1
|
Lu H, Wang Y, Liu H, Wang N, Zhang Y, Li X. Review of the Presence and Phage-Mediated Transfer of ARGs in Biofilms. Microorganisms 2025; 13:997. [PMID: 40431170 PMCID: PMC12114417 DOI: 10.3390/microorganisms13050997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/02/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
The widespread use of antibiotics has led to the emergence of a large number of drug-resistant bacteria, accelerating the dissemination and spread of antibiotic resistance genes (ARGs) in the environment. Bacterial biofilms, serving as reservoirs of ARGs, pose potential risks to environmental health that should not be ignored. Studies on the presence and transfer of ARGs in biofilms have been conducted both domestically and internationally. This article summarises the research progress on ARGs in various environments and analyses the mechanisms and factors influencing the dissemination and transfer of ARGs in microplastics, activated sludge, and pipe wall biofilms, with a particular focus on phage-mediated ARG transfer. We also discuss current research gaps in this field to provide references for future biofilm management and health risk control of ARGs.
Collapse
Affiliation(s)
- Han Lu
- National Key Laboratory of Efficient Utilization of Nutrient Resources, Shandong Academy of Agricultural Sciences, Jinan 250100, China; (H.L.)
- National Technological Innovation Center for Comprehensive Utilization of Saline-Alkali Land, Dongying 257347, China
- Shandong Provincial Key Laboratory of Water Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Shandong University, Qingdao 266237, China
| | - Yanjun Wang
- National Key Laboratory of Efficient Utilization of Nutrient Resources, Shandong Academy of Agricultural Sciences, Jinan 250100, China; (H.L.)
- National Technological Innovation Center for Comprehensive Utilization of Saline-Alkali Land, Dongying 257347, China
| | - Hongyuan Liu
- National Key Laboratory of Efficient Utilization of Nutrient Resources, Shandong Academy of Agricultural Sciences, Jinan 250100, China; (H.L.)
- National Technological Innovation Center for Comprehensive Utilization of Saline-Alkali Land, Dongying 257347, China
| | - Nana Wang
- National Key Laboratory of Efficient Utilization of Nutrient Resources, Shandong Academy of Agricultural Sciences, Jinan 250100, China; (H.L.)
- National Technological Innovation Center for Comprehensive Utilization of Saline-Alkali Land, Dongying 257347, China
| | - Yan Zhang
- National Key Laboratory of Efficient Utilization of Nutrient Resources, Shandong Academy of Agricultural Sciences, Jinan 250100, China; (H.L.)
- National Technological Innovation Center for Comprehensive Utilization of Saline-Alkali Land, Dongying 257347, China
| | - Xinhua Li
- National Key Laboratory of Efficient Utilization of Nutrient Resources, Shandong Academy of Agricultural Sciences, Jinan 250100, China; (H.L.)
- National Technological Innovation Center for Comprehensive Utilization of Saline-Alkali Land, Dongying 257347, China
| |
Collapse
|
2
|
Kaczmarczyk O, Augustyniak D, Żak A. Imaging of Hydrated and Living Cells in Transmission Electron Microscope: Summary, Challenges, and Perspectives. ACS NANO 2025; 19:12710-12733. [PMID: 40156542 PMCID: PMC11984313 DOI: 10.1021/acsnano.5c00871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 04/01/2025]
Abstract
Transmission electron microscopy (TEM) is well-known for performing in situ studies in the nanoscale. Hence, scientists took this opportunity to explore the subtle processes occurring in living organisms. Nevertheless, such observations are complex─they require delicate samples kept in the liquid phase, low electron dose, and proper cell viability verification methods. Despite being highly demanding, so-called "live-cell" experiments have seen some degree of success. The presented review consists of an exhaustive literature review on reported "live-cell" studies and associated subjects, including liquid phase imaging, electron radiation interactions with liquids, and methods for cell viability testing. The challenges of modern, reliable research on living organisms are widely explained and discussed, and future perspectives for developing these techniques are presented.
Collapse
Affiliation(s)
- Olga Kaczmarczyk
- Institute
of Advanced Materials, Wroclaw University
of Science and Technology, 50-370 Wroclaw, Poland
| | - Daria Augustyniak
- Department
of Pathogen Biology and Immunology, Faculty of Biological Sciences, University of Wroclaw, 51-148 Wroclaw, Poland
| | - Andrzej Żak
- Institute
of Advanced Materials, Wroclaw University
of Science and Technology, 50-370 Wroclaw, Poland
- Department
of Material Science and Engineering, Massachusetts
Institute of Science and Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
3
|
Alhadrami HA, Sayed AM, Hassan HM, Rateb ME, Taha MN. Optimized peptide inhibitor Aqs1C targets LasR to disrupt quorum sensing and biofilm formation in Pseudomonas aeruginosa: Insights from MD simulations and in vitro studies. Int J Biol Macromol 2025; 300:140119. [PMID: 39855517 DOI: 10.1016/j.ijbiomac.2025.140119] [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: 09/01/2024] [Revised: 01/07/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
Pseudomonas aeruginosa (PA) is a critical pathogen, and its antibiotic resistance is largely driven by the quorum-sensing regulator LasR. Herein, we report the design, synthesis, and characterization of Aqs1C, a mutated peptide derivative of Aqs1, optimized to inhibit LasR and its quorum-sensing pathway. By introducing a targeted mutation, Aqs1C exhibited enhanced stability and binding affinity for LasR protein compared to its predecessor, Aqs1B. Using molecular dynamics simulations (MDS), the Aqs1C-LasR complex demonstrated a marked increase in structural stability, reflected in reduced root mean square deviation (RMSD) values and lower binding free energy. Electrostatic complementarity analysis showed stronger and more favorable interactions between Aqs1C and LasR. Further, GaMD experiments were able to reproduce the binding state between Aqs1C and LasR, indicating the binding mechanism between them. These molecular insights correlated with functional in vitro assays. Aqs1C effectively inhibited quorum-sensing-associated virulence factors in PA, involving biofilm formation (77.6 % inhibition), pyocyanin production (75.7 % inhibition), protease secretion (61.1 % inhibition), and rhamnolipid production (74.1 % inhibition), at a 100 μg/mL concentration, in a comparable or superior pattern to azithromycin (AZM). Molecular modelling, MDS, and GaMD insights and in vitro assays established Aqs1C as a promising candidate for therapeutic development to mitigate PA infections through targeted quorum-sensing disruption.
Collapse
Affiliation(s)
- Hani A Alhadrami
- Faculty of Applied Medical Sciences, Department of Medical Laboratory Sciences, King Abdulaziz University, P.O. Box 80402, Jeddah 21589, Saudi Arabia; King Fahd Medical Research Centre, King Abdulaziz University, P.O. Box 80402, Jeddah 21589, Saudi Arabia
| | - Ahmed M Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Hossam M Hassan
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, Beni-Suef 62513, Egypt; Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Mostafa E Rateb
- School of Computing, Engineering & Physical Sciences, University of the West of Scotland, Paisley PA1 2BE, UK.
| | - Mostafa N Taha
- Microbiology and Immunology Department, Faculty of Pharmacy, Nahda University, Beni-Suef 62764, Egypt
| |
Collapse
|
4
|
Smith E, Matthews A, Westra ER, Custodio R. Disruption of Pseudomonas aeruginosa quorum sensing influences biofilm formation without affecting antibiotic tolerance. MICROBIOLOGY (READING, ENGLAND) 2025; 171:001557. [PMID: 40279159 PMCID: PMC12032407 DOI: 10.1099/mic.0.001557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 04/07/2025] [Indexed: 04/26/2025]
Abstract
The opportunistic bacterial pathogen Pseudomonas aeruginosa is a leading cause of antimicrobial resistance-related deaths, and novel antimicrobial therapies are urgently required. P. aeruginosa infections are difficult to treat due to the bacterium's propensity to form biofilms, whereby cells aggregate to form a cooperative, protective structure. Autolysis, the self-killing of bacterial cells, and the bacterial cell-to-cell communication system, quorum sensing (QS), play essential roles in biofilm formation. Strains of P. aeruginosa that have lost the lasI/R QS system commonly develop in patients, and previous studies have characterized distinctive autolysis phenotypes in these strains. Yet, the underlying causes and implications of these autolysis phenotypes remain unknown. This study confirmed these autolysis phenotypes in the PA14 QS mutant strains, ΔlasI and ΔlasR, and investigated the consequences of QS loss and associated autolysis on biofilm formation and antibiotic susceptibility. QS mutants exhibited delayed biofilm formation but ultimately surpassed the wild-type (WT) in biofilm mass. However, the larger biofilm mass of the QS mutants was not reflected in higher live-cell numbers, indicating an altered biofilm structure. Nevertheless, QS mutant biofilms were not more susceptible to antibiotics than the WT. Artificial supplementation of ΔlasI with a QS signal molecule (autoinducer) restored the strain's QS system without the associated costs of QS, enabling ΔlasI to achieve higher pre-treatment and post-treatment live-cell numbers. Overall, the lack of QS functioning was not detrimental to biofilm antibiotic tolerance, though the artificial disruption of QS may reduce the advantages of QS mutants within in vivo mixed-strain populations. Much remains to be understood regarding the regulation and induction of the autolysis phenotypes observed in these strains, and future research to fully elucidate the control and consequences of autolysis may offer potential for novel antimicrobial therapies.
Collapse
Affiliation(s)
- Elvina Smith
- Environment and Sustainability Institute, Biosciences, University of Exeter, Penryn Campus, Penryn TR10 9FE, UK
| | - Andrew Matthews
- Environment and Sustainability Institute, Biosciences, University of Exeter, Penryn Campus, Penryn TR10 9FE, UK
| | - Edze R. Westra
- Environment and Sustainability Institute, Biosciences, University of Exeter, Penryn Campus, Penryn TR10 9FE, UK
| | - Rafael Custodio
- Environment and Sustainability Institute, Biosciences, University of Exeter, Penryn Campus, Penryn TR10 9FE, UK
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
5
|
Carey JN, Lamont S, Wozniak DJ, Dandekar AA, Parsek MR. Quorum sensing regulation of Psl polysaccharide production by Pseudomonas aeruginosa. J Bacteriol 2024; 206:e0031224. [PMID: 39530727 PMCID: PMC11656772 DOI: 10.1128/jb.00312-24] [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: 08/12/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Pseudomonas aeruginosa is a common opportunistic pathogen and a model organism for studying bacterial sociality. A social behavior of P. aeruginosa that is critical for its success as a pathogen is its ability to form protective biofilms. Many of P. aeruginosa's social phenotypes are regulated by quorum sensing-a type of cell-cell communication that allows bacteria to respond to population density. Although biofilm formation is known to be affected by quorum sensing, evidence for direct regulation of biofilm production by quorum regulators has remained elusive. In this work, we show that production of the major biofilm matrix polysaccharide Psl in P. aeruginosa PAO1 is regulated by the quorum regulators LasR and RhlR in stationary-phase cultures. Secretion of Psl into the culture medium requires LasR, RhlR, and the quorum signal molecules N-3-oxo-dodecanoyl-homoserine lactone and N-butanoyl homoserine lactone. Psl production in strains unable to synthesize the homoserine lactone signals can be restored by exogenous introduction of the signal molecules. We found that LasR and RhlR perform different roles in the regulation of Psl production: LasR acts at the promoter of the psl operon and activates transcription of the Psl biosynthetic genes, while RhlR activates translation of the psl transcripts. This work contributes to our understanding of the overlapping but distinct functions of the Las and Rhl quorum-sensing systems and implicates both in the direct regulation of biofilm matrix production.IMPORTANCEPseudomonas aeruginosa biofilms are responsible for many treatment-resistant infections in humans. Many cooperative behaviors in P. aeruginosa are controlled by quorum sensing, but evidence for a direct role of quorum sensing in the regulation of biofilm matrix production has been scant. In this work, we show that the Las and Rhl quorum-sensing systems have distinct roles in regulating production of the matrix polysaccharide Psl and that this regulation happens at the level of transcription (Las) and translation (Rhl) of the psl operon. These findings deepen our understanding of overlapping functions of Las and Rhl quorum sensing and the complex regulation of biofilm development in P. aeruginosa.
Collapse
Affiliation(s)
- Jeffrey N. Carey
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Sabrina Lamont
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Daniel J. Wozniak
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Ajai A. Dandekar
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Matthew R. Parsek
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
6
|
Zhang ZS, He Z, Shi Y, Guan M, Zhao DS, Zhu D, Xiong LT, Li Y, Deng X, Cui ZN. Structure-Based Discovery of Symmetric Disulfides from Garlic Extract as Pseudomonas aeruginosa Quorum Sensing Inhibitors. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20299-20307. [PMID: 39231265 DOI: 10.1021/acs.jafc.4c04404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Microorganisms are the most common cause of food spoilage. Pseudomonas aeruginosa is a common foodborne pathogen that causes food spoilage and poses a serious threat to food safety. As a crucial target in antitoxicity strategies, the quorum sensing (QS) system shows promising potential for further development. The garlic extract diallyl disulfide exhibits inhibitory activity against the QS system of P. aeruginosa, with disulfide bonds serving as the active component. However, the biological activity of other symmetric disulfides has not been investigated in this capacity. The study synthesized 39 disulfide bond-containing analogs and evaluated their activity as quorum sensing inhibitors (QSIs). The results showed that p-hydroxyphenyl substitution can replace the allyl groups while maintaining strong biological activity. The virulence factors production was reduced by compound 2i, with the strongest inhibitory effect being observed on elastase production. Synergistic inhibition was observed in the presence of antibiotics like ciprofloxacin and tobramycin. 2i successfully inhibited P. aeruginosa infection in the Galleria mellonella larvae model. Primary mechanism studies using transcriptome, surface plasmon resonance and molecular docking suggested that 2i inhibits the QS system by targeting the LasR protein. Thus, compound 2i could be used in developing QSIs for the control of P. aeruginosa infections.
