyclonal aptamer libraries not only in clinical microbiology.
Collapse
Affiliation(s)
- Dennis Kubiczek
- Institute of pharmaceutical biotechnologyUlm University89081UlmGermany
| | - Heinz Raber
- Institute of pharmaceutical biotechnologyUlm University89081UlmGermany
| | | | - Thomas Oswald
- Institute of pharmaceutical biotechnologyUlm University89081UlmGermany
| | - Melis Sahan
- Institute of pharmaceutical biotechnologyUlm University89081UlmGermany
| | - Daniel Mayer
- Institute for Medical Microbiology and HygieneUniversity Hospital Ulm89081UlmGermany
| | | | - Steffen Stenger
- Institute for Medical Microbiology and HygieneUniversity Hospital Ulm89081UlmGermany
| | - Tanja Weil
- Department Synthesis of MacromoleculesMax-Planck-Institute for Polymer Research Mainz55128MainzGermany
| | - Frank Rosenau
- Institute of pharmaceutical biotechnologyUlm University89081UlmGermany
- Department Synthesis of MacromoleculesMax-Planck-Institute for Polymer Research Mainz55128MainzGermany
| |
Collapse
|
179
|
Regmi RS, Khadka S, Sapkota S, Adhikari S, Dhakal KK, Dhakal B, Lamsal B, Kafle SC. Bacterial etiology of sputum from tuberculosis suspected patients and antibiogram of the isolates. BMC Res Notes 2020; 13:520. [PMID: 33172496 PMCID: PMC7654065 DOI: 10.1186/s13104-020-05369-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/31/2020] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE The current study aims to explore the bacteriology of sputum of tuberculosis (TB) suspected patients. A cross-sectional study was carried out in the sputum samples of 150 TB suspected patients visiting District Public Health Office, Bharatpur, Nepal. The samples were subjected to cultural, microscopic and biochemical analyses for the identification of the isolates. In addition, antibiotic susceptibility tests were carried out with a special focus on ESBL and MBL production following Clinical and Laboratory Standard Institute guidelines. RESULTS Bacterial growth was recovered in 47% (71/150) of the TB suspected patients of which 12.66% (19/150) had pulmonary TB infection. Streptococcus spp. (9%) and Pseudomonas aeruginosa (9%) were the most frequently isolated bacteria. Enterobacteriaceae accounted for 35% of the total isolates. Occurrence of bacterial pathogens was more in males (69%) than in females (31%).The incidence of bacterial pathogen was seen associated with gender of the patients and with the TB infection (p < 0.05) but independent with age of the patients and HIV infection (p > 0.05). Tetracycline was effective against Streptococcus spp. whereas gentamicin was effective against Bacillus species. Imipenem and co-trimoxazole were effective drugs for Gram-negative isolates. Among 83 isolates, 35 were multi-drug resistant, 9 were ESBL producers and 4 were MBL producers.
Collapse
Affiliation(s)
- Ramesh Sharma Regmi
- Department of Microbiology, Birendra Multiple Campus, Tribhuvan University, Bharatpur, Chitwan, 44200, Nepal
| | - Sujan Khadka
- Department of Microbiology, Birendra Multiple Campus, Tribhuvan University, Bharatpur, Chitwan, 44200, Nepal
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Sanjeep Sapkota
- Department of Microbiology, Birendra Multiple Campus, Tribhuvan University, Bharatpur, Chitwan, 44200, Nepal
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Sanjib Adhikari
- Department of Microbiology, Birendra Multiple Campus, Tribhuvan University, Bharatpur, Chitwan, 44200, Nepal.
| | - Khuma Kumari Dhakal
- Department of Microbiology, Birendra Multiple Campus, Tribhuvan University, Bharatpur, Chitwan, 44200, Nepal
| | - Bishnu Dhakal
- Department of Microbiology, Birendra Multiple Campus, Tribhuvan University, Bharatpur, Chitwan, 44200, Nepal
| | - Bandana Lamsal
- Department of Microbiology, Birendra Multiple Campus, Tribhuvan University, Bharatpur, Chitwan, 44200, Nepal
| | - Sarad Chandra Kafle
- Department of Microbiology, Birendra Multiple Campus, Tribhuvan University, Bharatpur, Chitwan, 44200, Nepal
| |
Collapse
|
180
|
The Landscape of Pseudomonas aeruginosa Membrane-Associated Proteins. Cells 2020; 9:cells9112421. [PMID: 33167383 PMCID: PMC7694347 DOI: 10.3390/cells9112421] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Pseudomonas aeruginosa cell envelope-associated proteins play a relevant role in infection mechanisms. They can contribute to the antibiotic resistance of the bacterial cells and be involved in the interaction with host cells. Thus, studies contributing to elucidating these key molecular elements are of great importance to find alternative therapeutics. Methods: Proteins and peptides were extracted by different methods and analyzed by Multidimensional Protein Identification Technology (MudPIT) approach. Proteomic data were processed by Discoverer2.1 software and multivariate statistics, i.e., Linear Discriminant Analysis (LDA), while the Immune Epitope Database (IEDB) resources were used to predict antigenicity and immunogenicity of experimental identified peptides and proteins. Results: The combination of 29 MudPIT runs allowed the identification of 10,611 peptides and 2539 distinct proteins. Following application of extraction methods enriching specific protein domains, about 15% of total identified peptides were classified in trans inner-membrane, inner-membrane exposed, trans outer-membrane and outer-membrane exposed. In this scenario, nine outer membrane proteins (OprE, OprI, OprF, OprD, PagL, OprG, PA1053, PAL and PA0833) were predicted to be highly antigenic. Thus, they were further processed and epitopes target of T cells (MHC Class I and Class II) and B cells were predicted. Conclusion: The present study represents one of the widest characterizations of the P. aeruginosa membrane-associated proteome. The feasibility of our method may facilitates the investigation of other bacterial species whose envelope exposed protein domains are still unknown. Besides, the stepwise prioritization of proteome, by combining experimental proteomic data and reverse vaccinology, may be useful for reducing the number of proteins to be tested in vaccine development.
