1
|
Swain J, Askenasy I, Rudland Nazeer R, Ho PM, Labrini E, Mancini L, Xu Q, Hollendung F, Sheldon I, Dickson C, Welch A, Agbamu A, Godlee C, Welch M. Pathogenicity and virulence of Pseudomonas aeruginosa: Recent advances and under-investigated topics. Virulence 2025; 16:2503430. [PMID: 40353451 PMCID: PMC12087490 DOI: 10.1080/21505594.2025.2503430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/23/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025] Open
Abstract
Pseudomonas aeruginosa is a model for the study of quorum sensing, protein secretion, and biofilm formation. Consequently, it has become one of the most intensely reviewed pathogens, with many excellent articles in the current literature focusing on these aspects of the organism's biology. Here, though, we aim to take a slightly different approach and consider some less well appreciated (but nonetheless important) factors that affect P. aeruginosa virulence. We start by reminding the reader of the global importance of P. aeruginosa infection and that the "virulome" is very niche-specific. Overlooked but obvious questions such as "what prevents secreted protein products from being digested by co-secreted proteases?" are discussed, and we suggest how the nutritional preference(s) of the organism might dictate its environmental reservoirs. Recent studies identifying host genes associated with genetic predisposition towards P. aeruginosa infection (and even infection by specific P. aeruginosa strains) and the role(s) of intracellular P. aeruginosa are introduced. We also discuss the fact that virulence is a high-risk strategy and touch on how expression of the two main classes of virulence factors is regulated. A particular focus is on recent findings highlighting how nutritional status and metabolism are as important as quorum sensing in terms of their impact on virulence, and how co-habiting microbial species at the infection site impact on P. aeruginosa virulence (and vice versa). It is our view that investigation of these issues is likely to dominate many aspects of research into this WHO-designated priority pathogen over the next decade.
Collapse
Affiliation(s)
- Jemima Swain
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Isabel Askenasy
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | - Pok-Man Ho
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Edoardo Labrini
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | - Qingqing Xu
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | | | - Camilla Dickson
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Amelie Welch
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Adam Agbamu
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Camilla Godlee
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Martin Welch
- Department of Biochemistry, Cambridge University, Cambridge, UK
| |
Collapse
|
2
|
Xu C, Feng J, Zhou Y, Ren H, Pan X, Chen S, Liu X, Li G, Li J, Geng B, Gao L, Cheng Z, Jin Y, Ha UH, Jin S, Lamont IL, Pletzer D, Wu W. Azithromycin represses evolution of ceftazidime/avibactam resistance by translational repression of rpoS in Pseudomonas aeruginosa. J Bacteriol 2025; 207:e0055224. [PMID: 40304512 DOI: 10.1128/jb.00552-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 03/07/2025] [Indexed: 05/02/2025] Open
Abstract
Antibiotic combinations can slow down resistance development and/or achieve synergistic therapeutic effects. In this study, we observed that a combined use of ceftazidime-avibactam (CZA) with azithromycin effectively repressed CZA resistance development in Pseudomonas aeruginosa. Transcriptome analysis revealed that subinhibitory concentrations of azithromycin reduced the expression of genes involved in stress-induced mutagenesis, including the stress response sigma factor rpoS. Interestingly, ribosome profiling revealed global redistribution of ribosomes by azithromycin, among which ribosome stalling was significantly intensified near the 5´ terminus of the rpoS mRNA. Further DNA mutational analysis revealed that azithromycin represses the translation of rpoS through its 5´-terminal rare codons, which in turn reduced its transcription. These in vitro observations have been recapitulated in vivo where azithromycin-repressed CZA resistance development when P. aeruginosa was passaged in mice. Overall, our study revealed the molecular mechanism of azithromycin-mediated repression of antibiotic resistance development, providing a promising antibiotic combination for the treatment of P. aeruginosa infections.IMPORTANCEAntibiotic resistance, a global public health challenge, demands the development of novel antibiotics and therapeutic strategies. Ceftazidime-avibactam (CZA) is a combination of a β-lactam antibiotic with a β-lactamase inhibitor that is effective against various gram-negative bacteria such as Pseudomonas aeruginosa. However, clinical CZA-resistant isolates have been reported. Here, we found that combining CZA with azithromycin can effectively suppress the development of resistance in P. aeruginosa in vitro and in vivo. Moreover, we found that azithromycin represses the translation initiation of rpoS through its 5´-terminal rare and less frequent codons, thereby subsequently reducing the mutational frequency of CZA resistance. Therefore, our work provides a promising antibiotic combination for the treatment of P. aeruginosa infections.
Collapse
Affiliation(s)
- Congjuan Xu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Jie Feng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yuchen Zhou
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Huan Ren
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaolei Pan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Shuiping Chen
- Department of Infection and Control, 5th Medical Center of PLA General Hospital, Beijing, China
| | - Xuehua Liu
- Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Guanxian Li
- Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Jinjin Li
- Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Bin Geng
- Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Linlin Gao
- Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China
| | - Zhihui Cheng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yongxin Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Un-Hwan Ha
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, South Korea
| | - Shouguang Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Iain L Lamont
- Department of Biochemistry, University of Otago, Dunedin, Otago, New Zealand
| | - Daniel Pletzer
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Weihui Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
3
|
Leitão MM, Gonçalves ASC, Borges F, Simões M, Borges A. Polypharmacological strategies for infectious bacteria. Pharmacol Rev 2025; 77:100038. [PMID: 40022769 DOI: 10.1016/j.pharmr.2025.100038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 01/03/2025] [Indexed: 03/04/2025] Open
Abstract
Polypharmacological approaches have significant potential for the treatment of various complex diseases, including infectious bacteria-related diseases. Actually, multitargeting agents can achieve better therapeutic effects and overcome the drawbacks of monotherapy. Although multidrug multitarget strategies have demonstrated the ability to inactivate infectious bacteria, several challenges have been pointed out. In this way, multitarget direct ligands approaches appear to be a rational and sustainable strategy to combat antibiotic resistance. By combining different pharmacophores, antibiotic hybrids stand out as a promising application in the field of bacterial infections. These new chemical entities can achieve synergistic interactions that allow to extend the spectrum of action and target multiple pathways. In addition, antibiotic hybrids can reduce the likelihood of resistance development and provide improved chemical stability. It is worth highlighting that despite the efforts of the scientific community to discover new solutions for the most complex diseases, there is a significant lack of studies on biofilm-associated infections. This review describes the different polypharmacological approaches that can be used to treat bacterial infections with a particular focus, whenever possible, on those promoted by biofilms. By exploring these innovative approaches, we aim to inspire further research and progress in the search for effective treatments for infectious bacteria-related diseases, including biofilm-related ones. SIGNIFICANCE STATEMENT: The importance of the proposed topic lies in the escalating challenge of antibiotic resistance, particularly in the context of infectious bacteria-related infections. Polypharmacological approaches, such as antibiotic hybrids, represent innovative strategies to combat bacterial infections. By targeting multiple signaling pathways, these approaches not only enhance therapeutic effect but also reduce the development of resistance while improving the drug's chemical stability. Despite the urgent need to combat bacterial infectious diseases, there is a notable research gap, in particular in biofilm-related ones. This review highlights the critical importance of exploring polypharmacological approaches with the aim of motivating further research and advances in effective treatments for infectious bacteria, including biofilm related infections.
Collapse
Affiliation(s)
- Miguel M Leitão
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal; CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Ariana S C Gonçalves
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Fernanda Borges
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Manuel Simões
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal; DEQB-Department of Chemical and Biological Engineering, Faculty of Engineering, University of Porto, Porto, Portugal.
| | - Anabela Borges
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal; DEQB-Department of Chemical and Biological Engineering, Faculty of Engineering, University of Porto, Porto, Portugal.
| |
Collapse
|
4
|
Naga NG, El-Badan DE, Mabrouk MEM, Rateb HS, Ghanem KM, Shaaban MI. Innovative application of ceftriaxone as a quorum sensing inhibitor in Pseudomonas aeruginosa. Sci Rep 2025; 15:5022. [PMID: 39934154 PMCID: PMC11814147 DOI: 10.1038/s41598-025-87609-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative bacterium that has the potential to induce various healthcare-related infections through its array of virulence factors. The control of virulence factor expression is mainly regulated by a communication process among cells called quorum sensing (QS). Blocking QS could be a viable tactic to suppress virulence factors and reduce pathogenicity without impacting bacterial growth. This approach has the potential to significantly decrease the multiple drug resistance emergence. In this study, we explored the impact of ceftriaxone (CRO), which is a commonly used β-lactam antibiotic, and its metal derivatives on the QS system and virulence factors of both standard strains and clinical isolates of P. aeruginosa. The quorum sensing inhibitory (QSI) activity of CRO and ceftriaxone Nickel complex (CRON) was evaluated. The minimum inhibitory concentration (MIC) was determined and the effect of sub-MICs of CRO and CRON was assessed on P. aeruginosa strains virulence factors. CRO and CRON effectively suppressed the virulence factors of P. aeruginosa strains at sub-MICs, without altering bacterial viability. Additionally, a molecular docking investigation was carried out to identify potential mechanisms of QSI. CRO and CRON exhibited high ICM scores, potentially displacing natural ligands when interacting with LasR, LasI, and PqsR receptors.