Collapse
Affiliation(s)
- Zhao-Sheng Zhang
- State Key Laboratory of Green Pesticide, Integrative Microbiology Research Centre, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, College of Plant Protection, South China Agricultural University, Guangzhou 510642, China
| | - Zhe He
- State Key Laboratory of Green Pesticide, Integrative Microbiology Research Centre, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, College of Plant Protection, South China Agricultural University, Guangzhou 510642, China
| | - Yu Shi
- State Key Laboratory of Green Pesticide, Integrative Microbiology Research Centre, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, College of Plant Protection, South China Agricultural University, Guangzhou 510642, China
- Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan 512005, China
| | - Mingming Guan
- State Key Laboratory of Green Pesticide, Integrative Microbiology Research Centre, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, College of Plant Protection, South China Agricultural University, Guangzhou 510642, China
| | - Dong-Sheng Zhao
- Department of Pharmacy, Quanzhou Medical College, Quanzhou 362100, China
| | - Di Zhu
- State Key Laboratory of Green Pesticide, Integrative Microbiology Research Centre, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, College of Plant Protection, South China Agricultural University, Guangzhou 510642, China
| | - Lan-Tu Xiong
- State Key Laboratory of Green Pesticide, Integrative Microbiology Research Centre, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, College of Plant Protection, South China Agricultural University, Guangzhou 510642, China
| | - Yasheng Li
- Department of Infectious Diseases, Anhui Province Key Laboratory of Infectious Diseases & Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xin Deng
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Kowloon, Hong Kong SAR 999077, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
| | - Zi-Ning Cui
- State Key Laboratory of Green Pesticide, Integrative Microbiology Research Centre, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, College of Plant Protection, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
7
|
Zermeño-Pérez D, Chouirfa H, Rodriguez BJ, Dürig T, Duffy P, Cróinín TÓ. Bioresorbable Polyester Coatings with Antifouling and Antimicrobial Properties for Prevention of Biofilm Formation in Early Stage Infections on Ti6Al4V Hard-Tissue Implants. ACS APPLIED BIO MATERIALS 2024; 7:5728-5739. [PMID: 39037897 PMCID: PMC11337155 DOI: 10.1021/acsabm.4c00832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/24/2024]
Abstract
Implants made from titanium are used as prostheses because of their biocompatibility and their mechanical properties close to those of human bone. However, the risk of bacterial infection is always a major concern during surgery, and the development of biofilm can make these infections difficult to treat. A promising strategy to mitigate against bacterial infections is the use of antifouling and antimicrobial coatings, where bioresorbable polymers can play an important role due to their controlled degradability and sustained drug release, as well as excellent biocompatibility. In the present study, poly(d,l-lactide) (PDLLA) and poly[d,l-lactide-co-methyl ether poly(ethylene glycol)] (PDLLA-PEG) were studied, varying the PEG content (20-40% w/w) to analyze the effectiveness of PEG as an antifouling molecule. In addition, silver sulfadiazine (AgSD) was used as an additional antimicrobial agent with a concentration ≤5% w/w and incorporated into the PEGylated polymers to create a polymer with both antifouling and antimicrobial properties. Polymers synthesized were applied using spin coating to obtain homogeneous coatings to protect samples made from titanium/aluminum/vanadium (Ti6Al4V). The polymer coatings had a smoothing effect in comparison to that of the uncoated material, decreasing the contact area available for bacterial colonization. It was also noted that PEG addition into the polymeric chain developed amphiphilic materials with a decrease in contact angle from the most hydrophobic (Ti6Al4V) to the most hydrophilic PDLLA-PEG (60/40), highlighting the increase in water uptake contributing to the hydration layer formation, which confers the antifouling effect on the coating. This study demonstrated that the addition of PEG above 20% w/w and AgSD above 1% w/v into the formulation was able to decrease bacterial adherence against clinically relevant biofilm former strains Staphylococcus aureus and Pseudomonas aeruginosa.
Collapse
Affiliation(s)
- David Zermeño-Pérez
- Ashland
Specialties Ireland Ltd., Mullingar N91 F6PD, Ireland
- School
of Biomolecular and Biomedical Science, University College Dublin, Dublin Dublin 4, Ireland
| | - Hamza Chouirfa
- Ashland
Specialties Ireland Ltd., Mullingar N91 F6PD, Ireland
| | | | - Thomas Dürig
- Ashland
Wilmington Centre, Wilmington 19808, Delaware, United States
| | - Patrick Duffy
- Ashland
Specialties Ireland Ltd., Mullingar N91 F6PD, Ireland
| | - Tadhg Ó Cróinín
- School
of Biomolecular and Biomedical Science, University College Dublin, Dublin Dublin 4, Ireland
| |
Collapse
|
8
|
Eghbalpoor F, Gorji M, Alavigeh MZ, Moghadam MT. Genetically engineered phages and engineered phage-derived enzymes to destroy biofilms of antibiotics resistance bacteria. Heliyon 2024; 10:e35666. [PMID: 39170521 PMCID: PMC11336853 DOI: 10.1016/j.heliyon.2024.e35666] [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: 03/12/2024] [Revised: 07/27/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
"An impregnable stronghold where one or more warrior clans can evade enemy attacks" may serve as a description of bacterial biofilm on a smaller level than human conflicts. Consider this hypothetical conflict: who would emerge victorious? The occupants of secure trenches or those carrying out relentless assault? Either faction has the potential for triumph; the defenders will prevail if they can fortify the trench with unwavering resolve, while the assailants will succeed if they can devise innovative means to breach the trench. Hence, bacterial biofilms pose a significant challenge and are formidable adversaries for medical professionals, often leading to the failure of antibiotic treatments in numerous hospital infections. Phage engineering has become the foundation for the targeted enhancement of various phage properties, facilitating the eradication of biofilms. Researchers across the globe have studied the impact of engineered phages and phage-derived enzymes on biofilms formed by difficult-to-treat bacteria. These novel biological agents have shown promising results in addressing biofilm-related challenges. The compilation of research findings highlights the impressive capabilities of engineered phages in combating antibiotic-resistant bacteria, superbugs, and challenging infections. Specifically, these engineered phages exhibit enhanced biofilm destruction, penetration, and prevention capabilities compared to their natural counterparts. Additionally, the engineered enzymes derived from phages demonstrate improved effectiveness in addressing bacterial biofilms. As a result, these novel solutions, which demonstrate high penetration, destruction, and inhibition of biofilms, can be regarded as a viable option for addressing infectious biofilms in the near future.
Collapse
Affiliation(s)
- Fatemeh Eghbalpoor
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdieh Gorji
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Zamani Alavigeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
9
|
Mini M, Jayakumar D, Kumar P. In-silico and in-vitro assessment of the antibiofilm potential of azo dye, carmoisine against Pseudomonas aeruginosa. J Biomol Struct Dyn 2024; 42:6700-6710. [PMID: 37485898 DOI: 10.1080/07391102.2023.2237579] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023]
Abstract
Biofilm is a community of microorganisms attached to the substrate and plays a significant role in microbial pathogenesis and medical device-related infection. Pseudomonas aeruginosa (PA) is a highly infectious gram-negative opportunistic biofilm-forming bacterium with high antibiotic resistance. Several reports underscore the antimicrobial activity of natural and synthetic food coloring agents, including carmoisine, turmeric dye, red amaranth dye, and phloxine B. However, their ability to suppress the PA biofilm is not clearly understood. Carmoisine is a red-colored synthetic azo dye containing naphthalene subunits and sulfonic groups and is widely used as a food coloring agent. This study investigated the antibiofilm potential and possible mechanism of biofilm inhibition by carmoisine against PA. Computational studies through molecular docking revealed that carmoisine strongly binds to QS regulator LasR (-12.7) and relatively less strongly but significantly with WspR (-6.9). Further analysis of the docked LasR-carmoisine complex using 100 ns MD simulation (Desmond, Schrödinger) validated the bonding strength and stability. Crystal violet assay, triphenyl tetrazolium chloride salt assay, and confocal microscopic studies were adopted for biofilm quantification, and the results indicated the dose-dependent antibiofilm activity of carmoisine against PA. We hypothesise that the carmoisine-mediated reduction of biofilm in PA is due to its interaction with LasR and interference with the QS system. The computational and biochemical analysis of another compound, 1,2-naphthoquinone-4-sulphonic acid, reiterated the role of the naphthalene ring in biofilm inhibition. Hence, this work will pave the way for the future discovery of antibiofilm drugs based on naphthalene ring-based lead compounds.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Minsa Mini
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| | - Devi Jayakumar
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| | - Praveen Kumar
- Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| |
Collapse
|
10
|
Al-Rabia MW, Asfour HZ, Alhakamy NA, Bazuhair MA, Ibrahim TS, Abbas HA, Mansour B, Hegazy WAH, Seleem NM. Cilostazol is a promising anti-pseudomonal virulence drug by disruption of quorum sensing. AMB Express 2024; 14:87. [PMID: 39090255 PMCID: PMC11294311 DOI: 10.1186/s13568-024-01740-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024] Open
Abstract
Resistance to antibiotics is a critical growing public health problem that desires urgent action to combat. To avoid the stress on bacterial growth that evokes the resistance development, anti-virulence agents can be an attractive strategy as they do not target bacterial growth. Quorum sensing (QS) systems play main roles in controlling the production of diverse virulence factors and biofilm formation in bacteria. Thus, interfering with QS systems could result in mitigation of the bacterial virulence. Cilostazol is an antiplatelet and a vasodilator FDA approved drug. This study aimed to evaluate the anti-virulence activities of cilostazol in the light of its possible interference with QS systems in Pseudomonas aeruginosa. Additionally, the study examines cilostazol's impact on the bacterium's ability to induce infection in vivo, using sub-inhibitory concentrations to minimize the risk of resistance development. In this context, the biofilm formation, the production of virulence factors and influence on the in vivo ability to induce infection were assessed in the presence of cilostazol at sub-inhibitory concentration. Furthermore, the outcome of combination with antibiotics was evaluated. Cilostazol interfered with biofilm formation in P. aeruginosa. Moreover, swarming motility, biofilm formation and production of virulence factors were significantly diminished. Histopathological investigation revealed that liver, spleen and kidney tissues damage was abolished in mice injected with cilostazol-treated bacteria. Cilostazol exhibited a synergistic outcome when used in combination with antibiotics. At the molecular level, cilostazol downregulated the QS genes and showed considerable affinity to QS receptors. In conclusion, Cilostazol could be used as adjunct therapy with antibiotics for treating Pseudomonal infections. This research highlights cilostazol's potential to combat bacterial infections by targeting virulence mechanisms, reducing the risk of antibiotic resistance, and enhancing treatment efficacy against P. aeruginosa. These findings open avenues for repurposing existing drugs, offering new, safer, and more effective infection control strategies.
Collapse
Affiliation(s)
- Mohammed W Al-Rabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Hani Z Asfour
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Nabil A Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Mohammed A Bazuhair
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Tarek S Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Hisham A Abbas
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Basem Mansour
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt
- Department of Pharmaceutical Chemistry, Kut University College, Al Kut, Wasit, 52001, Iraq
| | - Wael A H Hegazy
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
- Department of Pharmaceutical Sciences, Pharmacy Program, College of Health Sciences, 113, Muscat, Oman.
| | - Noura M Seleem
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
11
|
Zuberi A, Ahmad N, Ahmad H, Saeed M, Ahmad I. Beyond antibiotics: CRISPR/Cas9 triumph over biofilm-associated antibiotic resistance infections. Front Cell Infect Microbiol 2024; 14:1408569. [PMID: 39035353 PMCID: PMC11257871 DOI: 10.3389/fcimb.2024.1408569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/27/2024] [Indexed: 07/23/2024] Open
Abstract
A complex structure known as a biofilm is formed when a variety of bacterial colonies or a single type of cell in a group sticks to a surface. The extracellular polymeric compounds that encase these cells, often consisting of proteins, eDNA, and polysaccharides, exhibit strong antibiotic resistance. Concerns about biofilm in the pharmaceutical industry, public health, and medical fields have sparked a lot of interest, as antibiotic resistance is a unique capacity exhibited by these biofilm-producing bacteria, which increases morbidity and death. Biofilm formation is a complicated process that is controlled by several variables. Insights into the processes to target for the therapy have been gained from multiple attempts to dissect the biofilm formation process. Targeting pathogens within a biofilm is profitable because the bacterial pathogens become considerably more resistant to drugs in the biofilm state. Although biofilm-mediated infections can be lessened using the currently available medications, there has been a lot of focus on the development of new approaches, such as bioinformatics tools, for both treating and preventing the production of biofilms. Technologies such as transcriptomics, metabolomics, nanotherapeutics and proteomics are also used to develop novel anti-biofilm agents. These techniques help to identify small compounds that can be used to inhibit important biofilm regulators. The field of appropriate control strategies to avoid biofilm formation is expanding quickly because of this spurred study. As a result, the current article addresses our current knowledge of how biofilms form, the mechanisms by which bacteria in biofilms resist antibiotics, and cutting-edge treatment approaches for infections caused by biofilms. Furthermore, we have showcased current ongoing research utilizing the CRISPR/Cas9 gene editing system to combat bacterial biofilm infections, particularly those brought on by lethal drug-resistant pathogens, concluded the article with a novel hypothesis and aspirations, and acknowledged certain limitations.
