1
|
Dadkhah F, Moniri F, Zamani H, Hadavi M. Anti-virulence effects of Diclofenac sodium in combination with gentamicin on Pseudomonas aeruginosa: attenuation of quorum sensing related traits and efflux pump systems. Braz J Microbiol 2025; 56:1069-1081. [PMID: 39964668 PMCID: PMC12095730 DOI: 10.1007/s42770-025-01631-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/29/2025] [Indexed: 05/22/2025] Open
Abstract
Antibiotic resistance is a major clinical and public health problem. Non-steroidal anti-inflammatory drugs (NSAIDs) could increase the susceptibility of bacteria to antibiotics and have shown broad antimicrobial activity. In this work, the effect of Diclofenac sodium alone and in combination with Gentamicin on the expression of efflux pump genes and some virulence traits in clinical isolates of P. aeruginosa was investigated. The checkerboard titration assay was used to evaluate the synergistic effect of Diclofenac sodium and Gentamicin. The relative expression of MexAB-OprM and MexXY-OprM efflux pump genes was determined using qPCR. The impact of drugs on the activity of the efflux pump and some virulence traits, including biofilm formation, swarming, swimming and twitching, and bacterial proteolytic and hemolytic activities were assessed. The minimum inhibitory concentration (MIC) of Diclofenac sodium and Gentamicin for clinical P. aeruginosa strains was 5120 and 128 µg/mL and the drugs showed synergic antibacterial activity. Diclofenac sodium reduced the expression of the mexB, mexX, and mexY genes, and increased the expression of the mexA and oprM genes. In addition, Diclofenac sodium alone and in combination with Gentamicin inhibited the activity of the bacterial efflux pump and the simultaneous treatment of P. aeruginosa with Diclofenac sodium and Gentamicin significantly reduced biofilm formation, bacterial motility, proteolytic (40.78%) and hemolytic (85%) activities compared with untreated group. This study addresses clinically relevant questions about the efficacy and potential synergistic effects of diclofenac sodium and gentamicin in treating P. aeruginosa infections that can be applicable to clinical practice.
Collapse
Affiliation(s)
| | - Fatemeh Moniri
- Department of Biology, University of Guilan, Rasht, Iran
| | | | - Mahvash Hadavi
- Department of Biology, University of Guilan, Rasht, Iran
| |
Collapse
|
2
|
Beasley JM, Dorjsuren D, Jain S, Rath M, Tieghi RS, Tropsha A, Simeonov A, Zakharov AV, Muratov E. Breaking the Phalanx: Overcoming Bacterial Drug Resistance with Quorum Sensing Inhibitors that Enhance Therapeutic Activity of Antibiotics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633658. [PMID: 39896648 PMCID: PMC11785035 DOI: 10.1101/2025.01.17.633658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Antibiotic-resistant bacterial infections loom over humanity as an increasing deadly threat. There exists a dire need for new treatments, especially those that synergize with our existing arsenal of antibiotic drugs to help overcome the gap in antibiotic efficacy and attenuate the development of new antibiotic-resistance in the most dangerous pathogens. Quorum sensing systems in bacteria drive the formation of biofilms, increase surface motility, and enhance other virulence factors, making these systems attractive targets for the discovery of novel antibacterials. Quorum sensing inhibitors (QSIs) are hypothesized to synergize with existing antibiotics, making bacteria more sensitive to the effects of these drugs. In this study, we aimed to find the synergistic combinations between the QSIs and known antibiotics to combat the two deadliest hospital infections - Pseudomonas aeruginosa and Acinetobacter baumannii. We mined biochemical activity databases and literature to identify known, high efficacy QSIs against these bacteria. We used these data to develop and validate a Quantitative Structure-Activity Relationship (QSAR) model for predicting QSI activity and then employed this model to identify new potential QSIs from the Inxight database of approved and investigational drugs. We then tested binary mixtures of the identified QSIs with 11 existing antibiotics using a combinatorial matrix screening approach with ten (five of each) clinical isolates of P. aeruginosa and A. baumannii. Amongst explored drug combinations, 31 exhibited a synergistic effect, including mixtures involving naldemedine and telotristat, two drugs predicted by our model with previously undescribed QSI activity. Although no mixture inhibiting all the strains was found, piperacillin combined with curcumin, ketoprofen, indomethacin, and piroxicam demonstrated the broadest antimicrobial action. We anticipate that further preclinical investigation of these combinations of novel repurposed QSIs with a known antibiotic may lead to novel clinical candidates.
Collapse
Affiliation(s)
- Jon-Michael Beasley
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Dorjbal Dorjsuren
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, 20850
| | - Sankalp Jain
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, 20850
| | - Marielle Rath
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | | | - Alexander Tropsha
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, 20850
| | - Alexey V. Zakharov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, 20850
| | - Eugene Muratov
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
3
|
Khorramdel M, Ghadikolaii FP, Hashemy SI, Javid H, Tabrizi MH. Nanoformulated meloxicam and rifampin: inhibiting quorum sensing and biofilm formation in Pseudomonas aeruginosa. Nanomedicine (Lond) 2024; 19:615-632. [PMID: 38348578 DOI: 10.2217/nnm-2023-0268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024] Open
Abstract
Background: We aimed to investigate the simultaneous effects of meloxicam and rifampin nanoformulations with solid lipid nanoparticle (SLN) and nanostructured lipid carrier (NLC) substrates on inhibiting the quorum-sensing system of Pseudomonas aeruginosa and preventing biofilm formation by this bacterium. Methods: Antimicrobial activity of rifampin and meloxicam encapsulated with SLNs and NLCs against P. aeruginosa PAO1 was assessed by disk diffusion, minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC). Results: The SLN formulation was associated with lower doses for the MIC and minimum bactericidal concentration in comparison to NLC. Moreover, our results demonstrated that both nanoformulations were able to produce 100% inhibition of the biofilm formation of P. aeruginosa PAO1. Conclusion: All these findings suggest that meloxicam and rifampin encapsulated with SLNs could be the most effective formulation against P. aeruginosa.