Collapse
Affiliation(s)
- Nourhan G Naga
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Dalia E El-Badan
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria, Egypt
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut, Lebanon
| | - Mona E M Mabrouk
- Botany and Microbiology Department, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Heba S Rateb
- Department of Pharmaceutical and Medicinal Chemistry, Pharmacy College, Misr University for Science and Technology, Cairo, Egypt
| | - Khaled M Ghanem
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mona I Shaaban
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
5
|
San Mauro AJS, Høiby N, Ciofu O. Increased susceptibility to azithromycin of Pseudomonas aeruginosa biofilms using RPMI 1640 testing media. APMIS 2024; 132:1086-1095. [PMID: 38622982 PMCID: PMC11582341 DOI: 10.1111/apm.13413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 03/20/2024] [Indexed: 04/17/2024]
Abstract
Azithromycin (AZM) is efficient for treatment of chronic Pseudomonas aeruginosa biofilm lung infections, despite of resistance in conventional susceptibility testing. It has been shown that planktonic P. aeruginosa are more susceptible to AZM when tested in RPMI 1640 medium. The aim of the study was to test the susceptibility to AZM of P. aeruginosa biofilms in LB vs RPMI 1640 media. We investigated the effect of AZM on planktonic and biofilms of (WT) P. aeruginosa (PAO1), the hypermutable (ΔmutS) and the antibiotic-resistant phenotype(ΔnfxB) mutants. The effect of AZM on young and mature biofilms was investigated in the modified Calgary Biofilm Device by estimation of the minimal biofilm inhibitory concentration (MBIC). The AZM MBIC90 in LB/RPMI1640 on young biofilms treated for 24 h was 16/4 μg/mL for PAO1, 32/8 μg/mL for ΔmutS, and 256/16 μg/mL for ΔnfxB, while in mature biofilms was 256/2 μg/mL for PAO1 and ΔmutS and 16/1 μg/mL for ΔnfxB. The effect of AZM was improved when the treatment was prolonged to 72 h, supporting the intracellular accumulation of AZM. An increased susceptibility of P. aeruginosa biofilms to AZM was observed in RPMI 1640 than in LB medium. Our results might improve susceptibility testing and dosing of AZM for treatment of biofilm infections.
Collapse
Affiliation(s)
| | - Niels Høiby
- Institute of Immunology & Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Oana Ciofu
- Institute of Immunology & Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Dallal Bashi YH, Ali A, Al Ayoub Y, Assi KH, Mairs R, McCarthy HO, Tunney MM, Kett VL. Inhaled dry powder liposomal azithromycin for treatment of chronic lower respiratory tract infection. Int J Pharm 2024; 653:123841. [PMID: 38266939 DOI: 10.1016/j.ijpharm.2024.123841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/05/2024] [Accepted: 01/20/2024] [Indexed: 01/26/2024]
Abstract
A dry powder inhaled liposomal azithromycin formulation was developed for the treatment of chronic respiratory diseases such as cystic fibrosis and bronchiectasis. Key properties including liposome size, charge and encapsulation efficiency powder size, shape, glass transition temperature (Tg), water content and in vitro respiratory deposition were determined. Antimicrobial activity against cystic fibrosis (CF) respiratory pathogens was determined by MIC, MBC and biofilm assays. Cytotoxicity and cellular uptake studies were performed using A549 cells. The average liposome size was 105 nm, charge was 55 mV and encapsulation efficiency was 75 %. The mean powder particle size d[v,50] of 4.54 µm and Mass Median Aerodynamic Diameter (MMAD) was 5.23 µm with a mean Tg of 76˚C and water content of 2.1 %. These excellent physicochemical characteristics were maintained over one year. Liposomal loaded azithromycin demonstrated enhanced activity against P. aeruginosa clinical isolates grown in biofilm. The formulation was rapidly delivered into bacterial cells with > 75 % uptake in 1 h. Rapid uptake into A549 cells via a cholesterol-dependent endocytosis pathway with no cytotoxic effects apparent. These data demonstrate that this formulation could offer benefits over current treatment regimens for people with chronic respiratory infection.
Collapse
Affiliation(s)
| | - Ahlam Ali
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Yuosef Al Ayoub
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK; School of Pharmacy, University of Bradford, UK
| | - Khaled H Assi
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK; School of Pharmacy, University of Bradford, UK
| | - Rachel Mairs
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Michael M Tunney
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Vicky L Kett
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK.
| |
Collapse
|
7
|
Goodyear MC, Seidel L, Krieger JR, Geddes-McAlister J, Levesque RC, Khursigara CM. Quantitative proteomics reveals unique responses to antimicrobial treatments in clinical Pseudomonas aeruginosa isolates. mSystems 2023; 8:e0049123. [PMID: 37623324 PMCID: PMC10654054 DOI: 10.1128/msystems.00491-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/05/2023] [Indexed: 08/26/2023] Open
Abstract
IMPORTANCE Pseudomonas aeruginosa is an important pathogen often associated with hospital-acquired infections and chronic lung infections in people with cystic fibrosis. P. aeruginosa possesses a wide array of intrinsic and adaptive mechanisms of antibiotic resistance, and the regulation of these mechanisms is complex. Label-free quantitative proteomics is a powerful tool to compare susceptible and resistant strains of bacteria and their responses to antibiotic treatments. Here we compare the proteomes of three isolates of P. aeruginosa with different antibiotic resistance profiles in response to five challenge conditions. We uncover unique and shared proteome changes for the widely used laboratory strain PAO1 and two isolates of the Liverpool epidemic strain of P. aeruginosa, LESlike1 and LESB58. Our data set provides insight into antibiotic resistance in clinically relevant Pseudomonas isolates and highlights proteins, including those with uncharacterized functions, which can be further investigated for their role in adaptive responses to antibiotic treatments.
Collapse
Affiliation(s)
- Mara C. Goodyear
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Laura Seidel
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | | | | | - Roger C. Levesque
- Institut de biologie integrative et des systems (IBIS), Département de microbiologie-infectiologie et d'immunologie, Université Laval, Laval, Quebec, Canada
| | - Cezar M. Khursigara
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
8
|
Moshynets OV, Baranovskyi TP, Iungin OS, Krikunov AA, Potochilova VV, Rudnieva KL, Potters G, Pokholenko I. Therapeutic Potential of an Azithromycin-Colistin Combination against XDR K. pneumoniae in a 3D Collagen-Based In Vitro Wound Model of a Biofilm Infection. Antibiotics (Basel) 2023; 12:antibiotics12020293. [PMID: 36830203 PMCID: PMC9952533 DOI: 10.3390/antibiotics12020293] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/20/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023] Open
Abstract
A therapeutic combination of azithromycin (AZM) and colistin methanesulfonate (CMS) was shown to be effective against both non-PDR and PDR Klebsiella pneumoniae biofilms in vitro. These anti-biofilm effects, however, may not correlate with effects observed in standard plate assays, nor will they representative of in vivo therapeutic action. After all, biofilm-associated infection processes are also impacted by the presence of wound bed components, such as host cells or wound fluids, which can all affect the antibiotic effectiveness. Therefore, an in vitro wound model of biofilm infection which partially mimics the complex microenvironment of infected wounds was developed to investigate the therapeutic potential of an AZM-CMS combination against XDR K. pneumoniae isolates. The model consists of a 3D collagen sponge-like scaffold seeded with HEK293 cells submerged in a fluid milieu mimicking the wound bed exudate. Media that were tested were all based on different strengths of Dulbecco's modified Eagles/high glucose medium supplemented with fetal bovine serum, and/or Bacto Proteose peptone. Use of this model confirmed AZM to be a highly effective antibiofilm component, when applied alone or in combination with CMS, whereas CMS alone had little antibacterial effectiveness or even stimulated biofilm development. The wound model proposed here proves therefore, to be an effective aid in the study of drug combinations under realistic conditions.
Collapse
Affiliation(s)
- Olena V. Moshynets
- Biofilm Study Group, Department of Cell Regulatory Mechanisms, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Zabolotnoho Str. 150, 03680 Kyiv, Ukraine
- Correspondence: (O.V.M.); (G.P.)
| | - Taras P. Baranovskyi
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, A-1090 Vienna, Austria
| | - Olga S. Iungin
- Biofilm Study Group, Department of Cell Regulatory Mechanisms, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Zabolotnoho Str. 150, 03680 Kyiv, Ukraine
- Department of Biotechnology, Leather and Fur, Faculty of Chemical and Biopharmaceutical Technologies, Kyiv National University of Technologies and Design, Nemyrovycha-Danchenka Street 2, 01011 Kyiv, Ukraine
| | - Alexey A. Krikunov
- National Amosov Institute of Cardio-Vascular Surgery Affiliated to National Academy of Medical Sciences of Ukraine, Amosov Str. 6, 02000 Kyiv, Ukraine
| | | | - Kateryna L. Rudnieva
- Kyiv Regional Clinical Hospital, Baggovutovskaya Str. 1, 04107 Kyiv, Ukraine
- Department of Microbiology, Virology and Immunology, Bogomolets National Medical University, Shevchenka Blvd. 13, 01601 Kyiv, Ukraine
| | - Geert Potters
- Antwerp Maritime Academy, Noordkasteel Oost 6, 2030 Antwerp, Belgium
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
- Correspondence: (O.V.M.); (G.P.)
| | - Ianina Pokholenko
- Department of Cell Regulatory Mechanisms, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 150 Zabolotnoho Str., 03680 Kyiv, Ukraine
- The Polymer Chemistry & Biomaterials Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| |
Collapse
|
9
|
Ramakrishnan R, Singh AK, Singh S, Chakravortty D, Das D. Enzymatic dispersion of biofilms: An emerging biocatalytic avenue to combat biofilm-mediated microbial infections. J Biol Chem 2022; 298:102352. [PMID: 35940306 PMCID: PMC9478923 DOI: 10.1016/j.jbc.2022.102352] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 01/01/2023] Open
Abstract
Drug resistance by pathogenic microbes has emerged as a matter of great concern to mankind. Microorganisms such as bacteria and fungi employ multiple defense mechanisms against drugs and the host immune system. A major line of microbial defense is the biofilm, which comprises extracellular polymeric substances that are produced by the population of microorganisms. Around 80% of chronic bacterial infections are associated with biofilms. The presence of biofilms can increase the necessity of doses of certain antibiotics up to 1000-fold to combat infection. Thus, there is an urgent need for strategies to eradicate biofilms. Although a few physicochemical methods have been developed to prevent and treat biofilms, these methods have poor efficacy and biocompatibility. In this review, we discuss the existing strategies to combat biofilms and their challenges. Subsequently, we spotlight the potential of enzymes, in particular, polysaccharide degrading enzymes, for biofilm dispersion, which might lead to facile antimicrobial treatment of biofilm-associated infections.