Collapse
Affiliation(s)
- Azna Zuberi
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, CO, United States
- Department of Obs & Gynae, Northwestern University, Chicago, IL, United States
| | - Nayeem Ahmad
- Department of Biophysics, All India Institute of Medical Science, New Delhi, India
- Department of Microbiology, Immunology, and Infectious Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Hafiz Ahmad
- Department of Medical Microbiology & Immunology, Ras Al Khaimah (RAK) College of Medical Sciences, Ras Al Khaimah (RAK) Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Mohd Saeed
- Department of Biology, College of Science University of Hail, Hail, Saudi Arabia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
12
|
Liu Y, Zhou H, Wang J, Liu S, Xie GJ, Liu BF, Xing D. Regulatory Mechanisms of Quorum Sensing System of Bacteria in Response to Chlorine and Ozone Disinfection. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024. [PMID: 38937254 DOI: 10.1021/acs.est.3c08305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Waterborne pathogens invariably present considerable threats to public health. The quorum sensing (QS) system is instrumental in coordinating bacterial growth and metabolisms. However, the responses and regulatory mechanisms of bacteria to various disinfection technologies through quorum sensing are still unclear. This study examines the inactivation effect of chlorination and ozonation on biofilms and planktonic cells of QS signaling-deficient mutants of Pseudomonas aeruginosa. Cell counting and viability assessment revealed that the combined disinfection of chlorine and ozone was the most effective for inactivating planktonic P. aeruginosa within 10 min of exposure. Additionally, microfluidic chip culture demonstrated that the secretion of quinolone signals escalated biofilms' disinfection resistance. Disinfection exposure significantly altered the gene expression of wild-type strains and QS signaling-deficient mutants. Moreover, the QS system triggered multilayered gene expression programs as a responsive protection to disinfectant exposure, including oxidative stress, ribosome synthesis, and the nutrient absorption of bacteria. These insights broaden our understanding of bacterial QS in response to disinfection, promising potential strategies toward efficient disinfection processes.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
- School of Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
| | - Huihui Zhou
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
- School of Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
| | - Jing Wang
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
- School of Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
| | - Sitong Liu
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
- School of Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
| | - Guo-Jun Xie
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
- School of Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
| | - Bing-Feng Liu
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
- School of Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
| | - Defeng Xing
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
- School of Environment, Harbin Institute of Technology, Harbin 150090, P. R. China
| |
Collapse
|
13
|
Hultqvist LD, Andersen JB, Nilsson CM, Jansen CU, Rybtke M, Jakobsen TH, Nielsen TE, Qvortrup K, Moser C, Graz M, Qvortrup K, Tolker-Nielsen T, Givskov M. High efficacy treatment of murine Pseudomonas aeruginosa catheter-associated urinary tract infections using the c-di-GMP modulating anti-biofilm compound Disperazol in combination with ciprofloxacin. Antimicrob Agents Chemother 2024; 68:e0148123. [PMID: 38717093 PMCID: PMC11620490 DOI: 10.1128/aac.01481-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 04/08/2024] [Indexed: 12/07/2024] Open
Abstract
Persistent urinary tract infections (UTIs) in hospitalized patients constitute an important medical problem. It is estimated that 75% of nosocomial UTIs are associated with urinary tract catheters with P. aeruginosa being a species that forms biofilms on these catheters. These infections are highly resistant to standard-of-care antibiotics, and the effects of the host immune defenses, which allows for development of persistent infections. With antibiotics losing their efficacy, new treatment options against resilient infections, such as catheter-associated urinary tract infections (CAUTIs), are critically needed. Central to our anti-biofilm approach is the manipulation of the c-di-GMP signaling pathway in P. aeruginosa to switch bacteria from the protective biofilm to the unprotected planktonic mode of life. We recently identified a compound (H6-335-P1), that stimulates the c-di-GMP degrading activity of the P. aeruginosa BifA protein which plummets the intracellular c-di-GMP content and induces dispersal of P. aeruginosa biofilm bacteria into the planktonic state. In the present study, we formulated H6-335-P1 as a hydrochloride salt (Disperazol), which is water-soluble and facilitates delivery via injection or oral administration. Disperazol can work as a monotherapy, but we observed a 100-fold improvement in efficacy when treating murine P. aeruginosa CAUTIs with a Disperazol/ciprofloxacin combination. Biologically active Disperazol reached the bladder 30 min after oral administration. Our study provides proof of concept that Disperazol can be used in combination with a relevant antibiotic for effective treatment of CAUTIs.
Collapse
Affiliation(s)
- Louise Dahl Hultqvist
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bo Andersen
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carl Martin Nilsson
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Morten Rybtke
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tim Holm Jakobsen
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Eiland Nielsen
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Qvortrup
- Department of Biomedical Sciences, CFIM, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus Moser
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Michael Graz
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Qvortrup
- Department of Chemistry, Technical University of Denmark, Lyngby, Denmark
| | - Tim Tolker-Nielsen
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Givskov
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Tuli JF, Ramezanpour M, Cooksley C, Bouras GS, Ogi K, Feizi S, Nepal R, Psaltis AJ, Wormald P, Vreugde S. Increased antibiotic resistance of Pseudomonas aeruginosa isolates from chronic rhinosinusitis patients grown in anaerobic conditions. Laryngoscope Investig Otolaryngol 2024; 9:e1244. [PMID: 38736943 PMCID: PMC11081418 DOI: 10.1002/lio2.1244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/04/2024] [Accepted: 03/16/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction In chronic rhinosinusitis (CRS), the congestion and blockage of the nose can cause anaerobic conditions within the sinus cavities which may promote the expression of virulence and antibiotic resistance genes in invading pathogens. Pseudomonas aeruginosa is a facultative anaerobic bacteria and causes severe recalcitrant CRS. In this study, we aimed to evaluate the antimicrobial resistance of P. aeruginosa isolates of CRS patients in planktonic and biofilm form grown in aerobic and anaerobic conditions. Methods P. aeruginosa clinical isolates of CRS patients (n = 25) were grown in planktonic and biofilm form in aerobic and anaerobic conditions. Minimum inhibitory concentrations (MIC) of planktonic forms and minimum biofilm eradication concentrations (MBEC) were determined. Additionally, metabolic activity by fluorescein diacetate assay, biofilm biomass by crystal violet assay and eDNA concentration were assessed in both conditions. Results P. aeruginosa planktonic cells grown in anaerobic condition exhibited increased gentamicin resistance (p < .01), whereas P. aeruginosa biofilms grown in anaerobic condition displayed significantly increased MBEC values for gentamicin (p < .0001) and levofloxacin (p < .001). The metabolic activity of anaerobic biofilms was significantly higher compared with aerobic biofilms (p < .0001). However, the biofilm biomass of isolates grown in aerobic conditions was higher than anaerobic conditions (p < .5). Conclusion P. aeruginosa isolates from CRS patients grown in anaerobic conditions showed significantly increased resistance to antibiotics with an increased metabolic activity but decreased biofilm biomass. Level of Evidence NA.
Collapse
Affiliation(s)
- Jannatul Ferdoush Tuli
- Department of Surgery–Otolaryngology, Head and Neck SurgeryUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, The Queen Elizabeth HospitalWoodville SouthSouth AustraliaAustralia
| | - Mahnaz Ramezanpour
- Department of Surgery–Otolaryngology, Head and Neck SurgeryUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, The Queen Elizabeth HospitalWoodville SouthSouth AustraliaAustralia
| | - Clare Cooksley
- Department of Surgery–Otolaryngology, Head and Neck SurgeryUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, The Queen Elizabeth HospitalWoodville SouthSouth AustraliaAustralia
| | - George Spyro Bouras
- Department of Surgery–Otolaryngology, Head and Neck SurgeryUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, The Queen Elizabeth HospitalWoodville SouthSouth AustraliaAustralia
| | - Kazuhiro Ogi
- Division of Otorhinolaryngology Head and Neck Surgery, Department of Sensory and Locomotor Medicine, Faculty of Medical SciencesUniversity of FukuiFukuiJapan
| | - Sholeh Feizi
- Department of Surgery–Otolaryngology, Head and Neck SurgeryUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, The Queen Elizabeth HospitalWoodville SouthSouth AustraliaAustralia
| | - Roshan Nepal
- CSIRO Marine and Atmospheric ResearchThe Commonwealth Scientific and Industrial Research Organisation (CSORO)HobartTasmaniaAustralia
| | - Alkis James Psaltis
- Department of Surgery–Otolaryngology, Head and Neck SurgeryUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, The Queen Elizabeth HospitalWoodville SouthSouth AustraliaAustralia
| | - Peter‐John Wormald
- Department of Surgery–Otolaryngology, Head and Neck SurgeryUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, The Queen Elizabeth HospitalWoodville SouthSouth AustraliaAustralia
| | - Sarah Vreugde
- Department of Surgery–Otolaryngology, Head and Neck SurgeryUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, The Queen Elizabeth HospitalWoodville SouthSouth AustraliaAustralia
| |
Collapse
|
15
|
Leitão MM, Vieira TF, Sousa SF, Borges F, Simões M, Borges A. Dual action of benzaldehydes: Inhibiting quorum sensing and enhancing antibiotic efficacy for controlling Pseudomonas aeruginosa biofilms. Microb Pathog 2024; 191:106663. [PMID: 38679246 DOI: 10.1016/j.micpath.2024.106663] [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: 02/14/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
Quorum sensing (QS) has a central role in biofilm lifestyle and antimicrobial resistance, and disrupting these signaling pathways is a promising strategy to control bacterial pathogenicity and virulence. In this study, the efficacy of three structurally related benzaldehydes (4-hydroxybenzaldehyde, 4-hydroxy-3-methoxybenzaldehyde (vanillin) and 4-hydroxy-3,5-dimethoxybenzaldehyde (syringaldehyde)) in disrupting the las and pqs systems of Pseudomonas aeruginosa was investigated using bioreporter strains and computational simulations. Additionally, these benzaldehydes were combined with tobramycin and ciprofloxacin antibiotics to evaluate their ability to increase antibiotic efficacy in preventing and eradicating P. aeruginosa biofilms. To this end, the total biomass, metabolic activity and culturability of the biofilm cells were determined. In vitro assays results indicated that the aromatic aldehydes have potential to inhibit the las and pqs systems by > 80 %. Molecular docking studies supported these findings, revealing the aldehydes binding in the same pocket as the natural ligands or receptor proteins (LasR, PQSA, PQSE, PQSR). Benzaldehydes were shown to act as virulence factor attenuators, with vanillin achieving a 48 % reduction in pyocyanin production. The benzaldehyde-tobramycin combination led not only to a 60 % reduction in biomass production but also to a 90 % reduction in the metabolic activity of established biofilms. A similar result was observed when benzaldehydes were combined with ciprofloxacin. 4-Hydroxybenzaldehyde demonstrated relevant action in increasing biofilm susceptibility to ciprofloxacin, resulting in a 65 % reduction in biomass. This study discloses, for the first time, that the benzaldehydes studied are potent QS inhibitors and also enhancers of antibiotics antibiofilm activity against P. aeruginosa.
Collapse
Affiliation(s)
- Miguel M Leitão
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal; CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007, Porto, Portugal
| | - Tatiana F Vieira
- LAQV/REQUIMTE, BioSIM-Department of Biomedicine, Faculty of Medicine, University of Porto, Rua Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Sérgio F Sousa
- LAQV/REQUIMTE, BioSIM-Department of Biomedicine, Faculty of Medicine, University of Porto, Rua Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Fernanda Borges
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007, Porto, Portugal
| | - Manuel Simões
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal; DEQ-Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal
| | - Anabela Borges
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal; DEQ-Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal.
| |
Collapse
|
16
|
Sompiyachoke K, Elias MH. Engineering quorum quenching acylases with improved kinetic and biochemical properties. Protein Sci 2024; 33:e4954. [PMID: 38520282 PMCID: PMC10960309 DOI: 10.1002/pro.4954] [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: 10/25/2023] [Revised: 01/03/2024] [Accepted: 02/21/2024] [Indexed: 03/25/2024]
Abstract
Many Gram-negative bacteria use N-acyl-L-homoserine lactone (AHL) signals to coordinate phenotypes such as biofilm formation and virulence factor production. Quorum-quenching enzymes, such as AHL acylases, chemically degrade these molecules which prevents signal reception by bacteria and inhibits undesirable biofilm-related traits. These capabilities make acylases appealing candidates for controlling microbes, yet candidates with high activity levels and substrate specificity and that are capable of being formulated into materials are needed. In this work, we undertook engineering efforts against two AHL acylases, PvdQ and MacQ, to generate these improved properties using the Protein One-Stop Shop Server. The engineering of acylases is complicated by low-throughput enzymatic assays. Alleviating this challenge, we report a time-course kinetic assay for AHL acylases that monitors the real-time production of homoserine lactone. Using the assay, we identified variants of PvdQ that were significantly stabilized, with melting point increases of up to 13.2°C, which translated into high resistance against organic solvents and increased compatibility with material coatings. While the MacQ mutants were unexpectedly destabilized, they had considerably improved kinetic properties, with >10-fold increases against N-butyryl-L-homoserine lactone and N-hexanoyl-L-homoserine lactone. Accordingly, these changes resulted in increased quenching abilities using a biosensor model and greater inhibition of virulence factor production of Pseudomonas aeruginosa PA14. While the crystal structure of one of the MacQ variants, M1, did not reveal obvious structural determinants explaining the observed changes in kinetics, it allowed for the capture of an acyl-enzyme intermediate that confirms a previously hypothesized catalytic mechanism of AHL acylases.
Collapse
Affiliation(s)
- Kitty Sompiyachoke
- Department of Biochemistry, Molecular Biology and BiophysicsUniversity of MinnesotaSt. PaulMinnesotaUSA
| | - Mikael H. Elias
- Department of Biochemistry, Molecular Biology and BiophysicsUniversity of MinnesotaSt. PaulMinnesotaUSA
- Biotechnology InstituteSt. PaulMinnesotaUSA
| |
Collapse
|
17
|
Gattu R, Ramesh SS, Ramesh S. Role of small molecules and nanoparticles in effective inhibition of microbial biofilms: A ray of hope in combating microbial resistance. Microb Pathog 2024; 188:106543. [PMID: 38219923 DOI: 10.1016/j.micpath.2024.106543] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
Microbial biofilms pose a severe threat to global health, as they are associated with deadly chronic infections and antibiotic resistance. To date, very few drugs are in clinical practice that specifically target microbial biofilms. Therefore, there is an urgent need for the development of novel therapeutic options targeting biofilm-related infections. In this review, we discuss nearly seventy-five different molecular scaffolds published over the last decade (2010-2023) which have exhibited their biofilm inhibition potential. For convenience, we have classified these into five different sub-groups based on their origin and design (excluding peptides as they are placed in between small molecules and biologics), namely, heterocycles; inorganic small molecules & metal complexes; small molecules decorated nanoparticles; small molecules derived from natural products (both plant and marine sources); and small molecules designed by in-silico approach. These antibiofilm agents are capable of disrupting microbial biofilms and can offer a promising avenue for future developments in human medicine. A hitherto review of this kind will lay a platform for the researchers to find new molecular entities to curb the serious menace of antimicrobial resistance especially caused by biofilms.