Collapse
Affiliation(s)
- Malihe Khorramdel
- Department of Microbiology, Ayatollah Amoli Branch, Islamic Azad University, Amol, Iran
| | | | - Seyed Isaac Hashemy
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Javid
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
4
|
Mirpour M, Zahmatkesh H. Ketoprofen attenuates Las/Rhl quorum-sensing (QS) systems of Pseudomonas aeruginosa: molecular and docking studies. Mol Biol Rep 2024; 51:133. [PMID: 38236445 DOI: 10.1007/s11033-023-09071-3] [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: 07/06/2023] [Accepted: 11/02/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Quorum sensing (QS) is the leading cause of persistent infections and recalcitrance to antibiotic treatment of Pseudomonas aeruginosa. Hence, QS inhibitors are promising agents for the potential treatment of P. aeruginosa infections. METHODS AND RESULTS Herein, the reducing effect of ketoprofen on virulence factors production including protease, hemolysin, pyocyanin, hydrogen cyanide, biofilm, and motility of P. aeruginosa strains was investigated. Furthermore, the quorum quenching activity of ketoprofen at the molecular level was examined by real-time PCR assessment. Our results showed that ketoprofen significantly attenuates virulence factors and biofilm formation in P. aeruginosa strains. Moreover, ketoprofen down-regulated the expression of lasI, lasR, rhlI, and rhlR genes, by 35-47, 22-48, 34-67, and 43-56%, respectively. As well, molecular docking simulation showed a high binding affinity of ketoprofen with QS regulatory proteins. CONCLUSIONS Consequently, this study confirmed the quorum quenching activity of ketoprofen, which could be employed as a useful agent for the treatment of P. aeruginosa infections.
Collapse
Affiliation(s)
- Mirsasan Mirpour
- Department of Microbiology, Lahijan Branch, Islamic Azad University, Lahijan, Iran.
| | - Hossein Zahmatkesh
- Department of Microbiology, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| |
Collapse
|
5
|
Pisoni LA, Semple SJ, Liu S, Sykes MJ, Venter H. Combined Structure- and Ligand-Based Approach for the Identification of Inhibitors of AcrAB-TolC in Escherichia coli. ACS Infect Dis 2023; 9:2504-2522. [PMID: 37888944 DOI: 10.1021/acsinfecdis.3c00350] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The inhibition of efflux pumps is a promising approach to combating multidrug-resistant bacteria. We have developed a combined structure- and ligand-based model, using OpenEye software, for the identification of inhibitors of AcrB, the inner membrane protein component of the AcrAB-TolC efflux pump in Escherichia coli. From a database of 1391 FDA-approved drugs, 23 compounds were selected to test for efflux inhibition in E. coli. Seven compounds, including ivacaftor (25), butenafine (19), naftifine (27), pimozide (30), thioridazine (35), trifluoperazine (37), and meloxicam (26), enhanced the activity of at least one antimicrobial substrate and inhibited the efflux pump-mediated removal of the substrate Nile Red from cells. Ivacaftor (25) inhibited efflux dose dependently, had no effect on an E. coli strain with genomic deletion of the gene encoding AcrB, and did not damage the bacterial outer membrane. In the presence of a sub-minimum inhibitory concentration (MIC) of the outer membrane permeabilizer colistin, ivacaftor at 1 μg/mL reduced the MICs of erythromycin and minocycline by 4- to 8-fold. The identification of seven potential AcrB inhibitors shows the merits of a combined structure- and ligand-based approach to virtual screening.
Collapse
Affiliation(s)
- Lily A Pisoni
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
- Quality Use of Medicines and Pharmacy Research Centre, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Susan J Semple
- Quality Use of Medicines and Pharmacy Research Centre, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Sida Liu
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Matthew J Sykes
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Henrietta Venter
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| |
Collapse
|
6
|
Esnaashari F, Rostamnejad D, Zahmatkesh H, Zamani H. In vitro and in silico assessment of anti-quorum sensing activity of Naproxen against Pseudomonas aeruginosa. World J Microbiol Biotechnol 2023; 39:244. [PMID: 37407806 DOI: 10.1007/s11274-023-03690-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Serious infections caused by Pseudomonas aeruginosa are usually related to quorum sensing (QS)-dependent virulence factors. Hence, QS inhibition is a promising approach to overcoming P. aeruginosa infections. This study aimed to investigate the effect of naproxen on biofilm formation and QS-related virulence traits of P. aeruginosa. Furthermore, the anti-QS potential of naproxen was evaluated using real-time PCR and molecular docking analysis. Our findings supported the anti-QS activity of naproxen, as evidenced by down-regulation of the lasI and rhlI genes expression as well as the attenuation of bacterial protease, hemolysin, pyocyanin, biofilm, and motility. Additionally, the high binding affinity of naproxen with QS regulatory proteins was determined in the molecular docking simulation. Altogether, these findings suggest that naproxen has a promising potential in inhibiting QS-associated traits of P. aeruginosa.
Collapse
Affiliation(s)
- Fatemeh Esnaashari
- Department of Biology, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Dorna Rostamnejad
- Department of Microbiology, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Hossein Zahmatkesh
- Department of Microbiology, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | | |
Collapse
|
7
|
Diclofenac and Meloxicam Exhibited Anti-Virulence Activities Targeting Staphyloxanthin Production in Methicillin-Resistant Staphylococcus aureus. Antibiotics (Basel) 2023; 12:antibiotics12020277. [PMID: 36830188 PMCID: PMC9951919 DOI: 10.3390/antibiotics12020277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/21/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a worldwide leading versatile pathogen that causes a wide range of serious infections. The emergence of antimicrobial resistance against S. aureus resulted in an urgent need to develop new antimicrobials in the new era. The methicillin-resistant S. aureus (MRSA) prevalence in hospital and community settings necessitates the discovery of novel anti-pathogenic agents. Staphyloxanthin (STX) is a key virulence factor for the survival of MRSA against host innate immunity. The current work aimed to demonstrate the anti-virulence properties of meloxicam (MXM) as compared to diclofenac (DC), which was previously reported to mitigate the virulence of multidrug-resistant Staphylococcus aureus and test their activities in STX production. A total of 80 S. aureus clinical isolates were included, wherein a qualitative and quantitative assessment of STX inhibition by diclofenac and meloxicam was performed. The quantitative gene expression of STX biosynthetic genes (crtM, crtN and sigB) and hla (coded for α-hemolysin) as a virulence gene with and without DC and MXM was conducted, followed by molecular docking analysis for further confirmation. DC and MXM potently inhibited the synthesis of STX at 47 and 59 µg/mL to reach 79.3-98% and 80.6-96.7% inhibition, respectively. Treated cells also revealed a significant downregulation of virulence genes responsible for STX synthesis, such as crtM, crtN and global transcriptional regulator sigB along with the hla gene. Furthermore, computational studies unveiled strong interactions between the CrtM binding site and DC/MXM. In conclusion, this study highlights the potential role and repurposing of DC and MXM as adjuvants to conventional antimicrobials and as an anti-virulent to combat MRSA infections.