Collapse
Affiliation(s)
- Reshma Ramakrishnan
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | - Ashish Kumar Singh
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Simran Singh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Debasis Das
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, Karnataka, India.
| |
Collapse
|
10
|
Kalgudi R, Tamimi R, Kyazze G, Keshavarz T. Effect of quorum quenchers on virulence factors production and quorum sensing signalling pathway of non-mucoid, mucoid, and heavily mucoid Pseudomonas aeruginosa. World J Microbiol Biotechnol 2022; 38:163. [PMID: 35835899 PMCID: PMC9283346 DOI: 10.1007/s11274-022-03339-9] [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: 02/07/2022] [Accepted: 06/15/2022] [Indexed: 11/28/2022]
Abstract
Quorum quenching (QQ), a mechanism which inhibits, interferes or inactivates quorum sensing, has been investigated for control of biofilms instigated by quorum sensing process. Application of quorum quenchers (QQs) provides the possibility to investigate how different phenotypes of Pseudomonas aeruginosa (non-mucoid, mucoid, and heavily mucoid strains) modulate their gene expression to form biofilms, their quorum sensing (QS) mediated biofilm to be formed, and their virulence expressed. The mRNA expression of the AHL-mediated QS circuit and AHL-mediated virulence factors in P. aeruginosa was investigated in presence of QQs. qPCR analysis showed that farnesol and tyrosol actively reduce the expression of the synthase protein, LasI and RhlI, and prevent production of 3OC12-HSL and C4-HSL, respectively. Also, the use of farnesol and tyrosol significantly moderated gene expression for exo-proteins toxA, aprA, LasB, as well as rhlAB, which are responsible for rhamnolipid production. Our findings were promising, identifying several suppressive regulatory effects of furanone and Candida albicans QS signal molecules, tyrosol, and farnesol on the AHL-mediated P. aeruginosa QS network and related virulence factors.
Collapse
Affiliation(s)
- Rachith Kalgudi
- School of Life Sciences, University of Westminster, 115 New Cavendish Street, W1W 6UW, London, UK.
| | - Roya Tamimi
- School of Life Sciences, University of Westminster, 115 New Cavendish Street, W1W 6UW, London, UK
| | - Godfrey Kyazze
- School of Life Sciences, University of Westminster, 115 New Cavendish Street, W1W 6UW, London, UK
| | - Tajalli Keshavarz
- School of Life Sciences, University of Westminster, 115 New Cavendish Street, W1W 6UW, London, UK
| |
Collapse
|
11
|
Moshynets OV, Baranovskyi TP, Cameron S, Iungin OS, Pokholenko I, Jerdan R, Kamyshnyi A, Krikunov AA, Potochilova VV, Rudnieva KL, Spiers AJ. Azithromycin possesses biofilm–inhibitory activity and potentiates non-bactericidal colistin methanesulfonate (CMS) and polymyxin B against Klebsiella pneumonia. PLoS One 2022; 17:e0270983. [PMID: 35776759 PMCID: PMC9249213 DOI: 10.1371/journal.pone.0270983] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/21/2022] [Indexed: 11/30/2022] Open
Abstract
Novel antibiotic combinations may act synergistically to inhibit the growth of multidrug-resistant bacterial pathogens but predicting which combination will be successful is difficult, and standard antimicrobial susceptibility testing may not identify important physiological differences between planktonic free-swimming and biofilm-protected surface-attached sessile cells. Using a nominally macrolide-resistant model Klebsiella pneumoniae strain (ATCC 10031) we demonstrate the effectiveness of several macrolides in inhibiting biofilm growth in multi-well plates, and the ability of azithromycin (AZM) to improve the effectiveness of the antibacterial last-agent-of-choice for K. pneumoniae infections, colistin methanesulfonate (CMS), against biofilms. This synergistic action was also seen in biofilm tests of several K. pneumoniae hospital isolates and could also be identified in polymyxin B disc-diffusion assays on azithromycin plates. Our work highlights the complexity of antimicrobial-resistance in bacterial pathogens and the need to test antibiotics with biofilm models where potential synergies might provide new therapeutic opportunities not seen in liquid culture or colony-based assays.
Collapse
Affiliation(s)
- Olena V. Moshynets
- Institute of Molecular Biology and Genetics of the National Academy of Sciences of Ukraine, Kyiv, Ukraine
- * E-mail: (OVM); (AJS)
| | - Taras P. Baranovskyi
- Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine
- Kyiv Regional Clinical Hospital, Kyiv, Ukraine
| | - Scott Cameron
- School of Applied Sciences, Abertay University, Dundee, United Kingdom
| | - Olga S. Iungin
- Institute of Molecular Biology and Genetics of the National Academy of Sciences of Ukraine, Kyiv, Ukraine
- Kyiv National University of Technologies and Design, Kyiv, Ukraine
| | - Ianina Pokholenko
- Institute of Molecular Biology and Genetics of the National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Robyn Jerdan
- School of Applied Sciences, Abertay University, Dundee, United Kingdom
| | | | | | | | | | - Andrew J. Spiers
- School of Applied Sciences, Abertay University, Dundee, United Kingdom
- * E-mail: (OVM); (AJS)
| |
Collapse
|
12
|
Deryabin DG, Galadzhieva AA, Kosyan DB, Duskaev GK. Plant-Derived Inhibitors of Density-Dependent Communication in Bacteria: Diversity of Structures, Bioactivity Mechanisms, and Sources of Origin. Microbiology (Reading) 2021. [DOI: 10.1134/s0026261721060059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
13
|
Nolan C, Behrends V. Sub-Inhibitory Antibiotic Exposure and Virulence in Pseudomonas aeruginosa. Antibiotics (Basel) 2021; 10:antibiotics10111393. [PMID: 34827331 PMCID: PMC8615142 DOI: 10.3390/antibiotics10111393] [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: 10/22/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/20/2022] Open
Abstract
Pseudomonas aeruginosa is a prime opportunistic pathogen, one of the most important causes of hospital-acquired infections and the major cause of morbidity and mortality in cystic fibrosis lung infections. One reason for the bacterium's pathogenic success is the large array of virulence factors that it can employ. Another is its high degree of intrinsic and acquired resistance to antibiotics. In this review, we first summarise the current knowledge about the regulation of virulence factor expression and production. We then look at the impact of sub-MIC antibiotic exposure and find that the virulence-antibiotic interaction for P. aeruginosa is antibiotic-specific, multifaceted, and complex. Most studies undertaken to date have been in vitro assays in batch culture systems, involving short-term (<24 h) antibiotic exposure. Therefore, we discuss the importance of long-term, in vivo-mimicking models for future work, particularly highlighting the need to account for bacterial physiology, which by extension governs both virulence factor expression and antibiotic tolerance/resistance.
Collapse
|
14
|
Naga NG, El-Badan DE, Rateb HS, Ghanem KM, Shaaban MI. Quorum Sensing Inhibiting Activity of Cefoperazone and Its Metallic Derivatives on Pseudomonas aeruginosa. Front Cell Infect Microbiol 2021; 11:716789. [PMID: 34660340 PMCID: PMC8515130 DOI: 10.3389/fcimb.2021.716789] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/15/2021] [Indexed: 12/20/2022] Open
Abstract
The last decade has witnessed a massive increase in the rate of mortalities caused by multidrug-resistant Pseudomonas aeruginosa. Therefore, developing new strategies to control virulence factors and pathogenicity has received much attention. One of these strategies is quorum sensing inhibition (QSI) which was developed to control Pseudomonas infection. This study aims to validate the effect of one of the most used β-lactam antibiotics; cefoperazone (CFP) and its metallic-derivatives on quorum sensing (QS) and virulence factors of P. aeruginosa. Assessment of quorum sensing inhibitory activity of CFP, cefoperazone Iron complex (CFPF) and cefoperazone Cobalt complex (CFPC) was performed by using reporter strain Chromobacterium violaceum ATCC 12472. Minimal inhibitory concentration (MIC) was carried out by the microbroth dilution method. The influence of sub-MICs (1/4 and 1/2 MICs) of CFP, CFPF and CFPC on virulence factors of P. aeruginosa was evaluated. Data was confirmed on the molecular level by RT-PCR. Also, molecular docking analysis was conducted to figure out the possible mechanisms of QSI. CFP, CFPF, and CFPC inhibited violacein pigment production of C. violaceum ATCC 12472. Sub-MICs of CFP (128- 256 μg/mL), and significantly low concentrations of CFPC (0.5- 16 μg/mL) and CFPF (0.5- 64 μg/mL) reduced the production of QS related virulence factors such as pyocyanin, protease, hemolysin and eliminated biofilm assembly by P. aeruginosa standard strains PAO1 and PA14, and P. aeruginosa clinical isolates Ps1, Ps2, and Ps3, without affecting bacterial viability. In addition, CFP, CFPF, and CFPC significantly reduced the expression of lasI and rhlI genes. The molecular docking analysis elucidated that the QS inhibitory effect was possibly caused by the interaction with QS receptors. Both CFPF and CFPC interacted strongly with LasI, LasR and PqsR receptors with a much high ICM scores compared to CFP that could be the cause of elimination of natural ligand binding. Therefore, CFPC and CFPF are potent inhibitors of quorum sensing signaling and virulence factors of P. aeruginosa.