Collapse
Affiliation(s)
- Rohith Gattu
- Postgraduate Department of Chemistry, JSS College of Arts, Commerce and Science (A Recognized Research Centre of University of Mysore), Ooty Road, Mysuru, 570025, Karnataka, India
| | - Sanjay S Ramesh
- Postgraduate Department of Chemistry, JSS College of Arts, Commerce and Science (A Recognized Research Centre of University of Mysore), Ooty Road, Mysuru, 570025, Karnataka, India
| | - Suhas Ramesh
- Postgraduate Department of Chemistry, JSS College of Arts, Commerce and Science (A Recognized Research Centre of University of Mysore), Ooty Road, Mysuru, 570025, Karnataka, India.
| |
Collapse
|
18
|
Sousa AM, Ferreira D, Rodrigues LR, Pereira MO. Aptamer-based therapy for fighting biofilm-associated infections. J Control Release 2024; 367:522-539. [PMID: 38295992 DOI: 10.1016/j.jconrel.2024.01.061] [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: 09/30/2023] [Revised: 01/06/2024] [Accepted: 01/27/2024] [Indexed: 02/06/2024]
Abstract
Biofilms are key players in the pathogenesis of most of chronic infections associated with host tissue or fluids and indwelling medical devices. These chronic infections are hard to be treated due to the increased biofilms tolerance towards antibiotics in comparison to planktonic (or free living) cells. Despite the advanced understanding of their formation and physiology, biofilms continue to be a challenge and there is no standardized therapeutic approach in clinical practice to eradicate them. Aptamers offer distinctive properties, including excellent affinity, selectivity, stability, making them valuable tools for therapeutic purposes. This review explores the flexibility and designability of aptamers as antibiofilm drugs but, importantly, as targeting tools for diverse drug and delivery systems. It highlights specific examples of application of aptamers in biofilms of diverse species according to different modes of action including inhibition of motility and adhesion, blocking of quorum sensing molecules, and dispersal of biofilm-cells to planktonic state. Moreover, it discusses the limitations and challenges that impaired an increased success of the use of aptamers on biofilm management, as well as the opportunities related to aptamers modifications that can significantly expand their applicability on the biofilm field.
Collapse
Affiliation(s)
- Ana Margarida Sousa
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal.
| | - Débora Ferreira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Lígia Raquel Rodrigues
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
19
|
Jensen PØ, Olsen P, Dungu AM, Egelund GB, Jensen AV, Ravn P, Lindegaard B, Hertz FB, Bjarnsholt T, Faurholt-Jepsen D, Kolpen M. Bacterial aerobic respiration is a major consumer of oxygen in sputum from patients with acute lower respiratory tract infection. APMIS 2024. [PMID: 38284501 DOI: 10.1111/apm.13381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/15/2024] [Indexed: 01/30/2024]
Abstract
Bacterial aerobic respiration may determine the outcome of antibiotic treatment in experimental settings, but the clinical relevance of bacterial aerobic respiration for the outcome of antibiotic treatment has not been tested. Therefore, we hypothesized that bacterial aerobic respiration is higher in sputum from patients with acute lower respiratory tract infections (aLRTI), than in sputum from patients with chronic LRTI (cLRTI), where the bacteria persist despite antibiotic treatment. The bacterial aerobic respiration was determined according to the dynamics of the oxygen (O2 ) concentration in sputum from aLRTI patients (n = 52). This result was evaluated by comparison to previously published data from patients with cLRTI. O2 consumption resulting in anoxic zones was more frequent in sputum with detected bacterial pathogens. The bacterial aerobic respiration in aLRTI sputum approximated 55% of the total O2 consumption, which was significantly higher than previously published for cLRTI. The bacterial aerobic respiration in sputum was higher in aLRTI patients than previously seen in cLRTI patients, indicating the presence of bacteria with a sensitive physiology in aLRTI. These variations in bacterial physiology between aLRTI patients and cLRTI patients may contribute the huge difference in treatment success between the two patient groups.
Collapse
Affiliation(s)
- Peter Østrup Jensen
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
- Costerton Biofilm Center, Institute of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Pernille Olsen
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Arnold Matovu Dungu
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital - North Zealand, Hillerød, Denmark
| | - Gertrud Baunbaek Egelund
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital - North Zealand, Hillerød, Denmark
| | - Andreas Vestergaard Jensen
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital - North Zealand, Hillerød, Denmark
| | - Pernille Ravn
- Department of Medicine Section for Infectious Diseases, Herlev- Gentofte University Hospital, Hellerup, Denmark
| | - Birgitte Lindegaard
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital - North Zealand, Hillerød, Denmark
| | | | - Thomas Bjarnsholt
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
- Costerton Biofilm Center, Institute of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Mette Kolpen
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
20
|
Alasiri A, Soltane R, Taha MN, Abd El-Aleam RH, Alshehri F, Sayed AM. Bakuchiol inhibits Pseudomonas aeruginosa's quorum sensing-dependent biofilm formation by selectively inhibiting its transcriptional activator protein LasR. Int J Biol Macromol 2024; 255:128025. [PMID: 37979739 DOI: 10.1016/j.ijbiomac.2023.128025] [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: 08/06/2023] [Revised: 10/26/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023]
Abstract
In the present study, we characterized Bakuchiol (Bak) as a new potent quorum sensing (QS) inhibitor against Pseudomonas aeruginosa biofilm formation. Upon extensive in vitro investigations, Bak was found to suppress the P. aeruginosa biofilm formation (75.5 % inhibition) and its associated virulence factor e.g., pyocyanin and rhamnolipids (% of inhibition = 71.5 % and 66.9 %, respectively). Upon LuxR-type receptors assay, Bak was found to selectively inhibit P. aeruginosa's LasR in a dose-dependent manner. Further in-depth molecular investigations (e.g., sedimentation velocity and thermal shift assays) revealed that Bak destabilized LasR upon binding and disrupted its functioning quaternary structure (i.e., the functioning dimeric form). The subsequent modeling and molecular dynamics (MD) simulations explained in more molecular detail how Bak interacts with LasR and how it can induce its dimeric form disruption. In conclusion, our study identified Bak as a potent and specific LasR antagonist that should be widely used as a chemical probe of QS in P. aeruginosa, offering new insights into LasR antagonism processes. The new findings shed light on the cryptic world of LuxR-type QS in this important opportunistic pathogen.
Collapse
Affiliation(s)
- Ahlam Alasiri
- Department of Biology, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia.
| | - Raya Soltane
- Department of Biology, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia.
| | - Mostafa N Taha
- Microbiology and Immunology Department, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt.
| | - Rehab H Abd El-Aleam
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information MTI, Cairo 11571, Egypt.
| | - Fatma Alshehri
- Department of Biology, College of Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Ahmed M Sayed
- Pharmacognosy Department, Faculty of Pharmacy, Nahda University, Beni Suef, Egypt; Department of Pharmacognosy, College of Pharmacy, Almaaqal University, 61014 Basra, Iraq.
| |
Collapse
|
21
|
Yao T, Liu X, Li D, Huang Y, Yang W, Liu R, Wang Q, Li X, Zhou J, Jin C, Liu Y, Yang B, Pang Y. Two-component system RstAB promotes the pathogenicity of adherent-invasive Escherichia coli in response to acidic conditions within macrophages. Gut Microbes 2024; 16:2356642. [PMID: 38769708 PMCID: PMC11135836 DOI: 10.1080/19490976.2024.2356642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/14/2024] [Indexed: 05/22/2024] Open
Abstract
Adherent-invasive Escherichia coli (AIEC) strain LF82, isolated from patients with Crohn's disease, invades gut epithelial cells, and replicates in macrophages contributing to chronic inflammation. In this study, we found that RstAB contributing to the colonization of LF82 in a mouse model of chronic colitis by promoting bacterial replication in macrophages. By comparing the transcriptomes of rstAB mutant- and wild-type when infected macrophages, 83 significant differentially expressed genes in LF82 were identified. And we identified two possible RstA target genes (csgD and asr) among the differentially expressed genes. The electrophoretic mobility shift assay and quantitative real-time PCR confirmed that RstA binds to the promoters of csgD and asr and activates their expression. csgD deletion attenuated LF82 intracellular biofilm formation, and asr deletion reduced acid tolerance compared with the wild-type. Acidic pH was shown by quantitative real-time PCR to be the signal sensed by RstAB to activate the expression of csgD and asr. We uncovered a signal transduction pathway whereby LF82, in response to the acidic environment within macrophages, activates transcription of the csgD to promote biofilm formation, and activates transcription of the asr to promote acid tolerance, promoting its replication within macrophages and colonization of the intestine. This finding deepens our understanding of the LF82 replication regulation mechanism in macrophages and offers new perspectives for further studies on AIEC virulence mechanisms.
Collapse
Affiliation(s)
- Ting Yao
- TEDA (Tianjin Economic-Technological Development Area) Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Ministry of Education, Nankai University, Tianjin, China
| | - Xingmei Liu
- TEDA (Tianjin Economic-Technological Development Area) Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Ministry of Education, Nankai University, Tianjin, China
| | - Dan Li
- TEDA (Tianjin Economic-Technological Development Area) Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Ministry of Education, Nankai University, Tianjin, China
| | - Yu Huang
- TEDA (Tianjin Economic-Technological Development Area) Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Ministry of Education, Nankai University, Tianjin, China
| | - Wen Yang
- TEDA (Tianjin Economic-Technological Development Area) Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Ministry of Education, Nankai University, Tianjin, China
| | - Ruiying Liu
- TEDA (Tianjin Economic-Technological Development Area) Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Ministry of Education, Nankai University, Tianjin, China
| | - Qian Wang
- TEDA (Tianjin Economic-Technological Development Area) Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Ministry of Education, Nankai University, Tianjin, China
| | - Xueping Li
- TEDA (Tianjin Economic-Technological Development Area) Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Ministry of Education, Nankai University, Tianjin, China
| | - Jiarui Zhou
- TEDA (Tianjin Economic-Technological Development Area) Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Ministry of Education, Nankai University, Tianjin, China
| | - Chen Jin
- TEDA (Tianjin Economic-Technological Development Area) Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Ministry of Education, Nankai University, Tianjin, China
| | - Yutao Liu
- TEDA (Tianjin Economic-Technological Development Area) Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Ministry of Education, Nankai University, Tianjin, China
| | - Bin Yang
- TEDA (Tianjin Economic-Technological Development Area) Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Ministry of Education, Nankai University, Tianjin, China
| | - Yu Pang
- TEDA (Tianjin Economic-Technological Development Area) Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Ministry of Education, Nankai University, Tianjin, China
| |
Collapse
|
22
|
Sukmarini L, Atikana A, Hertiani T. Antibiofilm activity of marine microbial natural products: potential peptide- and polyketide-derived molecules from marine microbes toward targeting biofilm-forming pathogens. J Nat Med 2024; 78:1-20. [PMID: 37930514 DOI: 10.1007/s11418-023-01754-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023]
Abstract
Controlling and treating biofilm-related infections is challenging because of the widespread presence of multidrug-resistant microbes. Biofilm, a naturally occurring matrix of microbial aggregates, has developed intricate and diverse resistance mechanisms against many currently used antibiotics. This poses a significant problem, especially for human health, including clinically chronic infectious diseases. Thus, there is an urgent need to search for and develop new and more effective antibiotics. As the marine environment is recognized as a promising reservoir of new biologically active molecules with potential pharmacological properties, marine natural products, particularly those of microbial origin, have emerged as a promising source of antibiofilm agents. Marine microbes represent an untapped source of secondary metabolites with antimicrobial activity. Furthermore, marine natural products, owing to their self-defense mechanisms and adaptation to harsh conditions, encompass a wide range of chemical compounds, including peptides and polyketides, which are primarily found in microbes. These molecules can be exploited to provide novel and unique structures for developing alternative antibiotics as effective antibiofilm agents. This review focuses on the possible antibiofilm mechanism of these marine microbial molecules against biofilm-forming pathogens. It provides an overview of biofilm development, its recalcitrant mode of action, strategies for the development of antibiofilm agents, and their assessments. The review also revisits some selected peptides and polyketides from marine microbes reported between 2016 and 2023, highlighting their moderate and considerable antibiofilm activities. Moreover, their antibiofilm mechanisms, such as adhesion modulation/inhibition targeting biofilm-forming pathogens, quorum sensing intervention and inhibition, and extracellular polymeric substance disruption, are highlighted herein.
Collapse
Affiliation(s)
- Linda Sukmarini
- Research Center for Applied Microbiology, National Research and Innovation Agency (BRIN), KST Soekarno, Jl. Raya Jakarta-Bogor Km. 46, Cibinong, West Java, 16911, Indonesia.
- Indonesian Biofilm Research Collaboration Center, Jl. Farmako Sekip Utara, Yogyakarta, 55281, Indonesia.
| | - Akhirta Atikana
- Research Center for Applied Microbiology, National Research and Innovation Agency (BRIN), KST Soekarno, Jl. Raya Jakarta-Bogor Km. 46, Cibinong, West Java, 16911, Indonesia
- Indonesian Biofilm Research Collaboration Center, Jl. Farmako Sekip Utara, Yogyakarta, 55281, Indonesia
| | - Triana Hertiani
- Indonesian Biofilm Research Collaboration Center, Jl. Farmako Sekip Utara, Yogyakarta, 55281, Indonesia.