Collapse
|
8
|
Rattanachak N, Weawsiangsang S, Daowtak K, Thongsri Y, Ross S, Ross G, Nilsri N, Baldock RA, Pongcharoen S, Jongjitvimol T, Jongjitwimol J. High-Throughput Transcriptomic Profiling Reveals the Inhibitory Effect of Hydroquinine on Virulence Factors in Pseudomonas aeruginosa. Antibiotics (Basel) 2022; 11:1436. [PMID: 36290094 PMCID: PMC9598861 DOI: 10.3390/antibiotics11101436] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/08/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Hydroquinine is an organic alkaloid compound that exhibits antimicrobial activity against several bacterial strains including strains of both drug-sensitive and multidrug-resistant P. aeruginosa. Despite this, the effects of hydroquinine on virulence factors in P. aeruginosa have not yet been characterized. We therefore aimed to uncover the mechanism of P. aeruginosa hydroquinine-sensitivity using high-throughput transcriptomic analysis. We further confirmed whether hydroquinine inhibits specific virulence factors using RT-qPCR and phenotypic analysis. At half the minimum inhibitory concentration (MIC) of hydroquinine (1.250 mg/mL), 254 genes were differentially expressed (97 downregulated and 157 upregulated). We found that flagellar-related genes were downregulated by between −2.93 and −2.18 Log2-fold change. These genes were consistent with the analysis of gene ontology and KEGG pathway. Further validation by RT-qPCR showed that hydroquinine significantly suppressed expression of the flagellar-related genes. By analyzing cellular phenotypes, P. aeruginosa treated with ½MIC of hydroquinine exhibited inhibition of motility (30−54% reduction) and pyocyanin production (~25−27% reduction) and impaired biofilm formation (~57−87% reduction). These findings suggest that hydroquinine possesses anti-virulence factors, through diminishing flagellar, pyocyanin and biofilm formation.
Collapse
Affiliation(s)
- Nontaporn Rattanachak
- Biomedical Sciences Program, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Sattaporn Weawsiangsang
- Biomedical Sciences Program, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Krai Daowtak
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Yordhathai Thongsri
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Sukunya Ross
- Department of Chemistry, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
- Centre of Excellence in Biomaterials, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Gareth Ross
- Department of Chemistry, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
- Centre of Excellence in Biomaterials, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Nungruthai Nilsri
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Robert A. Baldock
- School of Pharmacy and Biomedical Sciences, Faculty of Science and Health, University of Portsmouth, Portsmouth PO1 2DT, UK
| | - Sutatip Pongcharoen
- Division of Immunology, Department of Medicine, Faculty of Medicine, Naresuan University, Phitsanulok 65000, Thailand
| | - Touchkanin Jongjitvimol
- Biology Program, Faculty of Science and Technology, Pibulsongkram Rajabhat University, Phitsanulok 65000, Thailand
| | - Jirapas Jongjitwimol
- Biomedical Sciences Program, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Centre of Excellence in Biomaterials, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| |
Collapse
|
9
|
Qi L, Liang R, Duan J, Song S, Pan Y, Liu H, Zhu M, Li L. Synergistic antibacterial and anti-biofilm activities of resveratrol and polymyxin B against multidrug-resistant Pseudomonas aeruginosa. J Antibiot (Tokyo) 2022; 75:567-575. [PMID: 35999263 DOI: 10.1038/s41429-022-00555-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 11/10/2022]
Abstract
Bacterial infection caused by multidrug-resistant Pseudomonas aeruginosa has become a challenge in clinical practice. Polymyxins are used as the last resort agent for otherwise untreatable Gram-negative bacteria, including multidrug-resistant P.aeruginosa. However, pharmacodynamic (PD) and pharmacokinetic (PK) data on polymyxins suggest that polymyxin monotherapy is unlikely to generate reliably efficacious plasma concentrations. Also, polymyxin resistance has been frequently reported, especially among multidrug-resistant P.aeruginosa, which further limits its clinical use. A strategy for improving the antibacterial activity of polymyxins and preventing the development of polymyxin resistance is to use polymyxins in combination with other agents. In this study, we have demonstrated that resveratrol, a well tolerated compound, has synergistic effects when tested in vitro with polymyxin B on antibacterial and anti-biofilm activities. However, its' systemic use is limited as the required high plasma levels of resveratrol are not achievable. This suggests that it could be a partner for the combination therapy of polymyxin B in the treatment of topical bacterial infection caused by MDR P.aeruginosa.
Collapse
Affiliation(s)
- Lin Qi
- Department of Clinical Laboratory, Jinzhou Medical University Graduate Training Base, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, P. R. China
| | - Rongxin Liang
- Department of Clinical Laboratory, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, P. R. China
| | - Jingjing Duan
- Department of Clinical Laboratory, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, P. R. China
| | - Songze Song
- Jinzhou Medical University, Jinzhou, Liaoning, 121001, P. R. China
| | - Yunjun Pan
- Department of Clinical Laboratory, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, P. R. China
| | - Hui Liu
- Department of Clinical Laboratory, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, P. R. China
| | - Mingan Zhu
- Department of Clinical Laboratory, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, P. R. China
| | - Lian Li
- Department of Clinical Laboratory, Jinzhou Medical University Graduate Training Base, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, P. R. China. .,Department of Clinical Laboratory, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, P. R. China.