Collapse
Affiliation(s)
- Nourhan G Naga
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Dalia E El-Badan
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Heba S Rateb
- Department of Pharmaceutical and Medicinal Chemistry, Pharmacy College, Misr University for Science and Technology, Cairo, Egypt
| | - Khaled M Ghanem
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mona I Shaaban
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
15
|
Sedlmayer F, Woischnig AK, Unterreiner V, Fuchs F, Baeschlin D, Khanna N, Fussenegger M. 5-Fluorouracil blocks quorum-sensing of biofilm-embedded methicillin-resistant Staphylococcus aureus in mice. Nucleic Acids Res 2021; 49:e73. [PMID: 33856484 PMCID: PMC8287944 DOI: 10.1093/nar/gkab251] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 03/02/2021] [Accepted: 03/29/2021] [Indexed: 02/05/2023] Open
Abstract
Antibiotic-resistant pathogens often escape antimicrobial treatment by forming protective biofilms in response to quorum-sensing communication via diffusible autoinducers. Biofilm formation by the nosocomial pathogen methicillin-resistant Staphylococcus aureus (MRSA) is triggered by the quorum-sensor autoinducer-2 (AI-2), whose biosynthesis is mediated by methylthioadenosine/S-adenosylhomocysteine nucleosidase (MTAN) and S-ribosylhomocysteine lyase (LuxS). Here, we present a high-throughput screening platform for small-molecular inhibitors of either enzyme. This platform employs a cell-based assay to report non-toxic, bioavailable and cell-penetrating inhibitors of AI-2 production, utilizing engineered human cells programmed to constitutively secrete AI-2 by tapping into the endogenous methylation cycle via ectopic expression of codon-optimized MTAN and LuxS. Screening of a library of over 5000 commercial compounds yielded 66 hits, including the FDA-licensed cytostatic anti-cancer drug 5-fluorouracil (5-FU). Secondary screening and validation studies showed that 5-FU is a potent quorum-quencher, inhibiting AI-2 production and release by MRSA, Staphylococcus epidermidis, Escherichia coli and Vibrio harveyi. 5-FU efficiently reduced adherence and blocked biofilm formation of MRSA in vitro at an order-of-magnitude-lower concentration than that clinically relevant for anti-cancer therapy. Furthermore, 5-FU reestablished antibiotic susceptibility and enabled daptomycin-mediated prevention and clearance of MRSA infection in a mouse model of human implant-associated infection.
Collapse
Affiliation(s)
- Ferdinand Sedlmayer
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Anne-Kathrin Woischnig
- Laboratory of Infection Biology, Department of Biomedicine, University and University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Vincent Unterreiner
- Novartis Institutes for BioMedical Research (NIBR), Chemical Biology and Therapeutics (CBT), CH-4033, Basel, Switzerland
| | - Florian Fuchs
- Novartis Institutes for BioMedical Research (NIBR), Chemical Biology and Therapeutics (CBT), CH-4033, Basel, Switzerland
| | - Daniel Baeschlin
- Novartis Institutes for BioMedical Research (NIBR), Chemical Biology and Therapeutics (CBT), CH-4033, Basel, Switzerland
| | - Nina Khanna
- Laboratory of Infection Biology, Department of Biomedicine, University and University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, CH-4058 Basel, Switzerland
- Faculty of Science, University of Basel, Mattenstrasse 26, CH-4058 Basel, Switzerland
| |
Collapse
|
16
|
Konikkat S, Scribner MR, Eutsey R, Hiller NL, Cooper VS, McManus J. Quantitative mapping of mRNA 3' ends in Pseudomonas aeruginosa reveals a pervasive role for premature 3' end formation in response to azithromycin. PLoS Genet 2021; 17:e1009634. [PMID: 34252072 PMCID: PMC8297930 DOI: 10.1371/journal.pgen.1009634] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/22/2021] [Accepted: 06/01/2021] [Indexed: 01/06/2023] Open
Abstract
Pseudomonas aeruginosa produces serious chronic infections in hospitalized patients and immunocompromised individuals, including patients with cystic fibrosis. The molecular mechanisms by which P. aeruginosa responds to antibiotics and other stresses to promote persistent infections may provide new avenues for therapeutic intervention. Azithromycin (AZM), an antibiotic frequently used in cystic fibrosis treatment, is thought to improve clinical outcomes through a number of mechanisms including impaired biofilm growth and quorum sensing (QS). The mechanisms underlying the transcriptional response to AZM remain unclear. Here, we interrogated the P. aeruginosa transcriptional response to AZM using a fast, cost-effective genome-wide approach to quantitate RNA 3’ ends (3pMap). We also identified hundreds of P. aeruginosa genes with high incidence of premature 3’ end formation indicative of riboregulation in their transcript leaders using 3pMap. AZM treatment of planktonic and biofilm cultures alters the expression of hundreds of genes, including those involved in QS, biofilm formation, and virulence. Strikingly, most genes downregulated by AZM in biofilms had increased levels of intragenic 3’ ends indicating premature transcription termination, transcriptional pausing, or accumulation of stable intermediates resulting from the action of nucleases. Reciprocally, AZM reduced premature intragenic 3’ end termini in many upregulated genes. Most notably, reduced termination accompanied robust induction of obgE, a GTPase involved in persister formation in P. aeruginosa. Our results support a model in which AZM-induced changes in 3’ end formation alter the expression of central regulators which in turn impairs the expression of QS, biofilm formation and stress response genes, while upregulating genes associated with persistence. Pseudomonas aeruginosa is a common source of hospital-acquired infections and causes prolonged illness in patients with cystic fibrosis. P. aeruginosa infections are often treated with the macrolide antibiotic azithromycin, which changes the expression of many genes involved in infection. By examining such expression changes at nucleotide resolution, we found azithromycin treatment alters the locations of mRNA 3’ ends suggesting most downregulated genes are subject to premature 3’ end formation. We further identified candidate RNA regulatory elements that P. aeruginosa may use to control gene expression. Our work provides new insights in P. aeruginosa gene regulation and its response to antibiotics.
Collapse
Affiliation(s)
- Salini Konikkat
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Michelle R. Scribner
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Rory Eutsey
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - N. Luisa Hiller
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Vaughn S. Cooper
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Joel McManus
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
17
|
Dynamic Adaptive Response of Pseudomonas aeruginosa to Clindamycin/Rifampicin-Impregnated Catheters. Antibiotics (Basel) 2021; 10:antibiotics10070752. [PMID: 34206280 PMCID: PMC8300626 DOI: 10.3390/antibiotics10070752] [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: 05/26/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 11/16/2022] Open
Abstract
Pseudomonas aeruginosa is the most common Gram-negative pathogen causing nosocomial multidrug resistant infections. It is a good biofilm producer and has the potential for contaminating medical devices. Despite the widespread use of antibacterial-impregnated catheters, little is known about the impacts of antibacterial coating on the pathogenesis of P. aeruginosa. In this study, we investigated the adaptive resistance potential of P. aeruginosa strain PAO1 in response to continuous antibiotic exposure from clindamycin/rifampicin-impregnated catheters (CR-IC). During exposure for 144 h to clindamycin and rifampicin released from CR-IC, strain PAO1 formed biofilms featuring elongated and swollen cells. There were 545 and 372 differentially expressed proteins (DEPs) identified in the planktonic and biofilm cells, respectively, by ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). Both Cluster of Orthologous Groups (COG) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses showed that the planktonic cells responded to the released antibiotics more actively than the biofilm cells, with metabolism and ribosomal biosynthesis-associated proteins being significantly over-expressed. Exposure to CR-IC increased the invasion capability of P. aeruginosa for Hela cells and upregulated the expression of certain groups of virulence proteins in both planktonic and biofilm cells, including the outer membrane associated (flagella, type IV pili and type III secretion system) and extracellular (pyoverdine) virulence proteins. Continuous exposure of P. aeruginosa to CR-IC also induced the overexpression of antibiotic resistance proteins, including porins, efflux pumps, translation and transcription proteins. However, these upregulations did not change phenotypic minimum inhibitory concentration (MIC) during the experimental timeframe. The concerning association between CR-IC and overexpression of virulence factors in P. aeruginosa suggests the need for additional investigation to determine if it results in adverse clinical outcomes.
Collapse
|
18
|
Thomsen K, Kobayashi O, Kishi K, Shirai R, Østrup Jensen P, Heydorn A, Hentzer M, Calum H, Christophersen L, Høiby N, Moser C. Animal models of chronic and recurrent Pseudomonas aeruginosa lung infection: significance of macrolide treatment. APMIS 2021; 130:458-476. [PMID: 34117660 DOI: 10.1111/apm.13161] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 11/28/2022]
Abstract
Animal models of human diseases are invaluable and inevitable elements in identifying and testing novel treatments for serious diseases, including severe infections. Planning and conducting investigator-initiated human trials are generally accepted as being enormously challenging. In contrast, it is often underestimated how much planning, including background and modifying experiments, is needed to establish a relevant infectious disease animal model. However, representative animal infectious models, well designed to test generated hypotheses, are useful to improve our understanding of pathogenesis, virulence factors and host response and to identify novel treatment candidates and therapeutic strategies. Such results can subsequently proceed to clinical testing if suitable. The present review aims at presenting all the pulmonary Pseudomonas aeruginosa infectious models we have knowledge of and the detailed descriptions of established animal models in our laboratory focusing on macrolide therapy are presented.