- Pharmaceutical Biology Department, Faculty of Pharmacy, Gadjah Mada University, Jl. Sekip Utara, Yogyakarta, 55281, Indonesia.
| |
Collapse
|
23
|
Porzio E, Andrenacci D, Manco G. Thermostable Lactonases Inhibit Pseudomonas aeruginosa Biofilm: Effect In Vitro and in Drosophila melanogaster Model of Chronic Infection. Int J Mol Sci 2023; 24:17028. [PMID: 38069351 PMCID: PMC10707464 DOI: 10.3390/ijms242317028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Pseudomonas aeruginosa is one of the six antimicrobial-resistant pathogens known as "ESKAPE" that represent a global threat to human health and are considered priority targets for the development of novel antimicrobials and alternative therapeutics. The virulence of P. aeruginosa is regulated by a four-chemicals communication system termed quorum sensing (QS), and one main class of QS signals is termed acylhomoserine lactones (acyl-HSLs), which includes 3-Oxo-dodecanoil homoserine lactone (3-Oxo-C12-HSL), which regulates the expression of genes implicated in virulence and biofilm formation. Lactonases, like Paraoxonase 2 (PON2) from humans and the phosphotriesterase-like lactonases (PLLs) from thermostable microorganisms, are able to hydrolyze acyl-HSLs. In this work, we explored in vitro and in an animal model the effect of some lactonases on the production of Pseudomonas virulence factors. This study presents a model of chronic infection in which bacteria were administered by feeding, and Drosophila adults were treated with enzymes and the antibiotic tobramycin, alone or in combination. In vitro, we observed significant effects of lactonases on biofilm formation as well as effects on bacterial motility and the expression of virulence factors. The treatment in vivo by feeding with the lactonase SacPox allowed us to significantly increase the biocidal effect of tobramycin in chronic infection.
Collapse
Affiliation(s)
- Elena Porzio
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Via P. Castellino 111, 80131 Naples, Italy
| | - Davide Andrenacci
- CNR Institute of Molecular Genetics “Luigi-Luca Cavalli-Sforza” Unit of Bologna, 40136 Bologna, Italy
| | - Giuseppe Manco
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Via P. Castellino 111, 80131 Naples, Italy
| |
Collapse
|
24
|
Fernández-Billón M, Llambías-Cabot AE, Jordana-Lluch E, Oliver A, Macià MD. Mechanisms of antibiotic resistance in Pseudomonas aeruginosa biofilms. Biofilm 2023; 5:100129. [PMID: 37205903 PMCID: PMC10189392 DOI: 10.1016/j.bioflm.2023.100129] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 05/21/2023] Open
Abstract
Pseudomonas aeruginosa is a major cause of life-threatening acute infections and life-long lasting chronic infections. The characteristic biofilm mode of life in P. aeruginosa chronic infections severely limits the efficacy of antimicrobial therapies, as it leads to intrinsic tolerance, involving physical and physiological factors in addition to biofilm-specific genes that can confer a transient protection against antibiotics promoting the development of resistance. Indeed, a striking feature of this pathogen is the extraordinary capacity to develop resistance to nearly all available antibiotics through the selection of chromosomal mutations, evidenced by its outstanding and versatile mutational resistome. This threat is dramatically amplified in chronic infections, driven by the frequent emergence of mutator variants with enhanced spontaneous mutation rates. Thus, this mini review is focused on describing the complex interplay of antibiotic resistance mechanisms in P. aeruginosa biofilms, to provide potentially useful information for the design of effective therapeutic strategies.
Collapse
Affiliation(s)
- María Fernández-Billón
- Department of Microbiology, Hospital Universitario Son Espases, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC), 28029, Madrid, Spain
| | - Aina E. Llambías-Cabot
- Department of Microbiology, Hospital Universitario Son Espases, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC), 28029, Madrid, Spain
| | - Elena Jordana-Lluch
- Department of Microbiology, Hospital Universitario Son Espases, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC), 28029, Madrid, Spain
| | - Antonio Oliver
- Department of Microbiology, Hospital Universitario Son Espases, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC), 28029, Madrid, Spain
| | - María D. Macià
- Department of Microbiology, Hospital Universitario Son Espases, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC), 28029, Madrid, Spain
- Corresponding author. Department of Microbiology, Hospital Universitario Son Espases, Crta. Vallemossa 79, 07120, Palma de Mallorca, Spain.
| |
Collapse
|
25
|
Soltane R, Alasiri A, Taha MN, Abd El-Aleam RH, Alghamdi KS, Ghareeb MA, Keshek DEG, Cardoso SM, Sayed AM. Norlobaridone Inhibits Quorum Sensing-Dependent Biofilm Formation and Some Virulence Factors in Pseudomonas aeruginosa by Disrupting Its Transcriptional Activator Protein LasR Dimerization. Biomolecules 2023; 13:1573. [PMID: 38002255 PMCID: PMC10669572 DOI: 10.3390/biom13111573] [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: 09/20/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 11/26/2023] Open
Abstract
In the present study, norlobaridone (NBD) was isolated from Parmotrema and then evaluated as a new potent quorum sensing (QS) inhibitor against Pseudomonas aeruginosa biofilm development. This phenolic natural product was found to reduce P. aeruginosa biofilm formation (64.6% inhibition) and its related virulence factors, such as pyocyanin and rhamnolipids (% inhibition = 61.1% and 55%, respectively). In vitro assays inhibitory effects against a number of known LuxR-type receptors revealed that NBD was able to specifically block P. aeruginosa's LasR in a dose-dependent manner. Further molecular studies (e.g., sedimentation velocity and thermal shift assays) demonstrated that NBD destabilized LasR upon binding and damaged its functional quaternary structure (i.e., the functional dimeric form). The use of modelling and molecular dynamics (MD) simulations also allowed us to further understand its interaction with LasR, and how this can disrupt its dimeric form. Finally, our findings show that NBD is a powerful and specific LasR antagonist that should be widely employed as a chemical probe in QS of P. aeruginosa, providing new insights into LasR antagonism processes. The new discoveries shed light on the mysterious world of LuxR-type QS in this key opportunistic pathogen.
Collapse
Affiliation(s)
- Raya Soltane
- Department of Basic Sciences, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Ahlam Alasiri
- Department of Basic Sciences, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Mostafa N. Taha
- Microbiology and Immunology Department, Faculty of Pharmacy, Nahda University, Beni-Suef 62764, Egypt;
| | - Rehab H. Abd El-Aleam
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11571, Egypt;
| | - Kawthar Saad Alghamdi
- Department of Biology, College of Science, University of Hafr Al Batin, Hafar Al Batin 39511, Saudi Arabia;
| | - Mosad A. Ghareeb
- Medicinal Chemistry Department, Theodor Bilharz Research Institute Kornaish El Nile, Warrak El-Hadar, Imbaba, P.O. Box 30, Giza 12411, Egypt;
| | - Doaa El-Ghareeb Keshek
- Department of Biology, Jumum College University, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
- Agriculture Genetic Engineering Research Institute (AGERI), Agriculture Research Center, Giza 11571, Egypt
| | - Susana M. Cardoso
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Ahmed M. Sayed
- Pharmacognosy Department, Faculty of Pharmacy, Nahda University, Beni-Suef 62513, Egypt
| |
Collapse
|
26
|
Bano S, Hassan N, Rafiq M, Hassan F, Rehman M, Iqbal N, Ali H, Hasan F, Kang YQ. Biofilms as Battlefield Armor for Bacteria against Antibiotics: Challenges and Combating Strategies. Microorganisms 2023; 11:2595. [PMID: 37894253 PMCID: PMC10609369 DOI: 10.3390/microorganisms11102595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/25/2023] [Accepted: 09/04/2023] [Indexed: 10/29/2023] Open
Abstract
Bacterial biofilms are formed by communities, which are encased in a matrix of extracellular polymeric substances (EPS). Notably, bacteria in biofilms display a set of 'emergent properties' that vary considerably from free-living bacterial cells. Biofilms help bacteria to survive under multiple stressful conditions such as providing immunity against antibiotics. Apart from the provision of multi-layered defense for enabling poor antibiotic absorption and adaptive persistor cells, biofilms utilize their extracellular components, e.g., extracellular DNA (eDNA), chemical-like catalase, various genes and their regulators to combat antibiotics. The response of biofilms depends on the type of antibiotic that comes into contact with biofilms. For example, excessive production of eDNA exerts resistance against cell wall and DNA targeting antibiotics and the release of antagonist chemicals neutralizes cell membrane inhibitors, whereas the induction of protein and folic acid antibiotics inside cells is lowered by mutating genes and their regulators. Here, we review the current state of knowledge of biofilm-based resistance to various antibiotic classes in bacteria and genes responsible for biofilm development, and the key role of quorum sensing in developing biofilms and antibiotic resistance is also discussed. In this review, we also highlight new and modified techniques such as CRISPR/Cas, nanotechnology and bacteriophage therapy. These technologies might be useful to eliminate pathogens residing in biofilms by combating biofilm-induced antibiotic resistance and making this world free of antibiotic resistance.
Collapse
Affiliation(s)
- Sara Bano
- Applied Environmental and Geomicrobiology Laboratory, Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Noor Hassan
- Industrial Biotechnology Division, National Institute for Biotechnology and Genetic Engineering-College, Pakistan Institute of Engineering and Applied Sciences, Islamabad 44000, Pakistan
| | - Muhammad Rafiq
- Department of Microbiology, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta 87300, Pakistan
| | - Farwa Hassan
- Industrial Biotechnology Division, National Institute for Biotechnology and Genetic Engineering-College, Pakistan Institute of Engineering and Applied Sciences, Islamabad 44000, Pakistan
| | - Maliha Rehman
- Department of Microbiology, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta 87300, Pakistan
| | - Naveed Iqbal
- Department of Biotechnology & Informatics, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta 87300, Pakistan
- The Department of Paediatrics and Child Health, Aga Khan University, Karachi 74800, Pakistan
| | - Hazrat Ali
- Industrial Biotechnology Division, National Institute for Biotechnology and Genetic Engineering-College, Pakistan Institute of Engineering and Applied Sciences, Islamabad 44000, Pakistan
| | - Fariha Hasan
- Applied Environmental and Geomicrobiology Laboratory, Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Ying-Qian Kang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education of Guizhou, Guiyang 550025, China
- Key Laboratory of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China
| |
Collapse
|
27
|
Sompiyachoke K, Elias MH. Engineering Quorum Quenching Acylases with Improved Kinetic and Biochemical Properties. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.01.555929. [PMID: 37693529 PMCID: PMC10491313 DOI: 10.1101/2023.09.01.555929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Many Gram-negative bacteria respond to N-acyl-L-homoserine lactone (AHL) signals to coordinate phenotypes such as biofilm formation and virulence factor production. Quorum-quenching enzymes, such as acylases, chemically degrade AHL signals, prevent signal reception by bacteria, and inhibit undesirable traits related to biofilm. These capabilities make these enzymes appealing candidates for controlling microbes. Yet, enzyme candidates with high activity levels, high substrate specificity for specific interference, and that are capable of being formulated into materials are needed. In this work, we undertook engineering efforts against two AHL acylases, PvdQ and MacQ, to obtain improved acylase variants. The engineering of acylase is complicated by low-throughput enzymatic assays. To alleviate this challenge, we report a time-course kinetic assay for AHL acylase that tracks the real-time production of homoserine lactone. Using the protein one-stop shop server (PROSS), we identified variants of PvdQ that were significantly stabilized, with melting point increases of up to 13.2 °C, which translated into high resistance against organic solvents and increased compatibility with material coatings. We also generated mutants of MacQ with considerably improved kinetic properties, with >10-fold increases against N-butyryl-L-homoserine lactone and N-hexanoyl-L-homoserine lactone. In fact, the variants presented here exhibit unique combinations of stability and activity levels. Accordingly, these changes resulted in increased quenching abilities using a biosensor model and greater inhibition of virulence factor production of Pseudomonas aeruginosa PA14. While the crystal structure of one of the MacQ variants, M1, did not reveal obvious structural determinants explaining the observed changes in kinetics, it allowed for the capture of an acyl-enzyme intermediate that confirms a previously hypothesized catalytic mechanism of AHL acylases.
Collapse
Affiliation(s)
- Kitty Sompiyachoke
- University of Minnesota, Department of Biochemistry, Molecular Biology and Biophysics, St. Paul, MN, 55108, USA
| | - Mikael H. Elias
- University of Minnesota, Department of Biochemistry, Molecular Biology and Biophysics, St. Paul, MN, 55108, USA
- University of Minnesota, Biotechnology Institute, St. Paul, MN, 55108, USA
| |
Collapse
|
28
|
Stewart PS, Owkes M. Simulation of catalase-dependent tolerance of microbial biofilm to hydrogen peroxide with a biofilm computer model. NPJ Biofilms Microbiomes 2023; 9:60. [PMID: 37612330 PMCID: PMC10447567 DOI: 10.1038/s41522-023-00426-z] [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: 05/03/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023] Open
Abstract
Hydrogen peroxide (HP) is a common disinfectant and antiseptic. When applied to a biofilm, it may be expected that the top layer of the biofilm would be killed by HP, the HP would penetrate further, and eventually eradicate the entire biofilm. However, using the Biofilm.jl computer model, we demonstrate a mechanism by which the biofilm can persist, and even become thicker, in the indefinite treatment with an HP solution at concentrations that are lethal to planktonic microorganisms. This surprising result is found to be dependent on the neutralization of HP by dead biomass, which provides protection for living biomass deeper within the biofilm. Practically, to control a biofilm, this result leads to the concept of treating with an HP dose exceeding a critical threshold concentration rather than a sustained, lower-concentration treatment.