| |
Collapse
|
10
|
Sionov RV, Steinberg D. Targeting the Holy Triangle of Quorum Sensing, Biofilm Formation, and Antibiotic Resistance in Pathogenic Bacteria. Microorganisms 2022; 10:1239. [PMID: 35744757 PMCID: PMC9228545 DOI: 10.3390/microorganisms10061239] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic and recurrent bacterial infections are frequently associated with the formation of biofilms on biotic or abiotic materials that are composed of mono- or multi-species cultures of bacteria/fungi embedded in an extracellular matrix produced by the microorganisms. Biofilm formation is, among others, regulated by quorum sensing (QS) which is an interbacterial communication system usually composed of two-component systems (TCSs) of secreted autoinducer compounds that activate signal transduction pathways through interaction with their respective receptors. Embedded in the biofilms, the bacteria are protected from environmental stress stimuli, and they often show reduced responses to antibiotics, making it difficult to eradicate the bacterial infection. Besides reduced penetration of antibiotics through the intricate structure of the biofilms, the sessile biofilm-embedded bacteria show reduced metabolic activity making them intrinsically less sensitive to antibiotics. Moreover, they frequently express elevated levels of efflux pumps that extrude antibiotics, thereby reducing their intracellular levels. Some efflux pumps are involved in the secretion of QS compounds and biofilm-related materials, besides being important for removing toxic substances from the bacteria. Some efflux pump inhibitors (EPIs) have been shown to both prevent biofilm formation and sensitize the bacteria to antibiotics, suggesting a relationship between these processes. Additionally, QS inhibitors or quenchers may affect antibiotic susceptibility. Thus, targeting elements that regulate QS and biofilm formation might be a promising approach to combat antibiotic-resistant biofilm-related bacterial infections.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Biofilm Research Laboratory, The Institute of Biomedical and Oral Research, The Faculty of Dental Medicine, Hadassah Medical School, The Hebrew University, Jerusalem 9112102, Israel;
| | | |
Collapse
|
11
|
Khodaparast S, Ghanbari F, Zamani H. Evaluation of the effect of ibuprofen in combination with ciprofloxacin on the virulence-associated traits, and efflux pump genes of Pseudomonas aeruginosa. World J Microbiol Biotechnol 2022; 38:125. [PMID: 35657493 DOI: 10.1007/s11274-022-03316-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/20/2022] [Indexed: 12/01/2022]
Abstract
Biofilm formation and antibiotic efflux are two determinant factors in the development of drug resistance phenotype by Pseudomonas aeruginosa. Non-steroid anti-inflammatory drugs have shown the antimicrobial potential to be used in combination with antibiotics against bacterial pathogens. In this work, the effect of ibuprofen alone and in combination with ciprofloxacin on some virulence traits and the expression of the alginate synthesis and efflux pump genes of clinical isolates of P. aeruginosa was investigated. The checkerboard titration assay was used to evaluate the synergism of the drugs. P. aeruginosa strains were grown in the presence of sub-inhibitory concentrations of the drug and their biofilm formation level, swarming, swimming, and hemolytic activity were assessed. Also, the relative expression of the alg44, algT/U, mexB, and oprM genes was determined by qPCR assay. The MIC of ibuprofen and ciprofloxacin were measured 2048 and 32 µg/mL and the drugs showed synergic antibacterial activity (FIC = 0.4). Moreover, ibuprofen alone and in combination with ciprofloxacin, significantly reduced the expression of alg44 (0.22 and 0.25 folds) and algT/U (0.26 and 0.37 folds) genes, while increased the expression of the mexB (1.64 and 1.83 folds) and oprM (1.36 and 1.92 folds) genes. Simultaneous treatment of bacterial cells with ibuprofen and ciprofloxacin significantly decreased bacterial biofilm formation (65%), swimming, swarming, and hemolytic activity (85%), compared with the control. This work suggests that ibuprofen has considerable anti-virulence potential against P. aeruginosa and could be employed for combination therapy with antibiotics after further characterizations.
Collapse
Affiliation(s)
- Samira Khodaparast
- Depertment of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Fatemeh Ghanbari
- Department of Biology, University of Guilan, University Campus 2, Rasht, Iran
| | - Hojjatolah Zamani
- Depertment of Biology, Faculty of Science, University of Guilan, Rasht, Iran.
| |
Collapse
|
12
|
Jampilek J. Drug repurposing to overcome microbial resistance. Drug Discov Today 2022; 27:2028-2041. [PMID: 35561965 DOI: 10.1016/j.drudis.2022.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 12/15/2022]
Abstract
Infections are a growing global threat, and the number of resistant species of microbial pathogens is alarming. However, the rapid development of cross-resistant or multidrug-resistant strains and the development of so-called 'superbugs' are in stark contrast to the number of newly launched anti-infectives on the market. In this review, I summarize the causes of antimicrobial resistance, briefly discuss different approaches to the discovery and development of new anti-infective drugs, and focus on drug repurposing strategy, which is discussed from all possible perspectives. A comprehensive overview of drugs of other indications tested for their in vitro antimicrobial activity to support existing anti-infective therapeutics is provided, including several critical remarks on this strategy of repurposing non-antibiotics to antibacterial drugs.
Collapse
Affiliation(s)
- Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia; Department of Chemical Biology, Faculty of Science, Palacky University, Slechtitelu 27, 783 71 Olomouc, Czech Republic.
| |
Collapse
|
13
|
Di Bonaventura G, Lupetti V, De Fabritiis S, Piccirilli A, Porreca A, Di Nicola M, Pompilio A. Giving Drugs a Second Chance: Antibacterial and Antibiofilm Effects of Ciclopirox and Ribavirin against Cystic Fibrosis Pseudomonas aeruginosa Strains. Int J Mol Sci 2022; 23:ijms23095029. [PMID: 35563420 PMCID: PMC9102761 DOI: 10.3390/ijms23095029] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Drug repurposing is an attractive strategy for developing new antibacterial molecules. Herein, we evaluated the in vitro antibacterial, antibiofilm, and antivirulence activities of eight FDA-approved “non-antibiotic” drugs, comparatively to tobramycin, against selected Pseudomonas aeruginosa strains from cystic fibrosis patients. MIC and MBC values were measured by broth microdilution method. Time–kill kinetics was studied by the macro dilution method, and synergy studies were performed by checkerboard microdilution assay. The activity against preformed biofilms was measured by crystal violet and viable cell count assays. The effects on gene expression were studied by real-time quantitative PCR, while the cytotoxic potential was evaluated against IB3-1 bronchial CF cells. Ciclopirox, 5-fluorouracil, and actinomycin D showed the best activity against P. aeruginosa planktonic cells and therefore underwent further evaluation. Time–kill assays indicated actinomycin D and ciclopirox, contrarily to 5-fluorouracil and tobramycin, have the potential for bacterial eradication, although with strain-dependent efficacy. Ciclopirox was the most effective against the viability of the preformed biofilm. A similar activity was observed for other drugs, although they stimulate extracellular polymeric substance production. Ribavirin showed a specific antibiofilm effect, not dependent on bacterial killing. Exposure to drugs and tobramycin generally caused hyperexpression of the virulence traits tested, except for actinomycin D, which downregulated the expression of alkaline protease and alginate polymerization. Ciclopirox and actinomycin D revealed high cytotoxic potential. Ciclopirox and ribavirin might provide chemical scaffolds for anti-P. aeruginosa drugs. Further studies are warranted to decrease ciclopirox cytotoxicity and evaluate the in vivo protective effects.