Collapse
|
181
|
Bouffartigues E, Si Hadj Mohand I, Maillot O, Tortuel D, Omnes J, David A, Tahrioui A, Duchesne R, Azuama CO, Nusser M, Brenner-Weiss G, Bazire A, Connil N, Orange N, Feuilloley MGJ, Lesouhaitier O, Dufour A, Cornelis P, Chevalier S. The Temperature-Regulation of Pseudomonas aeruginosa cmaX-cfrX-cmpX Operon Reveals an Intriguing Molecular Network Involving the Sigma Factors AlgU and SigX. Front Microbiol 2020; 11:579495. [PMID: 33193206 PMCID: PMC7641640 DOI: 10.3389/fmicb.2020.579495] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/23/2020] [Indexed: 11/13/2022] Open
Abstract
Pseudomonas aeruginosa is a highly adaptable Gram-negative opportunistic pathogen, notably due to its large number of transcription regulators. The extracytoplasmic sigma factor (ECFσ) AlgU, responsible for alginate biosynthesis, is also involved in responses to cell wall stress and heat shock via the RpoH alternative σ factor. The SigX ECFσ emerged as a major regulator involved in the envelope stress response via membrane remodeling, virulence and biofilm formation. However, their functional interactions to coordinate the envelope homeostasis in response to environmental variations remain to be determined. The regulation of the putative cmaX-cfrX-cmpX operon located directly upstream sigX was investigated by applying sudden temperature shifts from 37°C. We identified a SigX- and an AlgU- dependent promoter region upstream of cfrX and cmaX, respectively. We show that cmaX expression is increased upon heat shock through an AlgU-dependent but RpoH independent mechanism. In addition, the ECFσ SigX is activated in response to valinomycin, an agent altering the membrane structure, and up-regulates cfrX-cmpX transcription in response to cold shock. Altogether, these data provide new insights into the regulation exerted by SigX and networks that are involved in maintaining envelope homeostasis.
Collapse
Affiliation(s)
- Emeline Bouffartigues
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Ishac Si Hadj Mohand
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Olivier Maillot
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Damien Tortuel
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Jordane Omnes
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Audrey David
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Ali Tahrioui
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Rachel Duchesne
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Cecil Onyedikachi Azuama
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Michael Nusser
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Gerald Brenner-Weiss
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Alexis Bazire
- Laboratoire de Biotechnologie et Chimie Marines (LBCM) EA3884, IUEM, Université de Bretagne-Sud, Lorient, France
| | - Nathalie Connil
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Nicole Orange
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Marc G J Feuilloley
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Olivier Lesouhaitier
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Alain Dufour
- Laboratoire de Biotechnologie et Chimie Marines (LBCM) EA3884, IUEM, Université de Bretagne-Sud, Lorient, France
| | - Pierre Cornelis
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| | - Sylvie Chevalier
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Normandie Université, Université de Rouen Normandie, Centre de Sécurité Sanitaire de Normandie, Evreux, France
| |
Collapse
|
182
|
Xu R, Sun X, Lin H, Han F, Xiao E, Li B, Qiu L, Song B, Yang Z, Sun W. Microbial adaptation in vertical soil profiles contaminated by an antimony smelting plant. FEMS Microbiol Ecol 2020; 96:5910484. [DOI: 10.1093/femsec/fiaa188] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/16/2020] [Indexed: 12/24/2022] Open
Abstract
ABSTRACT
Antimony mining has resulted in considerable pollution to the soil environment. Although studies on antinomy contamination have been conducted, its effects on vertical soil profiles and depth-resolved microbial communities remain unknown. The current study selected three vertical soil profiles (0–2 m) from the world's largest antimony mining area to characterize the depth-resolved soil microbiota and investigate the effects of mining contamination on microbial adaptation. Results demonstrated that contaminated soil profiles showed distinct depth-resolved effects when compared to uncontaminated soil profiles. As soil depth increased, the concentrations of antimony and arsenic gradually declined in the contaminated soil profiles. Acidobacteria, Chloroflexi, Proteobacteria and Thaumarchaeota were the most variable phyla from surface to deep soil. The co-occurrence networks were loosely connected in surface soil, but obviously recovered and were well-connected in deep soil. The metagenomic results indicated that microbial metabolic potential also changed with soil depth. Genes encoding C metabolism pathways were negatively correlated with antimony and arsenic concentrations. Abundances of arsenic-related genes were enriched by severe contamination, but reduced with soil depth. Overall, soil depth-resolved characteristics are often many meters deep and such effects affected the indigenous microbial communities, as well as their metabolic potential due to different contaminants along vertical depths.
Collapse
Affiliation(s)
- Rui Xu
- Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Guangdong Institute of Eco-environmental Science & Technology, Guangdong Academy of Sciences, Guangzhou 510650, P.R. China
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangzhou 510650, P.R. China
| | - Xiaoxu Sun
- Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Guangdong Institute of Eco-environmental Science & Technology, Guangdong Academy of Sciences, Guangzhou 510650, P.R. China
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangzhou 510650, P.R. China
| | - Hanzhi Lin
- Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Guangdong Institute of Eco-environmental Science & Technology, Guangdong Academy of Sciences, Guangzhou 510650, P.R. China
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangzhou 510650, P.R. China
| | - Feng Han
- Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Guangdong Institute of Eco-environmental Science & Technology, Guangdong Academy of Sciences, Guangzhou 510650, P.R. China
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangzhou 510650, P.R. China
| | - Enzong Xiao
- Innovation Center and Key Laboratory of Waters Safety & Protection in the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, P.R. China
| | - Baoqin Li
- Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Guangdong Institute of Eco-environmental Science & Technology, Guangdong Academy of Sciences, Guangzhou 510650, P.R. China
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangzhou 510650, P.R. China
| | - Lang Qiu
- Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Guangdong Institute of Eco-environmental Science & Technology, Guangdong Academy of Sciences, Guangzhou 510650, P.R. China
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangzhou 510650, P.R. China
| | - Benru Song
- Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Guangdong Institute of Eco-environmental Science & Technology, Guangdong Academy of Sciences, Guangzhou 510650, P.R. China
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangzhou 510650, P.R. China
| | - Zhaohui Yang
- College of Environmental Science and Engineering, Hunan University, Changsha 410082, P.R. China
| | - Weimin Sun
- Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Guangdong Institute of Eco-environmental Science & Technology, Guangdong Academy of Sciences, Guangzhou 510650, P.R. China
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangzhou 510650, P.R. China
| |
Collapse
|
183
|
Nadeem SF, Gohar UF, Tahir SF, Mukhtar H, Pornpukdeewattana S, Nukthamna P, Moula Ali AM, Bavisetty SCB, Massa S. Antimicrobial resistance: more than 70 years of war between humans and bacteria. Crit Rev Microbiol 2020; 46:578-599. [PMID: 32954887 DOI: 10.1080/1040841x.2020.1813687] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Development of antibiotic resistance in bacteria is one of the major issues in the present world and one of the greatest threats faced by mankind. Resistance is spread through both vertical gene transfer (parent to offspring) as well as by horizontal gene transfer like transformation, transduction and conjugation. The main mechanisms of resistance are limiting uptake of a drug, modification of a drug target, inactivation of a drug, and active efflux of a drug. The highest quantities of antibiotic concentrations are usually found in areas with strong anthropogenic pressures, for example medical source (e.g., hospitals) effluents, pharmaceutical industries, wastewater influents, soils treated with manure, animal husbandry and aquaculture (where antibiotics are generally used as in-feed preparations). Hence, the strong selective pressure applied by antimicrobial use has forced microorganisms to evolve for survival. The guts of animals and humans, wastewater treatment plants, hospital and community effluents, animal husbandry and aquaculture runoffs have been designated as "hotspots for AMR genes" because the high density of bacteria, phages, and plasmids in these settings allows significant genetic exchange and recombination. Evidence from the literature suggests that the knowledge of antibiotic resistance in the population is still scarce. Tackling antimicrobial resistance requires a wide range of strategies, for example, more research in antibiotic production, the need of educating patients and the general public, as well as developing alternatives to antibiotics (briefly discussed in the conclusions of this article).