Collapse
Affiliation(s)
- Kim Thomsen
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Osamu Kobayashi
- Department of Infectious Diseases, National Cancer Center Hospital, Tokyo, Japan
| | - Kenji Kishi
- Oitaken Kouseiren Tsurumi Hospital, Tsurumi, Beppu City, Japan
| | - Ryo Shirai
- Department of Internal Medicine, Kawasaki Medical School, General Medical Center, Okayama, Japan
| | - Peter Østrup Jensen
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Arne Heydorn
- Department of Endocrinology and Nephrology, Nordsjaellands Hospital, Hillerød, Denmark
| | - Morten Hentzer
- Department of Molecular Pharmacology, H. Lundbeck A/S, Copenhagen, Denmark
| | - Henrik Calum
- Department of Clinical Microbiology, Hvidovre Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lars Christophersen
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Niels Høiby
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Claus Moser
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Kumar M, Rao M, Mathur T, Barman TK, Joshi V, Chaira T, Singhal S, Pandya M, Al Khodor S, Upadhyay DJ, Masuda N. Azithromycin Exhibits Activity Against Pseudomonas aeruginosa in Chronic Rat Lung Infection Model. Front Microbiol 2021; 12:603151. [PMID: 33967970 PMCID: PMC8102702 DOI: 10.3389/fmicb.2021.603151] [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: 09/05/2020] [Accepted: 03/15/2021] [Indexed: 12/19/2022] Open
Abstract
Pseudomonas aeruginosa forms biofilms in the lungs of chronically infected cystic fibrosis patients, which are tolerant to both the treatment of antibiotics and the host immune system. Normally, antibiotics are less effective against bacteria growing in biofilms; azithromycin has shown a potent efficacy in cystic fibrosis patients chronically infected with P. aeruginosa and improved their lung function. The present study was conducted to evaluate the effect of azithromycin on P. aeruginosa biofilm. We show that azithromycin exhibited a potent activity against P. aeruginosa biofilm, and microscopic observation revealed that azithromycin substantially inhibited the formation of solid surface biofilms. Interestingly, we observed that azithromycin restricted P. aeruginosa biofilm formation by inhibiting the expression of pel genes, which has been previously shown to play an essential role in bacterial attachment to solid-surface biofilm. In a rat model of chronic P. aeruginosa lung infection, we show that azithromycin treatment resulted in the suppression of quorum sensing-regulated virulence factors, significantly improving the clearance of P. aeruginosa biofilms compared to that in the placebo control. We conclude that azithromycin attenuates P. aeruginosa biofilm formation, impairs its ability to produce extracellular biofilm matrix, and increases its sensitivity to the immune system, which may explain the clinical efficacy of azithromycin in cystic fibrosis patients.
Collapse
Affiliation(s)
- Manoj Kumar
- Department of Microbiology, Daiichi Sankyo India Pharma Private Limited, Gurgaon, India.,Research Department, Sidra Medicine, Doha, Qatar
| | - Madhvi Rao
- Department of Microbiology, Daiichi Sankyo India Pharma Private Limited, Gurgaon, India
| | - Tarun Mathur
- Department of Microbiology, Daiichi Sankyo India Pharma Private Limited, Gurgaon, India
| | - Tarani Kanta Barman
- Department of Microbiology, Daiichi Sankyo India Pharma Private Limited, Gurgaon, India
| | - Vattan Joshi
- Department of Microbiology, Daiichi Sankyo India Pharma Private Limited, Gurgaon, India
| | - Tridib Chaira
- Department of Pharmacokinetics and Metabolism, Daiichi Sankyo India Pharma Private Limited, Gurgaon, India
| | - Smita Singhal
- Department of Microbiology, Daiichi Sankyo India Pharma Private Limited, Gurgaon, India
| | - Manisha Pandya
- Department of Microbiology, Daiichi Sankyo India Pharma Private Limited, Gurgaon, India
| | | | - Dilip J Upadhyay
- Department of Microbiology, Daiichi Sankyo India Pharma Private Limited, Gurgaon, India
| | - Nobuhisa Masuda
- Department of Microbiology, Daiichi Sankyo India Pharma Private Limited, Gurgaon, India
| |
Collapse
|
20
|
Leroy AG, Caillon J, Caroff N, Broquet A, Corvec S, Asehnoune K, Roquilly A, Crémet L. Could Azithromycin Be Part of Pseudomonas aeruginosa Acute Pneumonia Treatment? Front Microbiol 2021; 12:642541. [PMID: 33796090 PMCID: PMC8008145 DOI: 10.3389/fmicb.2021.642541] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/26/2021] [Indexed: 12/29/2022] Open
Abstract
Azithromycin (AZM) is a 15-membered-ring macrolide that presents a broad-spectrum antimicrobial activity against Gram-positive bacteria and atypical microorganisms but suffers from a poor diffusion across the outer-membrane of Gram-negative bacilli, including Pseudomonas aeruginosa (PA). However, AZM has demonstrated clinical benefits in patients suffering from chronic PA respiratory infections, especially cystic fibrosis patients. Since the rise of multidrug-resistant PA has led to a growing need for new therapeutic options, this macrolide has been proposed as an adjunctive therapy. Clinical trials assessing AZM in PA acute pneumonia are scarce. However, a careful examination of the available literature provides good rationales for its use in that context. In fact, 14- and 15-membered-ring macrolides have demonstrated immunomodulatory and immunosuppressive effects that could be of major interest in the management of acute illness. Furthermore, growing evidence supports a downregulation of PA virulence dependent on direct interaction with the ribosomes, and based on the modulation of several key regulators from the Quorum Sensing network. First highlighted in vitro, these interesting properties of AZM have subsequently been confirmed in the animal models. In this review, we systematically analyzed the literature regarding AZM immunomodulatory and anti-PA effects. In vitro and in vivo studies, as well as clinical trials were reviewed, looking for rationales for AZM use in PA acute pneumonia.
Collapse
Affiliation(s)
- Anne-Gaëlle Leroy
- Laboratoire EA 3826 "Thérapeutiques cliniques et expérimentales des infections", IRS2-Nantes Biotech, Université de Nantes, Nantes, France.,CHU de Nantes, Service de Bactériologie-Hygiène hospitalière, Nantes Université, Nantes, France
| | - Jocelyne Caillon
- Laboratoire EA 3826 "Thérapeutiques cliniques et expérimentales des infections", IRS2-Nantes Biotech, Université de Nantes, Nantes, France.,CHU de Nantes, Service de Bactériologie-Hygiène hospitalière, Nantes Université, Nantes, France
| | - Nathalie Caroff
- Laboratoire EA 3826 "Thérapeutiques cliniques et expérimentales des infections", IRS2-Nantes Biotech, Université de Nantes, Nantes, France
| | - Alexis Broquet
- Laboratoire EA 3826 "Thérapeutiques cliniques et expérimentales des infections", IRS2-Nantes Biotech, Université de Nantes, Nantes, France
| | - Stéphane Corvec
- CHU de Nantes, Service de Bactériologie-Hygiène hospitalière, Nantes Université, Nantes, France.,CRCINA, U1232, CHU Nantes, Nantes, France
| | - Karim Asehnoune
- Laboratoire EA 3826 "Thérapeutiques cliniques et expérimentales des infections", IRS2-Nantes Biotech, Université de Nantes, Nantes, France.,CHU de Nantes, Service Anesthésie Réanimation Chirurgicale, Nantes Université, Nantes, France
| | - Antoine Roquilly
- Laboratoire EA 3826 "Thérapeutiques cliniques et expérimentales des infections", IRS2-Nantes Biotech, Université de Nantes, Nantes, France.,CHU de Nantes, Service Anesthésie Réanimation Chirurgicale, Nantes Université, Nantes, France
| | - Lise Crémet
- Laboratoire EA 3826 "Thérapeutiques cliniques et expérimentales des infections", IRS2-Nantes Biotech, Université de Nantes, Nantes, France.,CHU de Nantes, Service de Bactériologie-Hygiène hospitalière, Nantes Université, Nantes, France
| |
Collapse
|
21
|
Lu L, Li M, Yi G, Liao L, Cheng Q, Zhu J, Zhang B, Wang Y, Chen Y, Zeng M. Screening strategies for quorum sensing inhibitors in combating bacterial infections. J Pharm Anal 2021; 12:1-14. [PMID: 35573879 PMCID: PMC9073242 DOI: 10.1016/j.jpha.2021.03.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 03/04/2021] [Accepted: 03/25/2021] [Indexed: 01/20/2023] Open
Abstract
Interference with quorum sensing (QS) represents an antivirulence strategy with a significant promise for the treatment of bacterial infections and a new approach to restoring antibiotic tolerance. Over the past two decades, a novel series of studies have reported that quorum quenching approaches and the discovery of quorum sensing inhibitors (QSIs) have a strong impact on the discovery of anti-infective drugs against various types of bacteria. The discovery of QSI was demonstrated to be an appropriate strategy to expand the anti-infective therapeutic approaches to complement classical antibiotics and antimicrobial agents. For the discovery of QSIs, diverse approaches exist and develop in-step with the scale of screening as well as specific QS systems. This review highlights the latest findings in strategies and methodologies for QSI screening, involving activity-based screening with bioassays, chemical methods to seek bacterial QS pathways for QSI discovery, virtual screening for QSI screening, and other potential tools for interpreting QS signaling, which are innovative routes for future efforts to discover additional QSIs to combat bacterial infections. Interference with QSrepresents a promising antivirulence strategy for the treatment of bacterial infections. The discovery ofQSIs was demonstrated as an appropriate strategy to expand the anti-infective therapeutic arsenal to complement classical antibiotics and antimicrobial agents. For the discovery of QSIs, diverse approaches exist and develop in-step with the scale of screening and targeted QS systems. Few previous reviews have summarized the strategies and approaches of QSI screening, whereas this review highlights the recent findings in QSI screening strategies and methodologies.