Collapse
Affiliation(s)
- Giovanni Di Bonaventura
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (V.L.); (A.P.); (M.D.N.); (A.P.)
- Center of Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Correspondence:
| | - Veronica Lupetti
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (V.L.); (A.P.); (M.D.N.); (A.P.)
- Center of Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Simone De Fabritiis
- Center of Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Department of Medicine and Aging Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Alessandra Piccirilli
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Annamaria Porreca
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (V.L.); (A.P.); (M.D.N.); (A.P.)
| | - Marta Di Nicola
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (V.L.); (A.P.); (M.D.N.); (A.P.)
| | - Arianna Pompilio
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (V.L.); (A.P.); (M.D.N.); (A.P.)
- Center of Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
| |
Collapse
|
14
|
Benny AT, Rathinam P, Dev S, Mathew B, Radhakrishnan EK. Perillaldehyde mitigates virulence factors and biofilm formation of Pseudomonas aeruginosa clinical isolates, by acting on the quorum sensing mechanism in vitro. J Appl Microbiol 2022; 133:385-399. [PMID: 35384183 DOI: 10.1111/jam.15565] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/17/2021] [Accepted: 03/07/2022] [Indexed: 12/01/2022]
Abstract
AIM The incidence of biofilm linked catheter-associated urinary tract infections (CAUTIs) are increasing worldwide and Pseudomonas aeruginosa is one of the major causes. Perillaldehyde (PLD): as a natural, widely used flavouring agent, has been reported to possess various pharmacological properties. We hypothesized that PLD can inhibit biofilm formation and virulence factor production by P. aeruginosa by hampering the quorum sensing (QS) system(s). METHODS AND RESULTS Minimum inhibitory concentration (MIC) of PLD was assessed for standard strain and two multi-drug resistant catheter isolates of P. aeruginosa utilizing the microdilution method. Microtiter plate assay, crystal violet staining and scanning electron microscopy were used to evaluate the biofilm inhibition property. CFU was utilized to assess the antifouling property of PLD. Detection of virulence factors (VFs) and expression analysis of virulence determinants were applied to investigate the anti-virulence activity. Gene expression and molecular docking studies were also executed to explore the QS inhibition and binding of PLD with QS receptors. In the present study, PLD has significantly inhibited biofilm formation and antivirulence activity at sub-MIC levels (2.5 mM and 3.5 mM) in all the tested strains. In addition, molecular docking studies revealed a significant affinity towards quorum sensing receptors. DISCUSSIONS Perillaldehyde (PLD), being a non-toxic food flavouring agent, significantly inhibited biofilm formation, and exhibited antifouling property. PLD exhibited significantly reduced levels of VFs (p<0.001) and their respective genetic determinants (p<0.001). Gene expression analysis and molecular docking studies confirmed the interactions of PLD to the QS receptors, indicating the plausible mechanism for the anti-virulence property. SIGNIFICANCE AND IMPACT OF STUDY This study identified the anti-virulence potential of PLD and provided mechanistic insights. Perillaldehyde can be a suitable, non-toxic candidate for countering biofilms and associated pathogens, contributing to the prevention of biofilm-associated nosocomial infections..
Collapse
Affiliation(s)
- Anjitha Theres Benny
- Department of Chemistry, School of Advanced Science, VIT, Vellore, Tamil Nadu-632014, India
| | - Prasanth Rathinam
- Medical Biotechnology Laboratory, Pushpagiri Research Centre, Pushpagiri Institute of Medical Sciences and Research Centre, , Thiruvalla, Kerala - 689101, India
| | - Sanal Dev
- Department of Pharmaceutical Chemistry, Al-Shifa College of Pharmacy, Perinthalmanna, Kerala, India
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| | | |
Collapse
|
15
|
Chen YC, Moseson DE, Richard CA, Swinney MR, Horava SD, Oucherif KA, Cox AL, Hawkins ED, Li Y, DeNeve DF, Lomeo J, Zhu A, Lyle LT, Munson EJ, Taylor LS, Park K, Yeo Y. Development of hot-melt extruded drug/polymer matrices for sustained delivery of meloxicam. J Control Release 2022; 342:189-200. [PMID: 34990702 DOI: 10.1016/j.jconrel.2021.12.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/02/2021] [Accepted: 12/29/2021] [Indexed: 10/19/2022]
Abstract
For effective resolution of regional subacute inflammation and prevention of biofouling formation, we have developed a polymeric implant that can release meloxicam, a selective cyclooxygenase (COX)-2 inhibitor, in a sustained manner. Meloxicam-loaded polymer matrices were produced by hot-melt extrusion, with commercially available biocompatible polymers, poly(ε-caprolactone) (PCL), poly(lactide-co-glycolide) (PLGA), and poly(ethylene vinyl acetate) (EVA). PLGA and EVA had a limited control over the drug release rate partly due to the acidic microenvironment and hydrophobicity, respectively. PCL allowed for sustained release of meloxicam over two weeks and was used as a carrier of meloxicam. Solid-state and image analyses indicated that the PCL matrices encapsulated meloxicam in crystalline clusters, which dissolved in aqueous medium and generated pores for subsequent drug release. The subcutaneously implanted meloxicam-loaded PCL matrices in rats showed pharmacokinetic profiles consistent with their in vitro release kinetics, where higher drug loading led to faster drug release. This study finds that the choice of polymer platform is crucial to continuous release of meloxicam and the drug release rate can be controlled by the amount of drug loaded in the polymer matrices.