Collapse
Affiliation(s)
- Syeda Fatima Nadeem
- Institute of Industrial Biotechnology, Government College University, Lahore, Pakistan
| | - Umar Farooq Gohar
- Institute of Industrial Biotechnology, Government College University, Lahore, Pakistan
| | - Syed Fahad Tahir
- Institute of Industrial Biotechnology, Government College University, Lahore, Pakistan
| | - Hamid Mukhtar
- Institute of Industrial Biotechnology, Government College University, Lahore, Pakistan
| | | | - Pikunthong Nukthamna
- Faculty of Food Industry, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand.,College of Research Methodology and Cognitive Science, Burapha University, Chonburi, Thailand
| | - Ali Muhammed Moula Ali
- Faculty of Food Industry, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | | | - Salvatore Massa
- Faculty of Food Industry, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand.,Department of Agricultural, Food and Environmental Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
184
|
Impey RE, Hawkins DA, Sutton JM, Soares da Costa TP. Overcoming Intrinsic and Acquired Resistance Mechanisms Associated with the Cell Wall of Gram-Negative Bacteria. Antibiotics (Basel) 2020; 9:E623. [PMID: 32961699 PMCID: PMC7558195 DOI: 10.3390/antibiotics9090623] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/19/2022] Open
Abstract
The global increase in multi-drug-resistant bacteria is severely impacting our ability to effectively treat common infections. For Gram-negative bacteria, their intrinsic and acquired resistance mechanisms are heightened by their unique cell wall structure. The cell wall, while being a target of some antibiotics, represents a barrier due to the inability of most antibacterial compounds to traverse and reach their intended target. This means that its composition and resulting mechanisms of resistance must be considered when developing new therapies. Here, we discuss potential antibiotic targets within the most well-characterised resistance mechanisms associated with the cell wall in Gram-negative bacteria, including the outer membrane structure, porins and efflux pumps. We also provide a timely update on the current progress of inhibitor development in these areas. Such compounds could represent new avenues for drug discovery as well as adjuvant therapy to help us overcome antibiotic resistance.
Collapse
Affiliation(s)
- Rachael E. Impey
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (R.E.I.); (D.A.H.)
| | - Daniel A. Hawkins
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (R.E.I.); (D.A.H.)
| | - J. Mark Sutton
- National Infection Service, Research and Development Institute, Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK;
| | - Tatiana P. Soares da Costa
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (R.E.I.); (D.A.H.)
| |
Collapse
|
185
|
Structure-activity relationship of diameter controlled Ag@Cu nanoparticles in broad-spectrum antibacterial mechanism. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111501. [PMID: 33321601 DOI: 10.1016/j.msec.2020.111501] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 01/27/2023]
Abstract
Current outbreaks associated with drug-resistant clinical strains are demanding for the development of broad-spectrum antibacterial agents. The bactericidal materials should be eco-friendly, economical and effective to suppress bacterial growth. Thus, in this work, diameter controlled spherical Cucore-Agshell nanoparticles (Ag@CuNPs) with diameter ranging from 70 to 100 nm by one-step co-reduction approach were designed and synthesized. The Ag@CuNPs were homogenous, stable, and positively charged. The 70 nm Ag@CuNPs showed a consistent and regular Ag shielding. We observed the 100 nm Ag@CuNPs achieved symmetrical doped Ag clusters on the Cu core surface. We used Gram-positive and Gram-negative models strains to test the wide-spectrum antibacterial activity. The Ag@CuNPs showed detrimental microbial viability in a dose-dependent manner; however, 70 nm Ag@CuNPs were superior to those of 100 nm Ag@CuNPs. Initially, Ag@CuNPs attached and translocated the membrane surface resulting in bacterial eradication. Our analyses exhibited that antibacterial mechanism was not governed by the bacterial genre, nonetheless, by cell type, morphology, growing ability and the NPs uptake capability. The Ag@CuNPs were highly tolerated by human fibroblasts, mainly by the use of starch as glucosidic capper and stabilizer, suggesting optimal biocompatibility and activity. The Ag@CuNPs open up a novel platform to study the potential action of bimetallic nanoparticles and their molecular role for biomedical, clinical, hospital and industrial-chemical applications.
Collapse
|
186
|
Camus L, Briaud P, Bastien S, Elsen S, Doléans-Jordheim A, Vandenesch F, Moreau K. Trophic cooperation promotes bacterial survival of Staphylococcus aureus and Pseudomonas aeruginosa. ISME JOURNAL 2020; 14:3093-3105. [PMID: 32814867 DOI: 10.1038/s41396-020-00741-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 07/30/2020] [Accepted: 08/06/2020] [Indexed: 01/07/2023]
Abstract
In the context of infection, Pseudomonas aeruginosa and Staphylococcus aureus are frequently co-isolated, particularly in cystic fibrosis (CF) patients. Within lungs, the two pathogens exhibit a range of competitive and coexisting interactions. In the present study, we explored the impact of S. aureus on the physiology of P. aeruginosa in the context of coexistence. Transcriptomic analyses showed that S. aureus significantly and specifically affects the expression of numerous genes involved in P. aeruginosa carbon and amino acid metabolism. In particular, 65% of the strains presented considerable overexpression of the genes involved in the acetoin catabolic (aco) pathway. We demonstrated that acetoin is (i) produced by clinical S. aureus strains, (ii) detected in sputa from CF patients and (iii) involved in P. aeruginosa's aco system induction. Furthermore, acetoin is catabolized by P. aeruginosa, a metabolic process that improves the survival of both pathogens by providing a new carbon source for P. aeruginosa and avoiding the toxic accumulation of acetoin on S. aureus. Due to its beneficial effects on both bacteria, acetoin catabolism could testify to the establishment of trophic cooperation between S. aureus and P. aeruginosa in the CF lung environment, thus promoting their persistence.