Collapse
Affiliation(s)
- Lan Lu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
- Corresponding author.
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Guojuan Yi
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| | - Li Liao
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| | - Qiang Cheng
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| | - Jie Zhu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| | - Bin Zhang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| | - Yingying Wang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| | - Yong Chen
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| | - Ming Zeng
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| |
Collapse
|
22
|
Magalhães C, Lima M, Trieu-Cuot P, Ferreira P. To give or not to give antibiotics is not the only question. THE LANCET. INFECTIOUS DISEASES 2020; 21:e191-e201. [PMID: 33347816 DOI: 10.1016/s1473-3099(20)30602-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 06/05/2020] [Accepted: 06/25/2020] [Indexed: 02/08/2023]
Abstract
In a 1945 Nobel Lecture, Sir Alexander Fleming warned against the overuse of antibiotics, particularly in response to public pressure. In the subsequent decades, evidence has shown that bacteria can become resistant to almost any available molecule. One key question is how the emergence and dissemination of resistant bacteria or resistance genes can be delayed. Although some clinicians remain sceptical, in this Personal View, we argue that the prescription of fewer antibiotics and shorter treatment duration is just as effective as longer regimens that remain the current guideline. Additionally, we discuss the fact that shorter antibiotic treatments exert less selective pressure on microorganisms, preventing the development of resistance. By contrast, longer treatments associated with a strong selective pressure favour the emergence of resistant clones within commensal organisms. We also emphasise that more studies are needed to identify the optimal duration of antibiotic therapy for common infections, which is important for making changes to the current guidelines, and to identify clinical biomarkers to guide antibiotic treatment in both hospital and ambulatory settings.
Collapse
Affiliation(s)
- Catarina Magalhães
- Department of Immuno-Physiology and Pharmacology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Margarida Lima
- Unidade de Investigação Biomédica do Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal; Department of Hematology, Hospital de Santo António, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Patrick Trieu-Cuot
- Institut Pasteur, Unité de Biologie des Bactéries Pathogènes à Gram-positif, Centre National de la Recherche Scientifique (CNRS UMR 2001), Paris, France
| | - Paula Ferreira
- Department of Immuno-Physiology and Pharmacology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
23
|
Identification of novel targets of azithromycin activity against Pseudomonas aeruginosa grown in physiologically relevant media. Proc Natl Acad Sci U S A 2020; 117:33519-33529. [PMID: 33318204 DOI: 10.1073/pnas.2007626117] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pseudomonas aeruginosa causes severe multidrug-resistant infections that often lead to bacteremia and sepsis. Physiologically relevant conditions can increase the susceptibility of pathogens to antibiotics, such as azithromycin (AZM). When compared to minimal-inhibitory concentrations (MICs) in laboratory media, AZM had a 16-fold lower MIC in tissue culture medium with 5% Mueller Hinton broth (MHB) and a 64-fold lower MIC in this tissue culture medium with 20% human serum. AZM also demonstrated increased synergy in combination with synthetic host-defense peptides DJK-5 and IDR-1018 under host-like conditions and in a murine abscess model. To mechanistically study the altered effects of AZM under physiologically relevant conditions, global transcriptional analysis was performed on P. aeruginosa with and without effective concentrations of AZM. This revealed that the arn operon, mediating arabinosaminylation of lipopolysaccharides and related regulatory systems, was down-regulated in host-like media when compared to MHB. Inactivation of genes within the arn operon led to increased susceptibility of P. aeruginosa to AZM and great increases in synergy between AZM and other antimicrobial agents, indicating that dysregulation of the arn operon might explain increased AZM uptake and synergy in host-like media. Furthermore, genes involved in central and energy metabolism and ribosome biogenesis were dysregulated more in physiologically relevant conditions treated with AZM, likely due to general changes in cell physiology as a result of the increased effectiveness of AZM in these conditions. These data suggest that, in addition to the arn operon, there are multiple factors in host-like environments that are responsible for observed changes in susceptibility.
Collapse
|
24
|
Sörensen M, Khakimov B, Nurjadi D, Boutin S, Yi B, Dalpke AH, Eigenbrod T. Comparative evaluation of the effect of different growth media on in vitro sensitivity to azithromycin in multi-drug resistant Pseudomonas aeruginosa isolated from cystic fibrosis patients. Antimicrob Resist Infect Control 2020; 9:197. [PMID: 33298147 PMCID: PMC7724801 DOI: 10.1186/s13756-020-00859-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/20/2020] [Indexed: 12/26/2022] Open
Abstract
Long-term treatment with azithromycin is a therapeutic option in Cystic Fibrosis (CF) patients chronically infected with P. aeruginosa. It was recently shown that azithromycin has direct antimicrobial activity when P. aeruginosa isolates are tested in Roswell Park Memorial Institute medium supplemented with fetal calf serum (RPMI 1640/FCS) by broth microdilution. We now investigated whether (i) azithromycin might also be active against multidrug resistant (MDR) P. aeruginosa isolated from CF patients and (ii) how in vitro sensitivity assays perform in synthetic cystic fibrosis sputum medium (SCFM), a medium that mimics the particular CF airway environment. In 17 (59%) out of 29 MDR P. aeruginosa CF isolates MICs for azithromycin ranged between 0.25 and 8 μg/ml and 12 isolates (41%) showed a MIC ≥512 μg/ml when measured in RPMI/FCS. In contrast, MICs were ≥ 256 μg/ml for all P. aeruginosa MDR isolates when tested in either SCFM or in conventional cation-adjusted Mueller Hinton Broth. High MIC values observed in CF adapted medium SCFM for both PAO1 and MDR P. aeruginosa CF isolates, as opposed to findings in RPMI, argue against routine azithromycin MIC testing of CF isolates.
Collapse
Affiliation(s)
- Michael Sörensen
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120, Heidelberg, Germany. .,Laboratory Enders and Partners, 70193, Stuttgart, Germany.
| | - Bakhodur Khakimov
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Dennis Nurjadi
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Sébastien Boutin
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Buqing Yi
- Institute of Medical Microbiology and Hygiene, Medical Faculty, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Alexander H Dalpke
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120, Heidelberg, Germany. .,Institute of Medical Microbiology and Hygiene, Medical Faculty, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
| | - Tatjana Eigenbrod
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120, Heidelberg, Germany. .,SLK-Kliniken Heilbronn, 74080, Heilbronn, Germany.
| |
Collapse
|
25
|
Thomsen K, Christophersen L, Lerche CJ, Holmgaard DB, Calum H, Høiby N, Moser C. Azithromycin potentiates avian IgY effect against Pseudomonas aeruginosa in a murine pulmonary infection model. Int J Antimicrob Agents 2020; 57:106213. [PMID: 33256950 DOI: 10.1016/j.ijantimicag.2020.106213] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 10/13/2020] [Accepted: 10/17/2020] [Indexed: 01/25/2023]
Abstract
Cystic fibrosis (CF) patients are at risk of acquiring chronic Pseudomonas aeruginosa lung infections. The biofilm mode of growth of P. aeruginosa induces tolerance to antibiotics and the host response; accordingly, treatment failure occurs. Supplemental azithromycin has proven beneficial in CF owing to potential immunomodulatory mechanisms. Clinical studies have demonstrated a reduction in exacerbations in CF patients by avian IgY anti-Pseudomonas immunotherapy. We hypothesise that azithromycin pre-treatment could potentiate the observed anti-Pseudomonas effect of IgY opsonisation in vivo. Evaluation of phagocytic cell capacity was performed using in vitro exposure of azithromycin pre-treated human polymorphonuclear neutrophils to IgY opsonised P. aeruginosa PAO3. A murine lung infection model using nasal planktonic P. aeruginosa inoculation and successive evaluation 24 h post-infection was used to determine lung bacteriology and subsequent pulmonary inflammation. Combined azithromycin treatment and IgY opsonisation significantly increased bacterial killing compared with the two single-treated groups and controls. In vivo, significantly increased bacterial pulmonary elimination was revealed by combining azithromycin and IgY. A reduction in the inflammatory markers mobiliser granulocyte colony-stimulating factor (G-CSF), macrophage inflammatory protein 2 (MIP-2) and interleukin 1 beta (IL-1β) paralleled this effect. Combination of azithromycin and anti-Pseudomonas IgY potentiated the killing and pulmonary elimination of P. aeruginosa in vitro and in vivo. The augmented effect of combinatory treatment with azithromycin and IgY constitutes a potential clinical application for improving anti-Pseudomonas strategies.