Collapse
Affiliation(s)
- Yun-Chu Chen
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Dana E Moseson
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Coralie A Richard
- Eli Lilly and Company, 893 Delaware Street, Indianapolis, IN 46225, USA
| | - Monica R Swinney
- Eli Lilly and Company, 450 Kendall Street, Cambridge, MA 02142, USA
| | - Sarena D Horava
- Eli Lilly and Company, 450 Kendall Street, Cambridge, MA 02142, USA
| | | | - Amy L Cox
- Eli Lilly and Company, 893 Delaware Street, Indianapolis, IN 46225, USA
| | - Eric D Hawkins
- Eli Lilly and Company, 893 Delaware Street, Indianapolis, IN 46225, USA
| | - Yongzhe Li
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Daniel F DeNeve
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Joshua Lomeo
- DigiM Solution LLC, 67 South Bedford Street, West Burlington, MA 01803, USA
| | - Aiden Zhu
- DigiM Solution LLC, 67 South Bedford Street, West Burlington, MA 01803, USA
| | - L Tiffany Lyle
- Department of Comparative Pathobiology, Purdue University, 625 Harrison Street, West Lafayette, IN 47907, USA
| | - Eric J Munson
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Kinam Park
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
16
|
Singh S, Bhatia S. Quorum Sensing Inhibitors: Curbing Pathogenic Infections through Inhibition of Bacterial Communication. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:486-514. [PMID: 34567177 PMCID: PMC8457738 DOI: 10.22037/ijpr.2020.113470.14318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Currently, most of the developed and developing countries are facing the problem of infectious diseases. The genius way of an exaggerated application of antibiotics led the infectious agents to respond by bringing a regime of persisters to resist antibiotics attacks prolonging their survival. Persisters have the dexterity to communicate among themself using signal molecules via the process of Quorum Sensing (QS), which regulates virulence gene expression and biofilms formation, making them more vulnerable to antibiotic attack. Our review aims at the different approaches applied in the ordeal to solve the riddle for QS inhibitors. QS inhibitors, their origin, structures and key interactions for QS inhibitory activity have been summarized. Solicitation of a potent QS inhibitor molecule would be beneficial, giving new life to the simplest antibiotics in adjuvant therapy.
Collapse
Affiliation(s)
- Shaminder Singh
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3 Milestone, Faridabad-Gurugram Expressway, Faridabad - 121 001, Haryana, India
| | - Sonam Bhatia
- Department of Pharmaceutical Science, SHALOM Institute of Health and Allied Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences (SHUATS), Naini-211007, Prayagraj, Uttar Pradesh, India
| |
Collapse
|
17
|
Tajani AS, Jangi E, Davodi M, Golmakaniyoon S, Ghodsi R, Soheili V, Fazly Bazzaz BS. Anti-quorum sensing potential of ketoprofen and its derivatives against Pseudomonas aeruginosa: insights to in silico and in vitro studies. Arch Microbiol 2021; 203:5123-5132. [PMID: 34319419 DOI: 10.1007/s00203-021-02499-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/04/2021] [Accepted: 07/19/2021] [Indexed: 11/25/2022]
Abstract
Antibiotics are usually used for the treatment of bacterial infections, but multidrug-resistant strains are a phenomenon that has been growing at an increasing rate worldwide. Thus, there is an increasing need for novel strategies for combatting infectious diseases. Many pathogenic bacteria apply quorum sensing (QS) to regulate their pathogenicity and virulence factors production. This circuit makes the QS system an attractive target for antibacterial therapy. In the present study, an important member of non-steroidal anti-inflammatory drugs (NSAIDs), by reducing the biofilm and producing QS-regulated virulence factors, ketoprofen and its synthetic derivatives were screened against the Pseudomonas aeruginosa PAO1. All compounds showed anti-biofilm activity (16-79%) and most of them presented anti-virulence activity. In the co-treatment of ketoprofen, G20, G21, or G77 with tobramycin, biofilm is significantly reduced (potentiated to > 50%) in the number of cells protected inside the impermeable matrix. The in silico studies in addition to the similarities between the chemical structures of PqsR natural ligands and ketoprofen derivatives reinforce the possibility that the mechanism of action is through PqsR inhibition. Based on the results, the anti-pathogenic effect was more appreciable in ketoprofen, G77, and G20.
Collapse
Affiliation(s)
- Amineh Sadat Tajani
- Department of Pharmaceutical Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Jangi
- Department of Pharmaceutical Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Davodi
- Department of Pharmaceutical Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sima Golmakaniyoon
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Razieh Ghodsi
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Soheili
- Department of Pharmaceutical Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Bibi Sedigheh Fazly Bazzaz
- Department of Pharmaceutical Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran. .,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
18
|
Paes Leme RC, da Silva RB. Antimicrobial Activity of Non-steroidal Anti-inflammatory Drugs on Biofilm: Current Evidence and Potential for Drug Repurposing. Front Microbiol 2021; 12:707629. [PMID: 34385992 PMCID: PMC8353384 DOI: 10.3389/fmicb.2021.707629] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022] Open
Abstract
It has been demonstrated that some non-steroidal anti-inflammatory drugs (NSAIDs), like acetylsalicylic acid, diclofenac, and ibuprofen, have anti-biofilm activity in concentrations found in human pharmacokinetic studies, which could fuel an interest in repurposing these well tolerated drugs as adjunctive therapies for biofilm-related infections. Here we sought to review the currently available data on the anti-biofilm activity of NSAIDs and its relevance in a clinical context. We performed a systematic literature review to identify the most commonly tested NSAIDs drugs in the last 5 years, the bacterial species that have demonstrated to be responsive to their actions, and the emergence of resistance to these molecules. We found that most studies investigating NSAIDs' activity against biofilms were in vitro, and frequently tested non-clinical bacterial isolates, which may not adequately represent the bacterial populations that cause clinically-relevant biofilm-related infections. Furthermore, studies concerning NSAIDs and antibiotic resistance are scarce, with divergent outcomes. Although the potential to use NSAIDs to control biofilm-related infections seems to be an exciting avenue, there is a paucity of studies that tested these drugs using appropriate in vivo models of biofilm infections or in controlled human clinical trials to support their repurposing as anti-biofilm agents.