Collapse
Affiliation(s)
- Laura Camus
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Paul Briaud
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Sylvère Bastien
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Sylvie Elsen
- Université Grenoble Alpes, CNRS ERL5261, CEA-IRIG-BCI, INSERM UMR1036, 38000, Grenoble, France
| | - Anne Doléans-Jordheim
- Institut des agents infectieux, Hospices Civils de Lyon, Lyon, France.,Bactéries Pathogènes Opportunistes et Environnement, UMR CNRS 5557 Ecologie Microbienne, Université Lyon 1 and VetAgro Sup, Villeurbanne, France
| | - François Vandenesch
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France.,Institut des agents infectieux, Hospices Civils de Lyon, Lyon, France.,Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, Lyon, France
| | - Karen Moreau
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France.
| |
Collapse
|
187
|
Carraro N, Sentchilo V, Polák L, Bertelli C, van der Meer JR. Insights into Mobile Genetic Elements of the Biocide-Degrading Bacterium Pseudomonas nitroreducens HBP-1. Genes (Basel) 2020; 11:genes11080930. [PMID: 32806781 PMCID: PMC7466150 DOI: 10.3390/genes11080930] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 02/07/2023] Open
Abstract
The sewage sludge isolate Pseudomonas nitroreducens HBP-1 was the first bacterium known to completely degrade the fungicide 2-hydroxybiphenyl. PacBio and Illumina whole-genome sequencing revealed three circular DNA replicons: a chromosome and two plasmids. Plasmids were shown to code for putative adaptive functions such as heavy metal resistance, but with unclarified ability for self-transfer. About one-tenth of strain HBP-1's chromosomal genes are likely of recent horizontal influx, being part of genomic islands, prophages and integrative and conjugative elements (ICEs). P. nitroreducens carries two large ICEs with different functional specialization, but with homologous core structures to the well-known ICEclc of Pseudomonas knackmussii B13. The variable regions of ICEPni1 (96 kb) code for, among others, heavy metal resistances and formaldehyde detoxification, whereas those of ICEPni2 (171 kb) encodes complete meta-cleavage pathways for catabolism of 2-hydroxybiphenyl and salicylate, a protocatechuate pathway and peripheral enzymes for 4-hydroxybenzoate, ferulate, vanillin and vanillate transformation. Both ICEs transferred at frequencies of 10-6-10-8 per P. nitroreducens HBP-1 donor into Pseudomonas putida, where they integrated site specifically into tRNAGly-gene targets, as expected. Our study highlights the underlying determinants and mechanisms driving dissemination of adaptive properties allowing bacterial strains to cope with polluted environments.
Collapse
Affiliation(s)
- Nicolas Carraro
- Department of Fundamental Microbiology, University of Lausanne, Biophore, Quartier UNIL-Sorge, 1015 Lausanne, Switzerland; (V.S.); (L.P.); (J.R.v.d.M.)
- Correspondence:
| | - Vladimir Sentchilo
- Department of Fundamental Microbiology, University of Lausanne, Biophore, Quartier UNIL-Sorge, 1015 Lausanne, Switzerland; (V.S.); (L.P.); (J.R.v.d.M.)
| | - Lenka Polák
- Department of Fundamental Microbiology, University of Lausanne, Biophore, Quartier UNIL-Sorge, 1015 Lausanne, Switzerland; (V.S.); (L.P.); (J.R.v.d.M.)
| | - Claire Bertelli
- Institute for Microbiology, Lausanne University Hospital and University of Lausanne, Bugnon 48, 1011 Lausanne, Switzerland;
| | - Jan Roelof van der Meer
- Department of Fundamental Microbiology, University of Lausanne, Biophore, Quartier UNIL-Sorge, 1015 Lausanne, Switzerland; (V.S.); (L.P.); (J.R.v.d.M.)
| |
Collapse
|
188
|
Wang S, Huang Z, Wan Q, Feng S, Xie X, Zhang R, Zhang Z. Comparative Genomic and Metabolomic Analyses of Two Pseudomonas aeruginosa Strains With Different Antifungal Activities. Front Microbiol 2020; 11:1841. [PMID: 32849439 PMCID: PMC7412747 DOI: 10.3389/fmicb.2020.01841] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022] Open
Abstract
Pseudomonas aeruginosa isolated from the plant rhizosphere has been widely used as an effective strain in biological control against plant disease. This bacterium promotes plant growth and protect plants against various phytopathogens through the production of phenazine metabolites. In this study, the strain P. aeruginosa Y12 with anti-Beauveria bassiana activity was isolated from the gut of housefly larvae. It was comparatively analyzed with the strain P. aeruginosa P18, which showed no anti-B. bassiana activity. Genomic and metabolomic methods were used to obtain a comprehensive understanding of the antimicrobial mechanism of Y12. After whole-genome resequencing of the two strains, a total of 7,087 non-synonymous single-nucleotide polymorphisms (nsSNPs), 1079 insertions and deletions (InDels), 62 copy-number variations (CNVs) and 42 structural variations (SV) were found in both strains. We analyzed the differentially abundant metabolites between Y12 and P18, and identified six bioactive compounds that could be associated with the antimicrobial activity of Y12. Additionally, we found that, unlike other previously reported rhizospheric P. aeruginosa strains, Y12 could produce both phenazine-1,6-dicarboxylic acid (PDC) and pyocyanin (PYO) at significantly higher concentrations than P18. As B. bassiana is an effective biological insecticide that can cause high mortality in adult houseflies but has little effect on housefly larvae, we believe that P. aeruginosa Y12, identified in housefly larvae but not in adults, were beneficial for the development of housefly larvae and could protect them from B. bassiana infection through the production of toxic metabolites.