Collapse
Affiliation(s)
- Kim Thomsen
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Henrik Harpestrengsvej 4A, 2100-DK Copenhagen, Denmark.
| | - Lars Christophersen
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Henrik Harpestrengsvej 4A, 2100-DK Copenhagen, Denmark
| | - Christian Johann Lerche
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Henrik Harpestrengsvej 4A, 2100-DK Copenhagen, Denmark
| | | | - Henrik Calum
- Department of Clinical Microbiology, Hvidovre Hospital, Copenhagen, Denmark
| | - Niels Høiby
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Henrik Harpestrengsvej 4A, 2100-DK Copenhagen, Denmark; Institute of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus Moser
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Henrik Harpestrengsvej 4A, 2100-DK Copenhagen, Denmark
| |
Collapse
|
26
|
Is combined medication with natural medicine a promising therapy for bacterial biofilm infection? Biomed Pharmacother 2020; 128:110184. [DOI: 10.1016/j.biopha.2020.110184] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/17/2020] [Accepted: 04/19/2020] [Indexed: 12/11/2022] Open
|
27
|
Bush A. Azithromycin is the answer in paediatric respiratory medicine, but what was the question? Paediatr Respir Rev 2020; 34:67-74. [PMID: 31629643 DOI: 10.1016/j.prrv.2019.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 02/07/2023]
Abstract
The first clinical indication of non-antibiotic benefits of macrolides was in the Far East, in adults with diffuse panbronchiolitis. This condition is characterised by chronic airway infection, often with Pseudomonas aeruginosa, airway inflammation, bronchiectasis and a high mortality. Low dose erythromycin, and subsequently other macrolides, led in many cases to complete remission of the condition, and abrogated the neutrophilic airway inflammation characteristic of the disease. This dramatic finding sparked a flurry of interest in the many hundreds of macrolides in nature, especially their anti-inflammatory and immunomodulatory effects. The biggest subsequent trials of azithromycin were in cystic fibrosis, which has obvious similarities to diffuse panbronchiolitis. There were unquestionable improvements in lung function and pulmonary exacerbations, but compared to diffuse panbronchiolitis, the results were disappointing. Case reports, case series and some randomised controlled trials followed in other conditions. Three trials of azithromycin in preschool wheeze gave contradictory results; a trial in pauci-inflammatory adult asthma, and a trial in non-cystic fibrosis bronchiectasis both showed a significant reduction in exacerbations, but none matched the dramatic results in diffuse panbronchiolitis. There is clearly a huge risk of antibacterial resistance if macrolides are used widely and uncritically in the community. In summary, Azithromycin is not the answer to anything in paediatric respiratory medicine; the paediatric respiratory community needs to refocus on the dramatic benefits of macrolides in diffuse panbronchiolitis, use modern - omics technologies to determine the endotypes of inflammatory diseases and discover in nature or synthesise designer macrolides to replicate the diffuse panbronchiolitis results. We must now find out how to do better!
Collapse
Affiliation(s)
- Andrew Bush
- Professor of Paediatrics and Paediatric Respirology, Imperial College Consultant Paediatric Chest Physician, Royal Brompton & Harefield NHS Foundation Trust, National Heart and Lung Institute, UK; Paediatric Chest Physician, Royal Brompton Harefield NHS Foundation Trust, UK.
| |
Collapse
|
28
|
Deryabin D, Galadzhieva A, Kosyan D, Duskaev G. Plant-Derived Inhibitors of AHL-Mediated Quorum Sensing in Bacteria: Modes of Action. Int J Mol Sci 2019; 20:E5588. [PMID: 31717364 PMCID: PMC6888686 DOI: 10.3390/ijms20225588] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/02/2019] [Accepted: 11/06/2019] [Indexed: 12/15/2022] Open
Abstract
Numerous gram-negative phytopathogenic and zoopathogenic bacteria utilise acylated homoserine lactone (AHL) in communication systems, referred to as quorum sensing (QS), for induction of virulence factors and biofilm development. This phenomenon positions AHL-mediated QS as an attractive target for anti-infective therapy. This review focused on the most significant groups of plant-derived QS inhibitors and well-studied individual compounds for which in silico, in vitro and in vivo studies provide substantial knowledge about their modes of anti-QS activity. The current data about sulfur-containing compounds, monoterpenes and monoterpenoids, phenylpropanoids, benzoic acid derivatives, diarylheptanoids, coumarins, flavonoids and tannins were summarized; their plant sources, anti-QS effects and bioactivity mechanisms have also been summarized and discussed. Three variants of plant-derived molecules anti-QS strategies are proposed: (i) specific, via binding with LuxI-type AHL synthases and/or LuxR-type AHL receptor proteins, which have been shown for terpenes (carvacrol and l-carvone), phenylpropanoids (cinnamaldehyde and eugenol), flavonoid quercetin and ellagitannins; (ii) non-specific, by affecting the QS-related intracellular regulatory pathways by lowering regulatory small RNA expression (sulphur-containing compounds ajoene and iberin) or c-di-GMP metabolism reduction (coumarin); and (iii) indirect, via alteration of metabolic pathways involved in QS-dependent processes (vanillic acid and curcumin).
Collapse
Affiliation(s)
- Dmitry Deryabin
- Federal Scientific Center of Biological Systems and Agrotechnologies of RAS, Orenburg 460000, Russia; (A.G.); (D.K.); (G.D.)
| | | | | | | |
Collapse
|
29
|
Jedrey H, Lilley KS, Welch M. Ciprofloxacin binding to GyrA causes global changes in the proteome of Pseudomonas aeruginosa. FEMS Microbiol Lett 2019; 365:5017444. [PMID: 29846552 PMCID: PMC5995189 DOI: 10.1093/femsle/fny134] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/25/2018] [Indexed: 11/14/2022] Open
Abstract
Ciprofloxacin is one of the most widely-used antibiotics, and has proven especially effective at controlling infections associated with the opportunistic human pathogen, Pseudomonas aeruginosa. In this work, we show that sub-inhibitory concentrations of ciprofloxacin induce discrete changes in the intracellular proteome. Central metabolism and cell envelope-associated functions are particularly affected. In spite of the low magnitude of the intracellular proteomic changes, we found that sub-lethal concentrations of ciprofloxacin had substantial effects on motility and exoprotein secretion. Crucially, the proteomic and phenotypic modulations that we observed were absolutely dependent upon the presence of wild-type GyrA; an isogenic strain of P. aeruginosa carrying a ciprofloxacin-insensitive form of GyrA (a T83→I mutant) did not display ciprofloxacin-dependent changes unless complemented with wild-type gyrA in trans. These results show that the diverse effects of sub-inhibitory ciprofloxacin on the cell are routed through its primary target in the cell, DNA gyrase.
Collapse
Affiliation(s)
- Hannah Jedrey
- Department of Biochemistry, Tennis Court Road, Cambridge, CB2 1QW, UK
| | - Kathryn S Lilley
- Department of Biochemistry, Tennis Court Road, Cambridge, CB2 1QW, UK
| | - Martin Welch
- Department of Biochemistry, Tennis Court Road, Cambridge, CB2 1QW, UK
| |
Collapse
|
30
|
Pattnaik S, Barik S, Muralitharan G, Busi S. Ferulic acid encapsulated chitosan-tripolyphosphate nanoparticles attenuate quorum sensing regulated virulence and biofilm formation in Pseudomonas aeruginosa PAO1. IET Nanobiotechnol 2019; 12:1056-1061. [PMID: 30964013 DOI: 10.1049/iet-nbt.2018.5114] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic nosocomial pathogenic microorganism causing majority of acute hospital-acquired infections and poses a serious public health concern. The persistence of bacterial infection can be attributed to the highly synchronised cell-to-cell communication phenomenon, quorum sensing (QS) which regulates the expression of a number of virulence factors and biofilm formation which eventually imparts resistance to the conventional antimicrobial therapy. In this study, the anti-quorum sensing and anti-biofilm potential of ferulic acid encapsulated chitosan-tripolyphosphate nanoparticles (FANPs) was investigated against P. aeruginosa PAO1 and compared with native ferulic acid. Dynamic light scattering and transmission electron microscopic analysis confirmed the synthesis of FANPs with mean diameter of 215.55 nm. FANPs showed significant anti-quorum sensing activity by downregulating QS-regulated virulence factors. In addition, FANPs also significantly attenuate the swimming and swarming motility of P. aeruginosa PAO1. The anti-biofilm efficacy of FANPs as compared to native ferulic acid was established by light and confocal laser scanning microscopic analysis. The promising results of FANPs in attenuating QS highlighted the slow and sustained release of ferulic acid at the target sites with greater efficacy suggesting its application towards the development of anti-infective agents.
Collapse
Affiliation(s)
- Subhaswaraj Pattnaik
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry-605 014, India
| | - Subhashree Barik
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry-605 014, India
| | - Gangatharan Muralitharan
- Department of Microbiology, School of Life Sciences, Bharathidasan University, Tiruchirappali-620 024, India
| | - Siddhardha Busi
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry-605 014, India.
| |
Collapse
|
31
|
Wu X, Siehnel RJ, Garudathri J, Staudinger BJ, Hisert KB, Ozer EA, Hauser AR, Eng JK, Manoil C, Singh PK, Bruce JE. In Vivo Proteome of Pseudomonas aeruginosa in Airways of Cystic Fibrosis Patients. J Proteome Res 2019; 18:2601-2612. [PMID: 31060355 DOI: 10.1021/acs.jproteome.9b00122] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chronic airway infection with P. aeruginosa (PA) is a hallmark of cystic fibrosis (CF) disease. The mechanisms producing PA persistence in CF therapies remain poorly understood. To gain insight on PA physiology in patient airways and better understand how in vivo bacterial functioning differs from in vitro conditions, we investigated the in vivo proteomes of PA in 35 sputum samples from 11 CF patients. We developed a novel bacterial-enrichment method that relies on differential centrifugation and detergent treatment to enrich for bacteria to improve identification of PA proteome with CF sputum samples. Using two nonredundant peptides as a cutoff, a total of 1304 PA proteins were identified directly from CF sputum samples. The in vivo PA proteomes were compared with the proteomes of ex vivo-grown PA populations from the same patient sample. Label-free quantitation and proteome comparison revealed the in vivo up-regulation of siderophore TonB-dependent receptors, remodeling in central carbon metabolism including glyoxylate cycle and lactate utilization, and alginate overproduction. Knowledge of these in vivo proteome differences or others derived using the presented methodology could lead to future treatment strategies aimed at altering PA physiology in vivo to compromise infectivity or improve antibiotic efficacy.