Collapse
Affiliation(s)
- Philip S Stewart
- Chemical & Biological Engineering, Montana State University, Bozeman, 59717, MT, USA.
| | - Mark Owkes
- Mechanical & Industrial Engineering, Montana State University, Bozeman, 59717, MT, USA.
| |
Collapse
|
29
|
Abisado-Duque RG, Townsend KA, Mckee BM, Woods K, Koirala P, Holder AJ, Craddock VD, Cabeen M, Chandler JR. An Amino Acid Substitution in Elongation Factor EF-G1A Alters the Antibiotic Susceptibility of Pseudomonas aeruginosa LasR-Null Mutants. J Bacteriol 2023; 205:e0011423. [PMID: 37191503 PMCID: PMC10294626 DOI: 10.1128/jb.00114-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 04/22/2023] [Indexed: 05/17/2023] Open
Abstract
The opportunistic bacterium Pseudomonas aeruginosa uses the LasR-I quorum-sensing system to increase resistance to the aminoglycoside antibiotic tobramycin. Paradoxically, lasR-null mutants are commonly isolated from chronic human infections treated with tobramycin, suggesting there may be a mechanism that permits the emergence of lasR-null mutants under tobramycin selection. We hypothesized that some other genetic mutations that emerge in these isolates might modulate the effects of lasR-null mutations on antibiotic resistance. To test this hypothesis, we inactivated lasR in several highly tobramycin-resistant isolates from long-term evolution experiments. In some of these isolates, inactivating lasR further increased resistance, compared with decreasing resistance of the wild-type ancestor. These strain-dependent effects were due to a G61A nucleotide polymorphism in the fusA1 gene encoding amino acid substitution A21T in the translation elongation factor EF-G1A. The EF-G1A mutational effects required the MexXY efflux pump and the MexXY regulator ArmZ. The fusA1 mutation also modulated ΔlasR mutant resistance to two other antibiotics, ciprofloxacin and ceftazidime. Our results identify a gene mutation that can reverse the direction of the antibiotic selection of lasR mutants, a phenomenon known as sign epistasis, and provide a possible explanation for the emergence of lasR-null mutants in clinical isolates. IMPORTANCE One of the most common mutations in Pseudomonas aeruginosa clinical isolates is in the quorum sensing lasR gene. In laboratory strains, lasR disruption decreases resistance to the clinical antibiotic tobramycin. To understand how lasR mutations emerge in tobramycin-treated patients, we mutated lasR in highly tobramycin-resistant laboratory strains and determined the effects on resistance. Disrupting lasR enhanced the resistance of some strains. These strains had a single amino acid substitution in the translation factor EF-G1A. The EF-G1A mutation reversed the selective effects of tobramycin on lasR mutants. These results illustrate how adaptive mutations can lead to the emergence of new traits in a population and are relevant to understanding how genetic diversity contributes to the progression of disease during chronic infections.
Collapse
Affiliation(s)
| | - Kade A. Townsend
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Brielle M. Mckee
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Kathryn Woods
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Pratik Koirala
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Alexandra J. Holder
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Vaughn D. Craddock
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Matthew Cabeen
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | | |
Collapse
|
30
|
Ambreetha S, Singh V. Genetic and environmental determinants of surface adaptations in Pseudomonas aeruginosa. MICROBIOLOGY (READING, ENGLAND) 2023; 169. [PMID: 37276014 DOI: 10.1099/mic.0.001335] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Pseudomonas aeruginosa
is a well-studied Gram-negative opportunistic bacterium that thrives in markedly varied environments. It is a nutritionally versatile microbe that can colonize a host as well as exist in the environment. Unicellular, planktonic cells of
P. aeruginosa
can come together to perform a coordinated swarming movement or turn into a sessile, surface-adhered population called biofilm. These collective behaviours produce strikingly different outcomes. While swarming motility rapidly disseminates the bacterial population, biofilm collectively protects the population from environmental stresses such as heat, drought, toxic chemicals, grazing by predators, and attack by host immune cells and antibiotics. The ubiquitous nature of
P. aeruginosa
is likely to be supported by the timely transition between planktonic, swarming and biofilm lifestyles. The social behaviours of this bacteria viz biofilm and swarm modes are controlled by signals from quorum-sensing networks, LasI-LasR, RhlI-RhlR and PQS-MvfR, and several other sensory kinases and response regulators. A combination of environmental and genetic cues regulates the transition of the
P. aeruginosa
population to specific states. The current review is aimed at discussing key factors that promote physiologically distinct transitioning of the
P. aeruginosa
population.
Collapse
Affiliation(s)
- Sakthivel Ambreetha
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka - 560012, India
| | - Varsha Singh
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka - 560012, India
| |
Collapse
|
31
|
Brockhurst M, Cavet J, Diggle SP, Grainger D, Mangenelli R, Sychrova H, Martin-Verstraete I, Welch M, Palmer T, Thomas GH. Shaping microbiology for 75 years: highlights of research published in Microbiology. Part 2 - Communities and evolution. MICROBIOLOGY (READING, ENGLAND) 2023; 169. [PMID: 37379228 DOI: 10.1099/mic.0.001357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Affiliation(s)
- Michael Brockhurst
- Division of Evolution, Infection and Genomics, University of Manchester, Michael Smith Building, Dover Street, Manchester M13 9PT, UK
| | - Jennifer Cavet
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Stephen P Diggle
- Center for Microbial Dynamics & Infection, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - David Grainger
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | | - Hana Sychrova
- Institute of Physiology of the Czech Academy of Sciences, Laboratory of Membrane Transport, 14200 Prague 4, Czech Republic
| | | | - Martin Welch
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Tracy Palmer
- Microbes in Health and Disease Theme, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Gavin H Thomas
- Department of Biology, University of York, Wentworth Way, York, UK
| |
Collapse
|
32
|
Ye X, Mao S, Li Y, Yang Z, Du A, Wang H. Design, Synthesis, and Biological Evaluation of Phenyloxadiazole Sulfoxide Derivatives as Potent Pseudomonas aeruginosa Biofilm Inhibitors. Molecules 2023; 28:molecules28093879. [PMID: 37175289 PMCID: PMC10180516 DOI: 10.3390/molecules28093879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
With the development of antimicrobial agents, researchers have developed new strategies through key regulatory systems to block the expression of virulence genes without affecting bacterial growth. This strategy can minimize the selective pressure that leads to the emergence of resistance. Quorum sensing (QS) is an intercellular communication system that plays a key role in the regulation of bacterial virulence and biofilm formation. Studies have revealed that the QS system controls 4-6% of the total number of P. aeruginosa genes, and quorum sensing inhibitors (QSIs) could be a promising target for developing new prevention and treatment strategies against P. aeruginosa infection. In this study, four series of phenyloxadiazole and phenyltetrazole sulfoxide derivatives were synthesized and evaluated for their inhibitory effects on P. aeruginosa PAO1 biofilm formation. Our results showed that 5b had biofilm inhibitory activity and reduced the production of QS-regulated virulence factors in P. aeruginosa. In addition, silico molecular docking studies have shown that 5b binds to the P. aeruginosa QS receptor protein LasR through hydrogen bond interaction. Preliminary structure-activity relationship and docking studies show that 5b has broad application prospects as an anti-biofilm compound, and further research will be carried out in the future to solve the problem of microbial resistance.
Collapse
Affiliation(s)
- Xinyi Ye
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Shen Mao
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yasheng Li
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Zhikun Yang
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Aoqi Du
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hong Wang
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
33
|
Abisado-Duquea RG, McKee BM, Townsend KA, Woods K, Koirala P, Holder AJ, Craddock VD, Cabeen MT, Chandler JR. Tobramycin adaptation alters the antibiotic susceptibility of Pseudomonas aeruginosa quorum sensing-null mutants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.13.523864. [PMID: 36711731 PMCID: PMC9882136 DOI: 10.1101/2023.01.13.523864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The opportunistic bacterium Pseudomonas aeruginosa uses the LasR-I quorum sensing system to increase resistance to the aminioglycoside antibiotic tobramycin. Paradoxically, lasR-null mutants are commonly isolated from chronic human infections treated with tobramycin, suggesting there may be a mechanism allowing the lasR-null mutants to persist under tobramycin selection. We hypothesized that the effects of inactivating lasR on tobramycin resistance might be dependent on the presence or absence of other gene mutations in that strain, a phenomenon known as epistasis. To test this hypothesis, we inactivated lasR in several highly tobramycin-resistant isolates from long-term evolution experiments. We show that the effects of ΔlasR on tobramycin resistance are strain dependent. The effects can be attributed to a point mutation in the gene encoding the translation elongation factor fusA1 (G61A nucleotide substitution), which confers a strong selective advantage to lasR-null PA14 under tobramycin selection. This fusA1 G61A mutation results in increased activity of the MexXY efflux pump and expression of the mexXY regulator ArmZ. The fusA1 mutation can also modulate ΔlasR mutant resistance to two other antibiotics, ciprofloxacin and ceftazidime. Our results demonstrate the importance of epistatic gene interactions on antibiotic susceptibility of lasR-null mutants. These results support of the idea that gene interactions might play a significant role in the evolution of quorum sensing in P. aeruginosa.
Collapse
|
34
|
Uruén C, García C, Fraile L, Tommassen J, Arenas J. How Streptococcus suis escapes antibiotic treatments. Vet Res 2022; 53:91. [DOI: 10.1186/s13567-022-01111-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
AbstractStreptococcus suis is a zoonotic agent that causes sepsis and meningitis in pigs and humans. S. suis infections are responsible for large economic losses in pig production. The lack of effective vaccines to prevent the disease has promoted the extensive use of antibiotics worldwide. This has been followed by the emergence of resistance against different classes of antibiotics. The rates of resistance to tetracyclines, lincosamides, and macrolides are extremely high, and resistance has spread worldwide. The genetic origin of S. suis resistance is multiple and includes the production of target-modifying and antibiotic-inactivating enzymes and mutations in antibiotic targets. S. suis genomes contain traits of horizontal gene transfer. Many mobile genetic elements carry a variety of genes that confer resistance to antibiotics as well as genes for autonomous DNA transfer and, thus, S. suis can rapidly acquire multiresistance. In addition, S. suis forms microcolonies on host tissues, which are associations of microorganisms that generate tolerance to antibiotics through a variety of mechanisms and favor the exchange of genetic material. Thus, alternatives to currently used antibiotics are highly demanded. A deep understanding of the mechanisms by which S. suis becomes resistant or tolerant to antibiotics may help to develop novel molecules or combinations of antimicrobials to fight these infections. Meanwhile, phage therapy and vaccination are promising alternative strategies, which could alleviate disease pressure and, thereby, antibiotic use.
Collapse
|
35
|
Jayakumar P, Figueiredo ART, Kümmerli R. Evolution of Quorum Sensing in Pseudomonas aeruginosa Can Occur via Loss of Function and Regulon Modulation. mSystems 2022; 7:e0035422. [PMID: 36190124 PMCID: PMC9600717 DOI: 10.1128/msystems.00354-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 09/12/2022] [Indexed: 12/24/2022] Open
Abstract
Pseudomonas aeruginosa populations evolving in cystic fibrosis lungs, animal hosts, natural environments and in vitro undergo extensive genetic adaption and diversification. A common mutational target is the quorum sensing (QS) system, a three-unit regulatory system that controls the expression of virulence factors and secreted public goods. Three evolutionary scenarios have been advocated to explain selection for QS mutants: (i) disuse of the regulon, (ii) cheating through the exploitation of public goods, or (ii) modulation of the QS regulon. Here, we examine these scenarios by studying a set of 61 QS mutants from an experimental evolution study. We observed nonsynonymous mutations in all three QS systems: Las, Rhl, and Pseudomonas Quinolone Signal (PQS). The majority of the Las mutants had large deletions of the Las regulon, resulting in loss of QS function and the inability to produce QS-regulated traits, thus supporting the first or second scenarios. Conversely, phenotypic and gene expression analyses of Rhl mutants support network modulation (third scenario), as these mutants overexpressed the Las and Rhl receptors and showed an altered QS-regulated trait production profile. PQS mutants also showed patterns of regulon modulation leading to strain diversification and phenotypic tradeoffs, where the upregulation of certain QS traits is associated with the downregulation of others. Overall, our results indicate that mutations in the different QS systems lead to diverging effects on the QS trait profile in P. aeruginosa populations. These mutations might not only affect the plasticity and diversity of evolved populations but could also impact bacterial fitness and virulence in infections. IMPORTANCE Pseudomonas aeruginosa uses quorum sensing (QS), a three-unit multilayered network, to coordinate expression of traits required for growth and virulence in the context of infections. Despite its importance for bacterial fitness, the QS regulon appears to be a common mutational target during long-term adaptation of P. aeruginosa in the host, natural environments, and experimental evolutions. This raises questions of why such an important regulatory system is under selection and how mutations change the profile of QS-regulated traits. Here, we examine a set of 61 experimentally evolved QS mutants to address these questions. We found that mutations involving the master regulator, LasR, resulted in an almost complete breakdown of QS, while mutations in RhlR and PqsR resulted in modulations of the regulon, where both the regulon structure and the QS-regulated trait profile changed. Our work reveals that natural selection drives diversification in QS activity patterns in evolving populations.
Collapse
Affiliation(s)
- Priyanikha Jayakumar
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Alexandre R. T. Figueiredo
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Department of Evolutionary Biology and Environmental Studies, University of Zurich, Zurich, Switzerland
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Rolf Kümmerli
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
36
|
Lichtenberg M, Kragh KN, Fritz B, Kirkegaard JB, Tolker-Nielsen T, Bjarnsholt T. Cyclic-di-GMP signaling controls metabolic activity in Pseudomonas aeruginosa. Cell Rep 2022; 41:111515. [DOI: 10.1016/j.celrep.2022.111515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 07/13/2022] [Accepted: 09/26/2022] [Indexed: 11/03/2022] Open
|
37
|
Bernabè G, Marzaro G, Di Pietra G, Otero A, Bellato M, Pauletto A, Scarpa M, Sut S, Chilin A, Dall’Acqua S, Brun P, Castagliuolo I. A novel phenolic derivative inhibits AHL-dependent quorum sensing signaling in Pseudomonas aeruginosa. Front Pharmacol 2022; 13:996871. [PMID: 36204236 PMCID: PMC9531014 DOI: 10.3389/fphar.2022.996871] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Increasing antibiotic resistance and the decline in the pharmaceutical industry’s investments have amplified the need for novel treatments for multidrug-resistant bacteria. Quorum sensing (QS) inhibitors reduce pathogens’ virulence without selective pressure on bacteria and provide an alternative to conventional antibiotic-based therapies. P. aeruginosa uses complex QS signaling to control virulence and biofilm formation. We aimed to identify inhibitors of P. aeruginosa QS acting on acyl-homoserine lactones (AHL)-mediated circuits. Bioluminescence and qRT-PCR assays were employed to screen a library of 81 small phenolic derivatives to reduce AHL-dependent signaling. We identified GM-50 as the most active compound inhibiting the expression of AHL-regulated genes but devoid of cytotoxic activity in human epithelial cells and biocidal effects on bacteria. GM-50 reduces virulence factors such as rhamnolipids, pyocyanin, elastase secretion, and swarming motility in P. aeruginosa PAO1 laboratory strain. By molecular docking, we provide evidence that GM-50 highly interacts with RhlR. GM-50 significantly improved aztreonam-mediated biofilm disruption. Moreover, GM-50 prevents adhesion of PAO1 and inflammatory damage in the human A549 cell line and protects Galleria mellonella from PAO1-mediated killing. GM-50 significantly reduces virulence factors in 20 P. aeruginosa clinical isolates from patients with respiratory tract infections. In conclusion, GM-50 inhibits AHL-signaling, reduces virulence factors, enhances the anti-biofilm activity of aztreonam, and protects G. mellonella larvae from damage induced by P. aeruginosa. Since GM-50 is active on clinical strains, it represents a starting point for identifying and developing new phenolic derivatives acting as QS-inhibitors in P. aeruginosa infections.