Collapse
Affiliation(s)
- Shumin Wang
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Zhendong Huang
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Qing Wan
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Shuo Feng
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Xiaochen Xie
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Ruiling Zhang
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Zhong Zhang
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| |
Collapse
|
189
|
Trifan A, Luca SV, Greige-Gerges H, Miron A, Gille E, Aprotosoaie AC. Recent advances in tackling microbial multidrug resistance with essential oils: combinatorial and nano-based strategies. Crit Rev Microbiol 2020; 46:338-357. [DOI: 10.1080/1040841x.2020.1782339] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Adriana Trifan
- Department of Pharmacognosy, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, Iasi, Romania
| | - Simon Vlad Luca
- Department of Pharmacognosy, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, Iasi, Romania
- Biothermodynamics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Hélène Greige-Gerges
- Bioactive Molecules Research Laboratory, Faculty of Sciences, Lebanese University, Jdaidet El-Matn, Lebanon
| | - Anca Miron
- Department of Pharmacognosy, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, Iasi, Romania
| | - Elvira Gille
- National Institute of Research & Development for Biological Sciences Bucuresti/Stejarul Biological Research Centre, Piatra Neamt, Romania
| | - Ana Clara Aprotosoaie
- Department of Pharmacognosy, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, Iasi, Romania
| |
Collapse
|
190
|
Terra ACG, Salvador EA. In silico integrative analysis predicts relevant properties of exotoxin-derived peptides for the design of vaccines against Pseudomonas aeruginosa. INFECTION GENETICS AND EVOLUTION 2020; 85:104424. [PMID: 32561294 DOI: 10.1016/j.meegid.2020.104424] [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: 04/10/2020] [Revised: 05/25/2020] [Accepted: 06/12/2020] [Indexed: 11/18/2022]
Abstract
Pseudomonas aeruginosa (PA) is an opportunistic human pathogen responsible for causing serious infections in patients with cystic fibrosis. Infections caused by PA are difficult to treat and eradicate due to intrinsic and added resistance to antibiotic therapy. Therefore, it is necessary to establish effective prevention strategies against this infectious agent. In this study, a combination of immunoinformatic tools was applied to predict immunogenic and immunodominant regions in the structure of exotoxins commonly secreted as virulence factors in PA infection (ExoA, ExoS, ExoT, ExoU and ExoY). The peptides derived from exotoxins were evaluated for the potential affinity for human leukocyte antigen (HLA) I and HLA-II molecules, antigenicity score and toxicity profile. From an initial screening of 941 peptides, 13 (1.38%) were successful in all analyzes. The peptides with relevant immunogenic properties were mainly those derived from Exo A (10 / 76.9%). All peptides selected in the last analysis present a high population coverage rate based on the interaction of HLA alleles (95.36 ± 7.83%). Therefore, the peptides characterized in this study are recommended for in vitro and in vivo studies and can provide the basis for the rational design of a vaccine against PA.
Collapse
|
191
|
Inhibition of HIV-1 Protease by Carpobrotus edulis (L.). EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9648056. [PMID: 32595755 PMCID: PMC7298281 DOI: 10.1155/2020/9648056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/07/2020] [Accepted: 05/19/2020] [Indexed: 12/15/2022]
Abstract
Carpobrotus edulis (L.) is a plant commonly found in the Eastern Cape Province of South Africa and is used for the general treatment of infections relating to the human immunodeficiency virus (HIV). HIV-1 protease plays an important role during HIV replication and maturation to its infectious form, and therefore inhibition of the enzyme is one of the main focus areas in drug development. The inhibitory effect of a water extract of C. edulis leaves against HIV-1 protease activity was determined using the SensoLyte® 520 HIV-1 protease assay fluorimetric kit and employing a HiLyte Fluor™488/QXL™520 fluorescence resonance energy transfer (FRET) peptide. Cytotoxicity of the extract towards HeLa Chang cell lines was determined using an in vitro MTT assay, and the phytochemical profile of the extract was determined with FT-IR and LC-MS. HIV-1 protease activity was inhibited 83.06% (IC50 1.6 mg/ml) (p < 0.0001) by the pepstatin A inhibitor control. Treatment with all C. edulis extract concentrations (16, 1.6, 0.16, and 0.016 mg/ml) inhibited HIV-1 protease activity significantly (p < 0.0001) in a typical dose response manner. With regards to cytotoxicity, the negative controls containing untreated HeLa Chang cells exhibited high formazan formation rates in contrast with the positive controls, containing curcumin, which reduced formazan formation significantly (p < 0.001), exhibiting cytotoxicity towards the cells. There was no significant (p > 0.05) difference in the formazan formation rates between the negative controls and 1, 0.5, 0.125, 0.065, 0.031, and 0.015 mg/ml plant extract, confirming no toxicity of C. edulis extracts towards HeLa Chang cells. Major functional phytochemical compounds identified included alcohols, phenols, alkanes, amines, carboxylic acids, and esters. LC-ESI-TOF/MS analysis revealed the putative identities of main compounds present in the aqueous leaves extract, including some that contribute to its anti-HIV-1 protease action.
Collapse
|
192
|
Sonnleitner E, Pusic P, Wolfinger MT, Bläsi U. Distinctive Regulation of Carbapenem Susceptibility in Pseudomonas aeruginosa by Hfq. Front Microbiol 2020; 11:1001. [PMID: 32528439 PMCID: PMC7264166 DOI: 10.3389/fmicb.2020.01001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 04/24/2020] [Indexed: 12/29/2022] Open
Abstract
Carbapenems are often the antibiotics of choice to combat life threatening infections caused by the opportunistic human pathogen Pseudomonas aeruginosa. The outer membrane porins OprD and OpdP serve as entry ports for carbapenems. Here, we report that the RNA chaperone Hfq governs post-transcriptional regulation of the oprD and opdP genes in a distinctive manner. Hfq together with the recently described small regulatory RNAs (sRNAs) ErsA and Sr0161 is shown to mediate translational repression of oprD, whereas opdP appears not to be regulated by sRNAs. At variance, our data indicate that opdP is translationally repressed by a regulatory complex consisting of Hfq and the catabolite repression protein Crc, an assembly known to be key to carbon catabolite repression in P. aeruginosa. The regulatory RNA CrcZ, which is up-regulated during growth of P. aeruginosa on less preferred carbon sources, is known to sequester Hfq, which relieves Hfq-mediated translational repression of genes. The differential carbapenem susceptibility during growth on different carbon sources can thus be understood in light of Hfq-dependent oprD/opdP regulation and of the antagonizing function of the CrcZ RNA on Hfq regulatory complexes.