Collapse
Affiliation(s)
- Rodrigo Cuiabano Paes Leme
- Laboratório Especial de Microbiologia Clínica (LEMC), Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil.,Department of Infectious Diseases, Centro Universitário de Volta Redonda, Volta Redonda, Brazil
| | | |
Collapse
|
19
|
Rathinam P, Murari BM, Viswanathan P. Biofilm inhibition and antifouling evaluation of sol-gel coated silicone implants with prolonged release of eugenol against Pseudomonas aeruginosa. BIOFOULING 2021; 37:521-537. [PMID: 34139901 DOI: 10.1080/08927014.2021.1933960] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/16/2021] [Accepted: 05/17/2021] [Indexed: 02/08/2023]
Abstract
The incidence of biofilm-linked catheter-associated urinary tract infections (CAUTIs) is increasing across the world. However, there is no clinical evidence to support the modifications of biomaterials, such as antimicrobial agent-coated catheters, that are known to reduce the risk of bacterial colonization and resistance development. The present study developed and tested silicone segments coated with an antivirulence agent, eugenol. The parameters for sol-gel preparation and coating were tailored to achieve a prolonged release of eugenol (for >35 days) at predefined antivirulence doses from dip-coated thin films. The eugenol-coated segments could prevent biofilm formation by Pseudomonas aeruginosa PAO1 as well as bacterial adhesion. Significant repression in the expression of virulence and biofilm-associated genes were recorded, confirming the antivirulence and biofilm inhibition properties of silicone segments coated with eugenol. The drug release profiles, efficacy analysis, neutrophil-response studies, and in vitro toxicity profiling further supported the contention that the activity of the eugenol-coated sections was effective and safe.
Collapse
Affiliation(s)
- Prasanth Rathinam
- Medical Biotechnology Laboratory, Pushpagiri Research Centre, Pushpagiri Institute of Medical Sciences and Research Centre, Thiruvalla, Kerala, India
- Renal Research Lab, Centre for Bio-Medical Research, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Bhasker Mohan Murari
- Sol-Gel-Biosensor Lab, Centre for Bio-Medical Research, School of Electronics Engineering, Vellore Institute of Technology, Vellore, India
| | - Pragasam Viswanathan
- Renal Research Lab, Centre for Bio-Medical Research, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
20
|
Aljalamdeh R, Price R, Jones MD, Bolhuis A. The effect of particle size of inhaled tobramycin dry powder on the eradication of Pseudomonas aeruginosa biofilms. Eur J Pharm Sci 2021; 158:105680. [PMID: 33346008 DOI: 10.1016/j.ejps.2020.105680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/26/2020] [Accepted: 12/14/2020] [Indexed: 10/22/2022]
Abstract
Pseudomonas aeruginosa is the predominant opportunistic bacterium that causes chronic respiratory infections in cystic fibrosis (CF) patients. This bacterium can form biofilms, which are structured communities of cells encased within a self-produced matrix. Such biofilms have a high level of resistance to multiple classes of antibiotics. A widely used treatment of P. aeruginosa lung infections in CF patients is tobramycin dry powder inhalation. The behaviour of particles in the lung has been well studied, and dry powder inhalers are optimised for optimal dispersion of the drug into different zones of the lung. However, one question that has not been addressed is whether the size of an antibiotic particle influences the antibiofilm activity against P. aeruginosa. We investigated this by fractionating tobramycin particles using a Next Generation Impactor (NGI). The fractions obtained were then tested in an in vitro model on P. aeruginosa biofilms. The results indicate that the antibiofilm activity of tobramycin dry powder inhaler can indeed be influenced by the particle size. Against P. aeruginosa biofilms of two clinical isolates, smaller tobramycin particles (aerodynamic diameter <2.82 µm) showed better efficacy by approximately 20% as compared to larger tobramycin particles (aerodynamic diameter <11.7 µm) However, this effect was only observed when biofilms were treated for 3 hours, whereas there was no difference after treatment for 24 hours. This suggests that in our model the rate of dissolution of larger particles limits the effectiveness of tobramycin over a 3-hour time period, which is relevant as this is equivalent to the time in which most tobramycin is cleared from the lung.
Collapse
Affiliation(s)
- Reham Aljalamdeh
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom
| | - Robert Price
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom
| | - Matthew D Jones
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom
| | - Albert Bolhuis
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom.
| |
Collapse
|
21
|
Khan F, Pham DTN, Oloketuyi SF, Kim YM. Antibiotics Application Strategies to Control Biofilm Formation in Pathogenic Bacteria. Curr Pharm Biotechnol 2020; 21:270-286. [PMID: 31721708 DOI: 10.2174/1389201020666191112155905] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/09/2019] [Accepted: 10/31/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND The establishment of a biofilm by most pathogenic bacteria has been known as one of the resistance mechanisms against antibiotics. A biofilm is a structural component where the bacterial community adheres to the biotic or abiotic surfaces by the help of Extracellular Polymeric Substances (EPS) produced by bacterial cells. The biofilm matrix possesses the ability to resist several adverse environmental factors, including the effect of antibiotics. Therefore, the resistance of bacterial biofilm-forming cells could be increased up to 1000 times than the planktonic cells, hence requiring a significantly high concentration of antibiotics for treatment. METHODS Up to the present, several methodologies employing antibiotics as an anti-biofilm, antivirulence or quorum quenching agent have been developed for biofilm inhibition and eradication of a pre-formed mature biofilm. RESULTS Among the anti-biofilm strategies being tested, the sub-minimal inhibitory concentration of several antibiotics either alone or in combination has been shown to inhibit biofilm formation and down-regulate the production of virulence factors. The combinatorial strategies include (1) combination of multiple antibiotics, (2) combination of antibiotics with non-antibiotic agents and (3) loading of antibiotics onto a carrier. CONCLUSION The present review paper describes the role of several antibiotics as biofilm inhibitors and also the alternative strategies adopted for applications in eradicating and inhibiting the formation of biofilm by pathogenic bacteria.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Marine-Integrated Bionics Research Center, Pukyong National University, Busan 48513, Korea.,Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201306, U.P., India
| | - Dung T N Pham
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Korea
| | - Sandra F Oloketuyi
- Laboratory for Environmental and Life Sciences, University of Nova Gorica 5000, Nova Gorica, Slovenia
| | - Young-Mog Kim
- Marine-Integrated Bionics Research Center, Pukyong National University, Busan 48513, Korea.,Department of Food Science and Technology, Pukyong National University, Busan 48513, Korea
| |
Collapse
|
22
|
Chen L, Zou Y, Kronfl AA, Wu Y. Type VI secretion system of Pseudomonas aeruginosa is associated with biofilm formation but not environmental adaptation. Microbiologyopen 2020; 9:e991. [PMID: 31961499 PMCID: PMC7066461 DOI: 10.1002/mbo3.991] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/13/2022] Open
Abstract
Pseudomonas aeruginosa encodes three type VI secretion systems (T6SSs), namely H1‐, H2‐, and H3‐T6SS. P. aeruginosa hemolysin‐coregulated protein (Hcp) is the effector protein and the hallmark of T6SS. Although T6SS is ubiquitous and affects ecology and human health, its general mechanism and physiological role are still not fully understood. Therefore, in this study, we investigated the impact of the P. aeruginosa T6SS on biofilm formation and environmental adaptation. To this end, we collected P. aeruginosa clinical isolates, divided them into strong biofilm formation (SBF) and nonbiofilm formation (NBF) groups based on their biofilm‐forming ability, and compared their associated clinical characteristics. The duration of hospitalization was longer in patients infected with SBF than those infected with NBF strains. The expression levels of T6SS‐related genes (hcp1 and hcp3) and a quorum‐sensing gene (lasR) were higher in the SBF group as compared to those in the NBF group. In addition, the expression level of lasR was negatively associated with that of hcp1, but was positively associated with those of hcp2 and hcp3. Moreover, we evaluated the expression of T6SS‐ and biofilm‐associated genes in planktonic and biofilm cells of the P. aeruginosa strain PAO1, and constructed strain PAO1△clpV1 to study the adaptation characteristics of H1‐T6SS. The expression levels of hcp1, hcp2, hcp3, lasR, and other biofilm‐associated genes were significantly higher in PAO1 biofilm cells as compared to those of planktonic cells. However, except for swarming ability as a vital feature for biofilm formation, there were no significant differences in the biofilm‐forming ability and expression of biofilm‐associated genes, adherence ability, growth characteristics, resistance to acid and osmotic pressure, surface structure, and morphology between the PAO1△clpV1 and PAO1 wild‐type strains. Collectively, our results suggest that T6SS might play a role in biofilm formation and that H1‐T6SS does not contribute to environmental adaptation in P. aeruginosa.