Collapse
Affiliation(s)
- Giulia Bernabè
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Giovanni Marzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | | | - Ana Otero
- Departamento de Microbioloxía e Parasitoloxía, Facultade de Bioloxía-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Massimo Bellato
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Anthony Pauletto
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Melania Scarpa
- Laboratory of Advanced Translational Research, Veneto Institute of Oncology IOV—IRCCS, Padua, Italy
| | - Stefania Sut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Adriana Chilin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Stefano Dall’Acqua
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Paola Brun
- Department of Molecular Medicine, University of Padua, Padua, Italy
- *Correspondence: Paola Brun,
| | | |
Collapse
|
38
|
Assessment of Bacteriocin-Antibiotic Synergy for the Inhibition and Disruption of Biofilms of Listeria monocytogenes and Vancomycin-Resistant Enterococcus. MICROBIOLOGY RESEARCH 2022. [DOI: 10.3390/microbiolres13030033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In this study, we have evaluated the effects of previously characterized bacteriocins produced by E. faecium strains ST651ea, ST7119ea, and ST7319ea, against biofilm formation and biofilms formed by L. monocytogenes ATCC15313 and vancomycin-resistant E. faecium VRE19. The effects of bacteriocins on the biofilms formed by L. monocytogenes ATCC151313 were evaluated by crystal violet assay and further confirmed by quantifying viable cells and cell metabolic activities through flow cytometry and TTC assay, respectively, indicating that bacteriocin activities required to completely eradicate biofilms are at least 1600 AU mL−1, 3200 AU mL−1, and 6400 AU mL−1, respectively for each bacteriocin evaluated. Furthermore, bacteriocins ST651ea and ST7119ea require at least 6400 AU mL−1 to completely eradicate the viability of cells within the biofilms formed by E. faecium VRE19, while bacteriocin ST7319ea requires at least 12800 AU mL−1 to obtain the same observations. Assessment of synergistic activities between selected conventional antibiotics (ciprofloxacin and vancomycin) with these bacteriocins was carried out to evaluate their effects on biofilm formation and pre-formed biofilms of both test microorganisms. Results showed that higher concentrations are needed to completely eradicate metabolic activities of cells within pre-formed biofilms in contrast with the biofilm formation abilities of the strains. Furthermore, synergistic activities of bacteriocins with both ciprofloxacin and vancomycin are more evident against vancomycin-resistant E. faecium VRE19 rather than L. monocytogenes ATCC15313. These observations can be further explored for possible applications of these combinations of antibiotics as a possible treatment of clinically relevant pathogens.
Collapse
|
39
|
Dutt Y, Dhiman R, Singh T, Vibhuti A, Gupta A, Pandey RP, Raj VS, Chang CM, Priyadarshini A. The Association between Biofilm Formation and Antimicrobial Resistance with Possible Ingenious Bio-Remedial Approaches. Antibiotics (Basel) 2022; 11:930. [PMID: 35884186 PMCID: PMC9312340 DOI: 10.3390/antibiotics11070930] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/03/2022] [Accepted: 07/06/2022] [Indexed: 02/01/2023] Open
Abstract
Biofilm has garnered a lot of interest due to concerns in various sectors such as public health, medicine, and the pharmaceutical industry. Biofilm-producing bacteria show a remarkable drug resistance capability, leading to an increase in morbidity and mortality. This results in enormous economic pressure on the healthcare sector. The development of biofilms is a complex phenomenon governed by multiple factors. Several attempts have been made to unravel the events of biofilm formation; and, such efforts have provided insights into the mechanisms to target for the therapy. Owing to the fact that the biofilm-state makes the bacterial pathogens significantly resistant to antibiotics, targeting pathogens within biofilm is indeed a lucrative prospect. The available drugs can be repurposed to eradicate the pathogen, and as a result, ease the antimicrobial treatment burden. Biofilm formers and their infections have also been found in plants, livestock, and humans. The advent of novel strategies such as bioinformatics tools in treating, as well as preventing, biofilm formation has gained a great deal of attention. Development of newfangled anti-biofilm agents, such as silver nanoparticles, may be accomplished through omics approaches such as transcriptomics, metabolomics, and proteomics. Nanoparticles' anti-biofilm properties could help to reduce antimicrobial resistance (AMR). This approach may also be integrated for a better understanding of biofilm biology, guided by mechanistic understanding, virtual screening, and machine learning in silico techniques for discovering small molecules in order to inhibit key biofilm regulators. This stimulated research is a rapidly growing field for applicable control measures to prevent biofilm formation. Therefore, the current article discusses the current understanding of biofilm formation, antibiotic resistance mechanisms in bacterial biofilm, and the novel therapeutic strategies to combat biofilm-mediated infections.
Collapse
Affiliation(s)
- Yogesh Dutt
- Department of Microbiology, SRM University, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat 131029, Haryana, India; (Y.D.); (R.D.); (A.V.); (A.G.); (R.P.P.); (V.S.R.)
| | - Ruby Dhiman
- Department of Microbiology, SRM University, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat 131029, Haryana, India; (Y.D.); (R.D.); (A.V.); (A.G.); (R.P.P.); (V.S.R.)
| | - Tanya Singh
- Department of Botany, TPS College, Patliputra University, Patna 800020, Bihar, India;
| | - Arpana Vibhuti
- Department of Microbiology, SRM University, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat 131029, Haryana, India; (Y.D.); (R.D.); (A.V.); (A.G.); (R.P.P.); (V.S.R.)
| | - Archana Gupta
- Department of Microbiology, SRM University, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat 131029, Haryana, India; (Y.D.); (R.D.); (A.V.); (A.G.); (R.P.P.); (V.S.R.)
| | - Ramendra Pati Pandey
- Department of Microbiology, SRM University, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat 131029, Haryana, India; (Y.D.); (R.D.); (A.V.); (A.G.); (R.P.P.); (V.S.R.)
| | - V. Samuel Raj
- Department of Microbiology, SRM University, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat 131029, Haryana, India; (Y.D.); (R.D.); (A.V.); (A.G.); (R.P.P.); (V.S.R.)
| | - Chung-Ming Chang
- Master & Ph.D. Program in Biotechnology Industry, Chang Gung University, No.259, Wenhua 1st Rd., Guishan Dist., Taoyuan City 33302, Taiwan
| | - Anjali Priyadarshini
- Department of Microbiology, SRM University, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat 131029, Haryana, India; (Y.D.); (R.D.); (A.V.); (A.G.); (R.P.P.); (V.S.R.)
| |
Collapse
|
40
|
Beenker WAG, Hoeksma J, den Hertog J. Gregatins, a Group of Related Fungal Secondary Metabolites, Inhibit Aspects of Quorum Sensing in Gram-Negative Bacteria. Front Microbiol 2022; 13:934235. [PMID: 35865924 PMCID: PMC9296082 DOI: 10.3389/fmicb.2022.934235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/02/2022] [Indexed: 12/11/2022] Open
Abstract
Quorum sensing (QS) is a process that regulates gene expression based on cell density. In bacteria, QS facilitates collaboration and controls a large number of pathways, including biofilm formation and virulence factor production, which lead to lower sensitivity to antibiotics and higher toxicity in the host, respectively. Inhibition of QS is a promising strategy to combat bacterial infections. In this study, we tested the potential of secondary metabolites from fungi to inhibit bacterial QS using a library derived from more than ten thousand different fungal strains. We used the reporter bacterium, Chromobacterium violaceum, and identified 39 fungal strains that produced QS inhibitor activity. These strains expressed two QS inhibitors that had been described before and eight QS inhibitors that had not been described before. Further testing for QS inhibitor activity against the opportunistic pathogen Pseudomonas aeruginosa led to the identification of gregatins as an interesting family of compounds with QS inhibitor activity. Although various gregatins inhibited QS in P. aeruginosa, these gregatins did not inhibit virulence factor production and biofilm formation. We conclude that gregatins inhibit some, but not all aspects of QS.
Collapse
Affiliation(s)
- Wouter A. G. Beenker
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Jelmer Hoeksma
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Jeroen den Hertog
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Institute Biology Leiden, Leiden University, Leiden, Netherlands
- *Correspondence: Jeroen den Hertog
| |
Collapse
|
41
|
Darvishi S, Tavakoli S, Kharaziha M, Girault HH, Kaminski CF, Mela I. Advances in the Sensing and Treatment of Wound Biofilms. Angew Chem Int Ed Engl 2022; 61:e202112218. [PMID: 34806284 PMCID: PMC9303468 DOI: 10.1002/anie.202112218] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Indexed: 12/02/2022]
Abstract
Wound biofilms represent a particularly challenging problem in modern medicine. They are increasingly antibiotic resistant and can prevent the healing of chronic wounds. However, current treatment and diagnostic options are hampered by the complexity of the biofilm environment. In this review, we present new chemical avenues in biofilm sensors and new materials to treat wound biofilms, offering promise for better detection, chemical specificity, and biocompatibility. We briefly discuss existing methods for biofilm detection and focus on novel, sensor-based approaches that show promise for early, accurate detection of biofilm formation on wound sites and that can be translated to point-of-care settings. We then discuss technologies inspired by new materials for efficient biofilm eradication. We focus on ultrasound-induced microbubbles and nanomaterials that can both penetrate the biofilm and simultaneously carry active antimicrobials and discuss the benefits of those approaches in comparison to conventional methods.
Collapse
Affiliation(s)
- Sorour Darvishi
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
- Department of Chemistry and Chemical EngineeringÉcole Polytechnique Fédérale de Lausanne1951SionSwitzerland
| | - Shima Tavakoli
- Department of Chemistry-Ångstrom LaboratoryUppsala UniversitySE75121UppsalaSweden
| | - Mahshid Kharaziha
- Department of Materials EngineeringIsfahan University of TechnologyIsfahan84156-83111Iran
| | - Hubert H. Girault
- Department of Chemistry and Chemical EngineeringÉcole Polytechnique Fédérale de Lausanne1951SionSwitzerland
| | - Clemens F. Kaminski
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Ioanna Mela
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
| |
Collapse
|
42
|
Darvishi S, Tavakoli S, Kharaziha M, Girault HH, Kaminski CF, Mela I. Advances in the Sensing and Treatment of Wound Biofilms. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 134:e202112218. [PMID: 38505642 PMCID: PMC10946914 DOI: 10.1002/ange.202112218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Indexed: 03/21/2024]
Abstract
Wound biofilms represent a particularly challenging problem in modern medicine. They are increasingly antibiotic resistant and can prevent the healing of chronic wounds. However, current treatment and diagnostic options are hampered by the complexity of the biofilm environment. In this review, we present new chemical avenues in biofilm sensors and new materials to treat wound biofilms, offering promise for better detection, chemical specificity, and biocompatibility. We briefly discuss existing methods for biofilm detection and focus on novel, sensor-based approaches that show promise for early, accurate detection of biofilm formation on wound sites and that can be translated to point-of-care settings. We then discuss technologies inspired by new materials for efficient biofilm eradication. We focus on ultrasound-induced microbubbles and nanomaterials that can both penetrate the biofilm and simultaneously carry active antimicrobials and discuss the benefits of those approaches in comparison to conventional methods.