Collapse
Affiliation(s)
- Elisabeth Sonnleitner
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, Vienna BioCenter (VBC), University of Vienna, Vienna, Austria
| | - Petra Pusic
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, Vienna BioCenter (VBC), University of Vienna, Vienna, Austria
| | - Michael T Wolfinger
- Department of Theoretical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria.,Research Group Bioinformatics and Computational Biology, Faculty of Computer Science, University of Vienna, Vienna, Austria
| | - Udo Bläsi
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, Vienna BioCenter (VBC), University of Vienna, Vienna, Austria
| |
Collapse
|
193
|
Owusu EGA, Yaghini E, Naasani I, Parkin IP, Allan E, MacRobert AJ. Synergistic interactions of cadmium-free quantum dots embedded in a photosensitised polymer surface: efficient killing of multidrug-resistant strains at low ambient light levels. NANOSCALE 2020; 12:10609-10622. [PMID: 32373810 PMCID: PMC7497474 DOI: 10.1039/c9nr10421f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/20/2020] [Indexed: 06/11/2023]
Abstract
Cadmium-free quantum dots (QD) were combined with crystal violet photosensitising dye and incorporated into medical grade polyurethane via a non-covalent dipping process known as 'swell-encapsulation-shrink'. The antibacterial efficacy of the prepared quantum dot-crystal violet polyurethane substrates (QD + CV PU) was investigated under low power visible light illumination at similar intensities (500 lux) to those present in clinical settings. The antibacterial performance of QD + CV PU was superior to the constituent polymer substrates, eliminating ∼99.9% of an environmental P. aeruginosa strain, a clinical P. aeruginosa strain from a cystic fibrosis patient and a clinical E. coli strain. The nature of the reactive oxygen species (ROS) involved in antibacterial activity of the QD + CV PU surface was investigated using ROS inhibitors and time-resolved optical spectroscopy. The photo-physical interactions of the green-emitting QDs with CV lead to a combination of Type I and II electron transfer and energy transfer processes, with the highly potent ROS singlet oxygen playing a dominant role. This study is the first to demonstrate highly efficient synergistic killing of clinical and environmental strains of intrinsically resistant and multi-drug resistant Gram-negative bacteria using light-activated surfaces containing biocompatible cadmium-free QDs and crystal violet dye at ambient light levels.
Collapse
Affiliation(s)
- Ethel G. A. Owusu
- UCL Division of Surgery and Interventional Science
, University College London
,
Charles Bell House
, 43-45 Foley Street
, London W1 W 7TS
, UK
.
- Materials Chemistry Research Centre
, Department of Chemistry
, University College London
,
20 Gordon Street
, London WC1H 0AJ
, UK
- Department of Microbial Diseases
, UCL Eastman Dental Institute
, University College London
,
256 Gray's Inn Road
, London WC1X 8LD
, UK
| | - Elnaz Yaghini
- UCL Division of Surgery and Interventional Science
, University College London
,
Charles Bell House
, 43-45 Foley Street
, London W1 W 7TS
, UK
.
| | - Imad Naasani
- Nanoco Technologies Ltd
,
46 Grafton Street
, Manchester M13 9NT
, UK
| | - Ivan P. Parkin
- Materials Chemistry Research Centre
, Department of Chemistry
, University College London
,
20 Gordon Street
, London WC1H 0AJ
, UK
| | - Elaine Allan
- Department of Microbial Diseases
, UCL Eastman Dental Institute
, University College London
,
256 Gray's Inn Road
, London WC1X 8LD
, UK
| | - Alexander J. MacRobert
- UCL Division of Surgery and Interventional Science
, University College London
,
Charles Bell House
, 43-45 Foley Street
, London W1 W 7TS
, UK
.
| |
Collapse
|
194
|
Jing H, Zhang X, Zou J, Yuan Y, Chen Z, Liu D, Wu W, Yang F, Lu D, Zou Q, Zhang J. Oligomerization of IC43 resulted in improved immunogenicity and protective efficacy against Pseudomonas aeruginosa lung infection. Int J Biol Macromol 2020; 159:174-182. [PMID: 32413471 DOI: 10.1016/j.ijbiomac.2020.05.057] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 12/27/2022]
Abstract
IC43, a truncate form of outer membrane proteins OprF190-342 and OprI21-83 from Pseudomonas aeruginosa, is a promising candidate antigen and exists as monomer in solution. In this study, we generated the heptamer of IC43 by carrier protein aided oligomerization, which was confirmed by gel-filtration and chemical cross-linking analysis. The carrier protein naturally exists as a homo-heptamer, and IC43 was displayed on the surface of the carrier protein in the fusion protein. Immunization with this fusion protein resulted in increased level of antigen specific IgG antibodies and higher survival rate after infection. The improved efficacy was correlated with lower bacteria burden, inflammation and tissue damage in the lungs of immunized mice. Further studies revealed that immunization with this fusion protein resulted in increased levels of IL-4 and antigen specific IgG1, suggesting a stronger Th2 immune response was induced. The improved immunogenicity may be attributed to the exposure of more epitopes on the antigen, which was confirmed by results from immune-dominant peptide mapping and passive immunization. These results demonstrated a possible strategy to improve the immunogenicity of an antigen by carrier protein aided oligomerization.