Collapse
Affiliation(s)
- Lihua Chen
- Department of Clinical Laboratory, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yaru Zou
- Department of Clinical Laboratory, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Asmaa Abbas Kronfl
- Department of Clinical Laboratory, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yong Wu
- Department of Clinical Laboratory, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
23
|
She P, Wang Y, Liu Y, Tan F, Chen L, Luo Z, Wu Y. Effects of exogenous glucose on Pseudomonas aeruginosa biofilm formation and antibiotic resistance. Microbiologyopen 2019; 8:e933. [PMID: 31532581 PMCID: PMC6925152 DOI: 10.1002/mbo3.933] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/13/2019] [Accepted: 08/16/2019] [Indexed: 11/29/2022] Open
Abstract
Pseudomonas aeruginosa is commonly found in nosocomial and life‐threatening infections in patients. Biofilms formed by P. aeruginosa exhibit much greater resistance to antibiotics than the planktonic form of the bacteria. Few groups have studied the effects of glucose, a major carbon source, and metabolite, on P. aeruginosa biofilm formation and on its metabolic pathways. In this study, we investigated the effect of glucose on the biofilm formation ability of P. aeruginosa and carried out a metabolomic analysis to identify whether glucose alters the metabolic activity of P. aeruginosa in biofilms. We found that glucose efficiently promoted P. aeruginosa biofilm formation by upregulating the expression of the extracellular polysaccharide‐related gene pslA. Treatment with glucose caused an increase in 7 metabolites (including 3‐hydroxypropionic acid, glucose‐6‐phosphate, and 2,3‐dimethylsuccinic acid) and a decrease in 18 metabolites (including myo‐inositol, glutamine, and methoxamedrine) in the biofilm. In addition, there was a synergistic effect between glucose and horse serum on biofilm formation when the two were added in combination, which also increased the resistance of biofilm to levofloxacin therapy. Thus, our work sheds light on the underlying mechanisms by which glucose may enhance biofilm formation and identifies novel targets for developing strategies to counteract biofilm formation.
Collapse
Affiliation(s)
- Pengfei She
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yanle Wang
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yiqing Liu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Fang Tan
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Lihua Chen
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhen Luo
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yong Wu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
24
|
Rajkumari J, Borkotoky S, Reddy D, Mohanty SK, Kumavath R, Murali A, Suchiang K, Busi S. Anti-quorum sensing and anti-biofilm activity of 5-hydroxymethylfurfural against Pseudomonas aeruginosa PAO1: Insights from in vitro, in vivo and in silico studies. Microbiol Res 2019; 226:19-26. [DOI: 10.1016/j.micres.2019.05.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 05/03/2019] [Accepted: 05/04/2019] [Indexed: 10/26/2022]
|
25
|
She P, Wang Y, Luo Z, Chen L, Tan R, Wang Y, Wu Y. Meloxicam inhibits biofilm formation and enhances antimicrobial agents efficacy by Pseudomonas aeruginosa. Microbiologyopen 2017; 7. [PMID: 29178590 PMCID: PMC5822345 DOI: 10.1002/mbo3.545] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/18/2017] [Accepted: 08/28/2017] [Indexed: 12/13/2022] Open
Abstract
Microbial biofilms are communities of surface‐adhered cells enclosed in a matrix of extracellular polymeric substances. Bacterial cells in biofilm are 10~1,000‐fold more resistant to antimicrobials than the planktonic cells. Burgeoning antibiotic resistance in Pseudomonas aeruginosa biofilm has necessitated the development of antimicrobial agents. Here, we have investigated the antibiofilm effect of meloxicam against P. aeruginosaPAO1 and its potential mechanisms. Further, we have explored whether meloxicam could enhance the susceptibility of bacterial biofilms to treatment with conventional antimicrobials. Here, we found that meloxicam could significantly inhibit PAO1 biofilm formation in a dose‐dependent manner at the concentration without influence on planktonic cell growth. Meloxicam could also significantly inhibit the motilities, production of extracellular matrix, and expression of quorum sensing‐related genes and virulence factors of PAO1. Furthermore, synergistic interaction was observed when meloxicam combined with tetracycline, gentamicin, tobramycin, ciprofloxacin, ceftriaxone, ofloxacin, norfloxacin, ceftazidime, and DNase at subminimal inhibitory concentrations against PAO1 bioiflm. Collectively, our study lays the foundation for further investigation of repurposing meloxicam as a topical antibiofilm agent to treat P. aeruginosa biofilm‐related infections.
Collapse
Affiliation(s)
- Pengfei She
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yangxia Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhen Luo
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Lihua Chen
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Ruichen Tan
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yanle Wang
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yong Wu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|