Collapse
|
32
|
Inchagova KS, Duskaev GK, Deryabin DG. Quorum Sensing Inhibition in Chromobacterium violaceum by Amikacin Combination with Activated Charcoal or Small Plant-Derived Molecules (Pyrogallol and Coumarin). Microbiology (Reading) 2019. [DOI: 10.1134/s0026261719010132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
33
|
Tse BN, Adalja AA, Houchens C, Larsen J, Inglesby TV, Hatchett R. Challenges and Opportunities of Nontraditional Approaches to Treating Bacterial Infections. Clin Infect Dis 2018; 65:495-500. [PMID: 28985671 DOI: 10.1093/cid/cix320] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/10/2017] [Indexed: 01/21/2023] Open
Abstract
Due to increasing rates of antimicrobial-resistant infections and the current inadequacy of the antibiotic pipeline, there is increasing interest in nontraditional approaches to antibacterial therapies. We define "traditional" agents as small-molecule agents that directly target bacterial components to exert a bacteriostatic or bactericidal effect, and "nontraditional approaches" as antimicrobial therapeutics that work through other means (ie, not a small molecule and/or utilizes a nontraditional target). Due to their atypical features, such therapies may be less susceptible to the emergence of resistance than traditional antibiotics. They include approaches such as monoclonal antibodies, virulence disruptors, immunomodulators, phage therapies, microbiome-based therapies, antibiotic potentiators, and antisense approaches. This article discusses both the developmental and regulatory advantages and challenges associated with each of these technologies. By identifying existing regulatory and developmental gaps, we hope to provide a sense of where focusing resources may provide the greatest impact on successful product development.
Collapse
Affiliation(s)
- Brian N Tse
- Division of Chemical, Biological, Radiological and Nuclear (CBRN) Medical Countermeasures, Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), US Department of Health and Human Services (US DHHS), Washington, District of Columbia
| | - Amesh A Adalja
- Center for Health Security, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.,Division of Infectious Disease, University of Pittsburgh School of Medicine, Pennsylvania
| | - Christopher Houchens
- Division of Chemical, Biological, Radiological and Nuclear (CBRN) Medical Countermeasures, Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), US Department of Health and Human Services (US DHHS), Washington, District of Columbia
| | - Joseph Larsen
- Division of Chemical, Biological, Radiological and Nuclear (CBRN) Medical Countermeasures, Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), US Department of Health and Human Services (US DHHS), Washington, District of Columbia
| | - Thomas V Inglesby
- Center for Health Security, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | |
Collapse
|
34
|
Synthesis and Biological Evaluation of Azamacrolide Comprising the Triazole Moiety as Quorum Sensing Inhibitors. Molecules 2018; 23:molecules23051086. [PMID: 29734673 PMCID: PMC6102594 DOI: 10.3390/molecules23051086] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 04/21/2018] [Accepted: 05/02/2018] [Indexed: 12/13/2022] Open
Abstract
Novel azamacrolides comprising the triazole moiety were synthesized and evaluated for their quorum sensing inhibitor activities on the Agrobacterium tumefaciens. It was found that the inhibition rate of compound Z12-3 at 200 mg/L (0.45 mM) can reach 67%. The potential binding modes between these molecules and the TraR QS receptor was performed by molecular docking. The results showed that the two nitrogen atoms in the triazole ring of Z12-3 formed hydrogen bonds with GLN-2, and the carbonyl group (C=O) in the amide formed hydrogen bonds with water. It was worth noting that the carbonyl group on the macrolides formed hydrogen bonds with the G-106 base in the DNA. These azamacrolides may block quorum sensing expression through key amino acid residues or DNA bases in the TraR QS receptor by hydrogen-bonded.
Collapse
|
35
|
Mion S, Rémy B, Plener L, Chabrière E, Daudé D. [Prevent bacteria from communicating: Divide to cure]. ANNALES PHARMACEUTIQUES FRANÇAISES 2018; 76:249-264. [PMID: 29598881 DOI: 10.1016/j.pharma.2018.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 02/07/2023]
Abstract
Quorum Sensing (QS) is a communication system used by numerous bacteria to synchronize their behavior according to the cell density. In this way, bacteria secrete and sense small mediating molecules, called autoinducers (AI), which concentration increases in the environment proportionally to bacterial cell number. QS induces major physiological and phenotypic changes such as virulence induction and biofilm formation. Biofilm represents a physical barrier which shelters bacteria poorly sensitive to antimicrobial treatments and favors the apparition of resistance mechanisms. Disturbing QS is referred to as quorum quenching (QQ). This strategy is used by microorganisms themselves to prevent the development of specific group behaviors. Two strategies are mainly employed: the use of quorum sensing inhibitors (QSI) and of quorum quenching enzymes (QQE) that degrades AI. Many studies have been dedicated to identifying QSI (natural or synthetic) as well as QQE and demonstrating their anti-virulence and anti-biofilm effects on numerous bacterial species. Synergistic effects between QQ and traditional treatments such as antibiotherapy or with reemerging phage therapy have been put forward. The efficiency of numerous QSI and QQE was thereby demonstrated either with in vitro or in vivo animal models leading to the development of medical devices containing QSI and QQE to improve already existing treatments.
Collapse
Affiliation(s)
- S Mion
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU-Méditerranée Infection, 19-21, boulevard Jean-Moulin, 13005 Marseille, France
| | - B Rémy
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU-Méditerranée Infection, 19-21, boulevard Jean-Moulin, 13005 Marseille, France; Gene&GreenTK, 19-21, boulevard Jean-Moulin, 13005 Marseille, France
| | - L Plener
- Gene&GreenTK, 19-21, boulevard Jean-Moulin, 13005 Marseille, France
| | - E Chabrière
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU-Méditerranée Infection, 19-21, boulevard Jean-Moulin, 13005 Marseille, France
| | - D Daudé
- Gene&GreenTK, 19-21, boulevard Jean-Moulin, 13005 Marseille, France.
| |
Collapse
|
36
|
Rémy B, Mion S, Plener L, Elias M, Chabrière E, Daudé D. Interference in Bacterial Quorum Sensing: A Biopharmaceutical Perspective. Front Pharmacol 2018; 9:203. [PMID: 29563876 PMCID: PMC5845960 DOI: 10.3389/fphar.2018.00203] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 02/22/2018] [Indexed: 12/12/2022] Open
Abstract
Numerous bacteria utilize molecular communication systems referred to as quorum sensing (QS) to synchronize the expression of certain genes regulating, among other aspects, the expression of virulence factors and the synthesis of biofilm. To achieve this process, bacteria use signaling molecules, known as autoinducers (AIs), as chemical messengers to share information. Naturally occurring strategies that interfere with bacterial signaling have been extensively studied in recent years, examining their potential to control bacteria. To interfere with QS, bacteria use quorum sensing inhibitors (QSIs) to block the action of AIs and quorum quenching (QQ) enzymes to degrade signaling molecules. Recent studies have shown that these strategies are promising routes to decrease bacterial pathogenicity and decrease biofilms, potentially enhancing bacterial susceptibility to antimicrobial agents including antibiotics and bacteriophages. The efficacy of QSIs and QQ enzymes has been demonstrated in various animal models and are now considered in the development of new medical devices against bacterial infections, including dressings, and catheters for enlarging the therapeutic arsenal against bacteria.
Collapse
Affiliation(s)
- Benjamin Rémy
- IRD, APHM, MEPHI, IHU-Méditerranée Infection, Aix Marseille Université, Marseille, France
- Gene&GreenTK, Marseille, France
| | - Sonia Mion
- IRD, APHM, MEPHI, IHU-Méditerranée Infection, Aix Marseille Université, Marseille, France
| | | | - Mikael Elias
- Department of Biochemistry, Molecular Biology and Biophysics, Biotechnology Institute, University of Minnesota, St. Paul, MN, United States
| | - Eric Chabrière
- IRD, APHM, MEPHI, IHU-Méditerranée Infection, Aix Marseille Université, Marseille, France
| | | |
Collapse
|
37
|
Pérez-Pérez M, Jorge P, Pérez Rodríguez G, Pereira MO, Lourenço A. Quorum sensing inhibition in Pseudomonas aeruginosa biofilms: new insights through network mining. BIOFOULING 2017; 33:128-142. [PMID: 28121162 DOI: 10.1080/08927014.2016.1272104] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/09/2016] [Indexed: 06/06/2023]
Abstract
Quorum sensing plays a pivotal role in Pseudomonas aeruginosa's virulence. This paper reviews experimental results on antimicrobial strategies based on quorum sensing inhibition and discusses current targets in the regulatory network that determines P. aeruginosa biofilm formation and virulence. A bioinformatics framework combining literature mining with information from biomedical ontologies and curated databases was used to create a knowledge network of potential anti-quorum sensing agents for P. aeruginosa. A total of 110 scientific articles, corresponding to 1,004 annotations, were so far included in the network and are analysed in this work. Information on the most studied agents, QS targets and methods is detailed. This knowledge network offers a unique view of existing strategies for quorum sensing inhibition and their main regulatory targets and may be used to readily access otherwise scattered information and to help generate new testable hypotheses. This knowledge network is publicly available at http://pcquorum.org/ .
Collapse
Affiliation(s)
- Martín Pérez-Pérez
- a ESEI - Escuela Superior de Ingeniería Informática , Universidad de Vigo , Ourense , Spain
| | - Paula Jorge
- b CEB - Centre of Biological Engineering, LIBRO - Laboratory of Research in Biofilms Rosário Oliveira , University of Minho , Braga , Portugal
| | - Gael Pérez Rodríguez
- a ESEI - Escuela Superior de Ingeniería Informática , Universidad de Vigo , Ourense , Spain
| | - Maria Olívia Pereira
- b CEB - Centre of Biological Engineering, LIBRO - Laboratory of Research in Biofilms Rosário Oliveira , University of Minho , Braga , Portugal
| | - Anália Lourenço
- a ESEI - Escuela Superior de Ingeniería Informática , Universidad de Vigo , Ourense , Spain
- c CEB - Centre of Biological Engineering , University of Minho , Braga , Portugal
| |
Collapse
|