Collapse
Affiliation(s)
- Haiming Jing
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Xiaoli Zhang
- Department of Clinical Hematology, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Jintao Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Yue Yuan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Zhifu Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Dong Liu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Weiru Wu
- Department of Clinical Hematology, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Feng Yang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Dongshui Lu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China.
| |
Collapse
|
195
|
Parducho KR, Beadell B, Ybarra TK, Bush M, Escalera E, Trejos AT, Chieng A, Mendez M, Anderson C, Park H, Wang Y, Lu W, Porter E. The Antimicrobial Peptide Human Beta-Defensin 2 Inhibits Biofilm Production of Pseudomonas aeruginosa Without Compromising Metabolic Activity. Front Immunol 2020; 11:805. [PMID: 32457749 PMCID: PMC7225314 DOI: 10.3389/fimmu.2020.00805] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 04/08/2020] [Indexed: 12/13/2022] Open
Abstract
Biofilm production is a key virulence factor that facilitates bacterial colonization on host surfaces and is regulated by complex pathways, including quorum sensing, that also control pigment production, among others. To limit colonization, epithelial cells, as part of the first line of defense, utilize a variety of antimicrobial peptides (AMPs) including defensins. Pore formation is the best investigated mechanism for the bactericidal activity of AMPs. Considering the induction of human beta-defensin 2 (HBD2) secretion to the epithelial surface in response to bacteria and the importance of biofilm in microbial infection, we hypothesized that HBD2 has biofilm inhibitory activity. We assessed the viability and biofilm formation of a pyorubin-producing Pseudomonas aeruginosa strain in the presence and absence of HBD2 in comparison to the highly bactericidal HBD3. At nanomolar concentrations, HBD2 - independent of its chiral state - significantly reduced biofilm formation but not metabolic activity, unlike HBD3, which reduced biofilm and metabolic activity to the same degree. A similar discrepancy between biofilm inhibition and maintenance of metabolic activity was also observed in HBD2 treated Acinetobacter baumannii, another Gram-negative bacterium. There was no evidence for HBD2 interference with the regulation of biofilm production. The expression of biofilm-related genes and the extracellular accumulation of pyorubin pigment, another quorum sensing controlled product, did not differ significantly between HBD2 treated and control bacteria, and in silico modeling did not support direct binding of HBD2 to quorum sensing molecules. However, alterations in the outer membrane protein profile accompanied by surface topology changes, documented by atomic force microscopy, was observed after HBD2 treatment. This suggests that HBD2 induces structural changes that interfere with the transport of biofilm precursors into the extracellular space. Taken together, these data support a novel mechanism of biofilm inhibition by nanomolar concentrations of HBD2 that is independent of biofilm regulatory pathways.
Collapse
Affiliation(s)
- Kevin R. Parducho
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, CA, United States
| | - Brent Beadell
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Tiffany K. Ybarra
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Mabel Bush
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Erick Escalera
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Aldo T. Trejos
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Andy Chieng
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, CA, United States
| | - Marlon Mendez
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Chance Anderson
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Hyunsook Park
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Yixian Wang
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, CA, United States
| | - Wuyuan Lu
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Edith Porter
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
196
|
Rémy B, Plener L, Decloquement P, Armstrong N, Elias M, Daudé D, Chabrière É. Lactonase Specificity Is Key to Quorum Quenching in Pseudomonas aeruginosa. Front Microbiol 2020; 11:762. [PMID: 32390993 PMCID: PMC7193897 DOI: 10.3389/fmicb.2020.00762] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/30/2020] [Indexed: 12/31/2022] Open
Abstract
The human opportunistic pathogen Pseudomonas aeruginosa orchestrates the expression of many genes in a cell density-dependent manner by using quorum sensing (QS). Two acyl-homoserine lactones (AHLs) are involved in QS circuits and contribute to the regulation of virulence factors production, biofilm formation, and antimicrobial sensitivity. Disrupting QS, a strategy referred to as quorum quenching (QQ) can be achieved using exogenous AHL-degrading lactonases. However, the importance of enzyme specificity on quenching efficacy has been poorly investigated. Here, we used two lactonases both targeting the signal molecules N-(3-oxododecanoyl)-L-homoserine lactone (3-oxo-C12 HSL) and butyryl-homoserine lactone (C4 HSL) albeit with different efficacies on C4 HSL. Interestingly, both lactonases similarly decreased AHL concentrations and comparably impacted the expression of AHL-based QS genes. However, strong variations were observed in Pseudomonas Quinolone Signal (PQS) regulation depending on the lactonase used. Both lactonases were also found to decrease virulence factors production and biofilm formation in vitro, albeit with different efficiencies. Unexpectedly, only the lactonase with lower efficacy on C4 HSL was able to inhibit P. aeruginosa pathogenicity in vivo in an amoeba infection model. Similarly, proteomic analysis revealed large variations in protein levels involved in antibiotic resistance, biofilm formation, virulence and diverse cellular mechanisms depending on the chosen lactonase. This global analysis provides evidences that QQ enzyme specificity has a significant impact on the modulation of QS-associated behavior in P. aeruginosa PA14.
Collapse
Affiliation(s)
- Benjamin Rémy
- Aix Marseille University, Institut de Recherche pour le Développement, Assistance Publique - Hôpitaux de Marseille, Microbes Evolution Phylogeny and Infections, Institut Hospitalo-Universitaire-Méditerranée Infection, Marseille, France.,Gene&GreenTK, Marseille, France
| | | | - Philippe Decloquement
- Aix Marseille University, Institut de Recherche pour le Développement, Assistance Publique - Hôpitaux de Marseille, Microbes Evolution Phylogeny and Infections, Institut Hospitalo-Universitaire-Méditerranée Infection, Marseille, France
| | - Nicholas Armstrong
- Aix Marseille University, Institut de Recherche pour le Développement, Assistance Publique - Hôpitaux de Marseille, Microbes Evolution Phylogeny and Infections, Institut Hospitalo-Universitaire-Méditerranée Infection, Marseille, France
| | - Mikael Elias
- Department of Biochemistry, Molecular Biology and Biophysics - BioTechnology Institute, University of Minnesota, St. Paul, MN, United States
| | | | - Éric Chabrière
- Aix Marseille University, Institut de Recherche pour le Développement, Assistance Publique - Hôpitaux de Marseille, Microbes Evolution Phylogeny and Infections, Institut Hospitalo-Universitaire-Méditerranée Infection, Marseille, France
| |
Collapse
|
197
|
Recombinant N-terminal outer membrane porin (OprF) of Pseudomonas aeruginosa is a promising vaccine candidate against both P. aeruginosa and some strains of Acinetobacter baumannii. Int J Med Microbiol 2020; 310:151415. [PMID: 32156509 DOI: 10.1016/j.ijmm.2020.151415] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/19/2020] [Accepted: 02/27/2020] [Indexed: 02/06/2023] Open
|
198
|
Svenningsen SW, Frederiksen RF, Counil C, Ficker M, Leisner JJ, Christensen JB. Synthesis and Antimicrobial Properties of a Ciprofloxacin and PAMAM-dendrimer Conjugate. Molecules 2020; 25:molecules25061389. [PMID: 32197523 PMCID: PMC7146445 DOI: 10.3390/molecules25061389] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 02/01/2023] Open
Abstract
Infections caused by bacteria resistant to antibiotics are an increasing problem. Multivalent antibiotics could be a solution. In the present study, a covalent conjugate between Ciprofloxacin and a G0-PAMAM dendrimer has been synthesized and tested against clinically relevant Gram-positive and Gram-negative bacteria. The conjugate has antimicrobial activity and there is a positive dendritic effect compared to Ciprofloxacin itself.
Collapse
Affiliation(s)
- Søren Wedel Svenningsen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark; (S.W.S.); (C.C.); (M.F.)
| | - Rikki Franklin Frederiksen
- Department of Veterinary and Animal Sciences, Food Safety and Zoonoses, University of Copenhagen, Grønnegårdsvej 15, DK-1870 Frederiksberg C, Denmark; (R.F.F.); (J.J.L.)
| | - Claire Counil
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark; (S.W.S.); (C.C.); (M.F.)
| | - Mario Ficker
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark; (S.W.S.); (C.C.); (M.F.)
| | - Jørgen J. Leisner
- Department of Veterinary and Animal Sciences, Food Safety and Zoonoses, University of Copenhagen, Grønnegårdsvej 15, DK-1870 Frederiksberg C, Denmark; (R.F.F.); (J.J.L.)
| | - Jørn Bolstad Christensen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark; (S.W.S.); (C.C.); (M.F.)
- Correspondence: ; Tel.: +45-3533-2452
| |
Collapse
|
199
|
Poblete-Castro I, Aravena-Carrasco C, Orellana-Saez M, Pacheco N, Cabrera A, Borrero-de Acuña JM. Engineering the Osmotic State of Pseudomonas putida KT2440 for Efficient Cell Disruption and Downstream Processing of Poly(3-Hydroxyalkanoates). Front Bioeng Biotechnol 2020; 8:161. [PMID: 32211393 PMCID: PMC7066983 DOI: 10.3389/fbioe.2020.00161] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/17/2020] [Indexed: 11/17/2022] Open
Abstract
In the last decade, the development of novel programmable cell lytic systems based on different inducible genetic constructs like the holin–endolysin and lysozyme appears as a promising alternative to circumvent the use of costly enzymes and mechanical disrupters for downstream processing of intracellular microbial products. Despite the advances, upon activation of these systems the cellular disruption of the biocatalyst occurs in an extended period, thus delaying the recovery of poly(3-hydroxyalkanoate) (PHA). Herein the osmotic state of Pseudomonas putida KT2440 was engineered by inactivating the inner-membrane residing rescue valve MscL, which is responsible mainly for circumventing low-osmolarity challenges. Then the major outer membrane porin OprF and the specific porin OprE were overproduced during PHA producing conditions on decanoate-grown cells. The engineered P. putida strains carrying each porin showed no impairment on growth rate and final biomass and PHA yield after 48 h cultivation. Expression of both porins in tandem in the mutant strain KTΔmscL-oprFE led to a slight reduction of the biomass synthesis (∼10%) but higher PHA accumulation (%wt) relative to the cell dry mass. Each strain was then challenged to an osmotic upshift for 1 h and subsequently to a rapid passage to a hypotonic condition where the membrane stability of the KTΔmscL-oprFE suffered damage, resulting in a rapid reduction of cell viability. Cell disruption accounted for >95% of the cell population within 3 h as reported by colony forming units (CFU), FACS analyses, and transmission electron microscopy. PHA recovery yielded 94.2% of the biosynthesized biopolymer displaying no significant alterations on the final monomer composition. This study can serve as an efficient genetic platform for the recovery of any microbial intracellular compound allowing less unit operation steps for cellular disruption.
Collapse
Affiliation(s)
- Ignacio Poblete-Castro
- Biosystems Engineering Laboratory, Center for Bioinformatics and Integrative Biology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Carla Aravena-Carrasco
- Biosystems Engineering Laboratory, Center for Bioinformatics and Integrative Biology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Matias Orellana-Saez
- Biosystems Engineering Laboratory, Center for Bioinformatics and Integrative Biology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Nicolás Pacheco
- Biosystems Engineering Laboratory, Center for Bioinformatics and Integrative Biology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Alex Cabrera
- Unidad de Citometría de Flujo, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José Manuel Borrero-de Acuña
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany.,Braunschweig Integrated Centre of Systems Biology, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
200
|
A Screen for Antibiotic Resistance Determinants Reveals a Fitness Cost of the Flagellum in Pseudomonas aeruginosa. J Bacteriol 2020; 202:JB.00682-19. [PMID: 31871033 DOI: 10.1128/jb.00682-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/18/2019] [Indexed: 01/09/2023] Open
Abstract
The intrinsic resistance of Pseudomonas aeruginosa to many antibiotics limits treatment options for pseudomonal infections. P. aeruginosa's outer membrane is highly impermeable and decreases antibiotic entry into the cell. We used an unbiased high-throughput approach to examine mechanisms underlying outer membrane-mediated antibiotic exclusion. Insertion sequencing (INSeq) identified genes that altered fitness in the presence of linezolid, rifampin, and vancomycin, antibiotics to which P. aeruginosa is intrinsically resistant. We reasoned that resistance to at least one of these antibiotics would depend on outer membrane barrier function, as previously demonstrated in Escherichia coli and Vibrio cholerae This approach demonstrated a critical role of the outer membrane barrier in vancomycin fitness, while efflux pumps were primary contributors to fitness in the presence of linezolid and rifampin. Disruption of flagellar assembly or function was sufficient to confer a fitness advantage to bacteria exposed to vancomycin. These findings clearly show that loss of flagellar function alone can confer a fitness advantage in the presence of an antibiotic.IMPORTANCE The cell envelopes of Gram-negative bacteria render them intrinsically resistant to many classes of antibiotics. We used insertion sequencing to identify genes whose disruption altered the fitness of a highly antibiotic-resistant pathogen, Pseudomonas aeruginosa, in the presence of antibiotics usually excluded by the cell envelope. This screen identified gene products involved in outer membrane biogenesis and homeostasis, respiration, and efflux as important contributors to fitness. An unanticipated fitness cost of flagellar assembly and function in the presence of the glycopeptide antibiotic vancomycin was further characterized. These findings have clinical relevance for individuals with cystic fibrosis who are infected with P. aeruginosa and undergo treatment with vancomycin for a concurrent Staphylococcus aureus infection.
Collapse
|