Collapse
Affiliation(s)
- Sorour Darvishi
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
- Department of Chemistry and Chemical EngineeringÉcole Polytechnique Fédérale de Lausanne1951SionSwitzerland
| | - Shima Tavakoli
- Department of Chemistry-Ångstrom LaboratoryUppsala UniversitySE75121UppsalaSweden
| | - Mahshid Kharaziha
- Department of Materials EngineeringIsfahan University of TechnologyIsfahan84156-83111Iran
| | - Hubert H. Girault
- Department of Chemistry and Chemical EngineeringÉcole Polytechnique Fédérale de Lausanne1951SionSwitzerland
| | - Clemens F. Kaminski
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Ioanna Mela
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhilippa Fawcett DriveCambridgeCB3 0ASUK
| |
Collapse
|
43
|
Mallick S, Nag M, Lahiri D, Pandit S, Sarkar T, Pati S, Nirmal NP, Edinur HA, Kari ZA, Ahmad Mohd Zain MR, Ray RR. Engineered Nanotechnology: An Effective Therapeutic Platform for the Chronic Cutaneous Wound. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:778. [PMID: 35269266 PMCID: PMC8911807 DOI: 10.3390/nano12050778] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/02/2022] [Accepted: 02/06/2022] [Indexed: 12/27/2022]
Abstract
The healing of chronic wound infections, especially cutaneous wounds, involves a complex cascade of events demanding mutual interaction between immunity and other natural host processes. Wound infections are caused by the consortia of microbial species that keep on proliferating and produce various types of virulence factors that cause the development of chronic infections. The mono- or polymicrobial nature of surface wound infections is best characterized by its ability to form biofilm that renders antimicrobial resistance to commonly administered drugs due to poor biofilm matrix permeability. With an increasing incidence of chronic wound biofilm infections, there is an urgent need for non-conventional antimicrobial approaches, such as developing nanomaterials that have intrinsic antimicrobial-antibiofilm properties modulating the biochemical or biophysical parameters in the wound microenvironment in order to cause disruption and removal of biofilms, such as designing nanomaterials as efficient drug-delivery vehicles carrying antibiotics, bioactive compounds, growth factor antioxidants or stem cells reaching the infection sites and having a distinct mechanism of action in comparison to antibiotics-functionalized nanoparticles (NPs) for better incursion through the biofilm matrix. NPs are thought to act by modulating the microbial colonization and biofilm formation in wounds due to their differential particle size, shape, surface charge and composition through alterations in bacterial cell membrane composition, as well as their conductivity, loss of respiratory activity, generation of reactive oxygen species (ROS), nitrosation of cysteines of proteins, lipid peroxidation, DNA unwinding and modulation of metabolic pathways. For the treatment of chronic wounds, extensive research is ongoing to explore a variety of nanoplatforms, including metallic and nonmetallic NPs, nanofibers and self-accumulating nanocarriers. As the use of the magnetic nanoparticle (MNP)-entrenched pre-designed hydrogel sheet (MPS) is found to enhance wound healing, the bio-nanocomposites consisting of bacterial cellulose and magnetic nanoparticles (magnetite) are now successfully used for the healing of chronic wounds. With the objective of precise targeting, some kinds of "intelligent" nanoparticles are constructed to react according to the required environment, which are later incorporated in the dressings, so that the wound can be treated with nano-impregnated dressing material in situ. For the effective healing of skin wounds, high-expressing, transiently modified stem cells, controlled by nano 3D architectures, have been developed to encourage angiogenesis and tissue regeneration. In order to overcome the challenge of time and dose constraints during drug administration, the approach of combinatorial nano therapy is adopted, whereby AI will help to exploit the full potential of nanomedicine to treat chronic wounds.
Collapse
Affiliation(s)
- Suhasini Mallick
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Nadia 741249, India;
| | - Moupriya Nag
- Department of Biotechnology, University of Engineering & Management, Kolkata 700156, India; (M.N.); (D.L.)
| | - Dibyajit Lahiri
- Department of Biotechnology, University of Engineering & Management, Kolkata 700156, India; (M.N.); (D.L.)
| | - Soumya Pandit
- Department of Life Sciences, Sharda University, Noida 201310, India;
| | - Tanmay Sarkar
- Department of Food Processing Technology, Malda Polytechnic, West Bengal State Council of Technical Education, Government of West Bengal, Malda 732102, India;
| | - Siddhartha Pati
- NatNov Bioscience Private Limited, Balasore 756001, India;
- Skills Innovation & Academic Network (SIAN) Institute, Association for Biodiversity Conservation & Research (ABC), Balasore 756001, India
| | - Nilesh Prakash Nirmal
- Institute of Nutrition, Mahidol University, 999 Phutthamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand;
| | - Hisham Atan Edinur
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia;
| | - Zulhisyam Abdul Kari
- Department of Agricultural Science, Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Jeli 17600, Malaysia
| | | | - Rina Rani Ray
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Nadia 741249, India;
| |
Collapse
|
44
|
Quality Related Safety Evaluation of a South African Traditional Formulation (PHELA®) as Novel Anti-Biofilm Candidate. Molecules 2022; 27:molecules27041219. [PMID: 35209008 PMCID: PMC8877180 DOI: 10.3390/molecules27041219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/04/2022] [Indexed: 11/16/2022] Open
Abstract
A South African traditional formulation, PHELA®, is consumed by the traditional people for severe chest problems with coughing, diarrhea, oral ulcers etc. The present study focused on establishing the anti-infective properties of a safe and standardized poly-herbal formulation through a series of criteria and specifications.
Collapse
|
45
|
Abstract
The opportunity of decreasing the development of biofilm on the implant surface is one of the biggest research problems. It is connected with the existing prevention of microorganism hyperplasia. The application of numerous modifications is concerned with surface treatments leading to minimizing bacterial colonization. In the case of non-use antibacterial therapy, this leads to tissue infection. It can lead to a decreased opportunity to fight infection using antibiotherapy. One way is to decrease the increasing biofilm application which requires a method of modification. These techniques ensure properties like homogeneity or repeatability. The structure and chemical composition are changed with methods like CVD (Chemical Vapor Deposition), PVD (Physical Vapor Deposition), sol–gel, or ALD (Atomic Layer Deposition). Antibacterial properties of metals are connected with their impact on proteins and the nuclear proliferation of fibroblasts, causing improvement in biocompatibility and also growth corrosion resistance, and the decline of biofilm adhesion. The prevention of biofilm with medicines and antibiotics is a crowded-out treatment. Traditional methods of preventing biofilm are based on compounds that kill or inhibit the growth of the microbes but at the same time lead to frequent development of resistance to antibiotics. This review summarizes the current knowledge of reducing and preventing the creation of biofilm.
Collapse
|
46
|
Toale C, Kelly A, Leahy F, Meagher H, Stapleton PJ, Moloney MA, Kavanagh EG. Effect of Pseudomonas colonisation on lower limb venous ulcer healing: a systematic review. J Wound Care 2022; 31:186-192. [PMID: 35148629 DOI: 10.12968/jowc.2022.31.2.186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Pseudomonas aeruginosa is a Gram-negative bacillus that commonly colonises lower limb venous ulcers. Its effects on venous ulcer healing are widely debated. It produces exotoxins and elastase, as well as forming biofilms in hard-to-heal wounds. It is postulated that these virulence factors lead to slower healing times in patients with lower limb venous ulcers colonised with Pseudomonas. This review aimed to summarise the available evidence pertaining to this topic. METHOD A systematic review was performed in August 2019, where the Pubmed, Cochrane and Embase databases were searched for relevant literature according to PRISMA guidelines. Retrospective and prospective studies examining the effect of Pseudomonas colonisation on any measure of ulcer healing were included. RESULTS Some 282 articles were screened, of which seven studies including 491 patients were ultimately included for analysis. Of these, no study demonstrated a significant association between Pseudomonas colonisation and delayed healing of venous ulcers. In five of the seven studies, the effect of Pseudomonas aeruginosa on initial ulcer size at presentation was recorded. CONCLUSION All the studies demonstrated an association between ulcer size and the presence of Pseudomonas aeruginosa. While Pseudomonas aeruginosa may colonise larger ulcers or those with a worse prognosis, no evidence was found to support the hypothesis that this colonisation had a negative impact on lower limb venous ulcer healing.
Collapse
Affiliation(s)
- Conor Toale
- Department of Vascular/Endovascular Surgery, University Hospital Limerick, Ireland
| | - Aisling Kelly
- Department of Vascular/Endovascular Surgery, University Hospital Limerick, Ireland
| | - Fiona Leahy
- Department of Vascular/Endovascular Surgery, University Hospital Limerick, Ireland
| | - Helen Meagher
- Department of Vascular/Endovascular Surgery, University Hospital Limerick, Ireland
| | | | - Michael A Moloney
- Department of Vascular/Endovascular Surgery, University Hospital Limerick, Ireland
| | - Eamon G Kavanagh
- Department of Vascular/Endovascular Surgery, University Hospital Limerick, Ireland.,Department of Microbiology, University Hospital Limerick, Ireland
| |
Collapse
|
47
|
Vilela PB, Mendonça Neto RP, Starling MCVM, da S Martins A, Pires GFF, Souza FAR, Amorim CC. Metagenomic analysis of MWWTP effluent treated via solar photo-Fenton at neutral pH: Effects upon microbial community, priority pathogens, and antibiotic resistance genes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 801:149599. [PMID: 34467925 PMCID: PMC8573595 DOI: 10.1016/j.scitotenv.2021.149599] [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: 06/25/2021] [Revised: 08/07/2021] [Accepted: 08/08/2021] [Indexed: 04/13/2023]
Abstract
The effectiveness of advanced technologies on eliminating antibiotic resistant bacteria (ARB) and resistance genes (ARGs) from wastewaters have been recently investigated. Solar photo-Fenton has been proven effective in combating ARB and ARGs from Municipal Wastewater Treatment Plant effluent (MWWTPE). However, most of these studies have relied solely on cultivable methods to assess ARB removal. This is the first study to investigate the effect of solar photo-Fenton upon ARB and ARGs in MWWTPE by high throughput metagenomic analysis (16S rDNA sequencing and Whole Genome Sequencing). Treatment efficiency upon priority pathogens and resistome profile were also investigated. Solar photo-Fenton (30 mg L-1 of Fe2+ intermittent additions and 50 mg L-1 of H2O2) reached 76-86% removal of main phyla present in MWWTPE. An increase in Proteobacteria abundance was observed after solar photo-Fenton and controls in which H2O2 was present as an oxidant (Fenton, H2O2 only, solar/H2O2). Hence, tolerance mechanisms presented by this group should be further assessed. Solar photo-Fenton achieved complete removal of high priority Staphylococcus and Enterococcus, as well as Klebsiella pneumoniae and Pseudomonas aeruginosa. Substantial reduction of intrinsically multi-drug resistant bacteria was detected. Solar photo-Fenton removed nearly 60% of ARGs associated with sulfonamides, macrolides, and tetracyclines, and complete removal of ARGs related to β-lactams and fluoroquinolones. These results indicate the potential of using solar-enhanced photo-Fenton to limit the spread of antimicrobial resistance, especially in developing tropical countries.
Collapse
Affiliation(s)
- Pâmela B Vilela
- Universidade Federal de Minas Gerais, Escola de Engenharia, Departamento de Engenharia Sanitária e Ambiental, Research Group on Environmental Applications of Advanced Oxidation Processes (GruPOA), Av. Pres. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - Rondon P Mendonça Neto
- Universidade Federal de Minas Gerais, Escola de Engenharia, Departamento de Engenharia Sanitária e Ambiental, Research Group on Environmental Applications of Advanced Oxidation Processes (GruPOA), Av. Pres. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Bioquímica e Imunologia, Pampulha, Belo Horizonte, MG, Brazil
| | - Maria Clara V M Starling
- Universidade Federal de Minas Gerais, Escola de Engenharia, Departamento de Engenharia Sanitária e Ambiental, Research Group on Environmental Applications of Advanced Oxidation Processes (GruPOA), Av. Pres. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - Alessandra da S Martins
- Universidade Federal de Minas Gerais, Escola de Engenharia, Departamento de Engenharia Sanitária e Ambiental, Research Group on Environmental Applications of Advanced Oxidation Processes (GruPOA), Av. Pres. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - Giovanna F F Pires
- Universidade Federal de Minas Gerais, Escola de Engenharia, Departamento de Engenharia Sanitária e Ambiental, Research Group on Environmental Applications of Advanced Oxidation Processes (GruPOA), Av. Pres. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - Felipe A R Souza
- Universidade Federal de Minas Gerais, Escola de Engenharia, Departamento de Engenharia Sanitária e Ambiental, Research Group on Environmental Applications of Advanced Oxidation Processes (GruPOA), Av. Pres. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - Camila C Amorim
- Universidade Federal de Minas Gerais, Escola de Engenharia, Departamento de Engenharia Sanitária e Ambiental, Research Group on Environmental Applications of Advanced Oxidation Processes (GruPOA), Av. Pres. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil.
| |
Collapse
|
48
|
Kim EJ, Hyun JE, Kang YH, Baek SJ, Hwang CY. In vitro antibacterial and antibiofilm effects of cold atmospheric microwave plasma against Pseudomonas aeruginosa causing canine skin and ear infections. Vet Dermatol 2021; 33:29-e10. [PMID: 34747063 DOI: 10.1111/vde.13030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Pseudomonas aeruginosa is an opportunist pathogen that causes purulent inflammation in the skin and in the ears of dogs. Among the various virulence factors of P. aeruginosa, biofilms have been reported to result in antibiotic resistance, leading to therapeutic limitations. Cold atmospheric microwave plasma (CAMP) is known to have a high antimicrobial effect, which causes physical cell wall rupture and DNA damage. HYPOTHESIS/OBJECTIVES The objective of this study was to evaluate the antibacterial and antibiofilm effects of CAMP against planktonic bacteria and the biofilm of P. aeruginosa. METHODS AND MATERIALS The antibacterial effect of CAMP against P. aeruginosa ATCC10145 and clinical isolates (n = 30) was evaluated using the colony count method. We also assessed the effect of CAMP on biofilm of P. aeruginosa ATCC strain by the colony count method, water-soluble tetrazolium salt (WST) assay and confocal laser scanning microscopy (CLSM). RESULTS The complete eradication of P. aeruginosa (ATCC strain and clinical isolates) was achieved within 120 s at 50 W, and clinical isolates required 60 s shorter than the ATCC strain for complete eradication at 50 W. We also confirmed the time-dependent bactericidal effect of CAMP at 50 W against ATCC strain biofilm. CONCLUSIONS AND CLINICAL IMPORTANCE CAMP was effective against both planktonic bacteria and biofilm formation of P. aeruginosa. However, further studies on in vivo efficacy and safety in canine skin and ears are necessary to fully validate its clinical application.
Collapse
Affiliation(s)
- Eun-Joo Kim
- Laboratory of Veterinary Dermatology and the Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Jae-Eun Hyun
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Korea
| | - Yeong-Hun Kang
- Laboratory of Veterinary Dermatology and the Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Seung-Joon Baek
- Laboratory of Signal Transduction, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Cheol-Yong Hwang
- Laboratory of Veterinary Dermatology and the Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| |
Collapse
|
49
|
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
|
50
|
Warrier A, Satyamoorthy K, Murali TS. Quorum-sensing regulation of virulence factors in bacterial biofilm. Future Microbiol 2021; 16:1003-1021. [PMID: 34414776 DOI: 10.2217/fmb-2020-0301] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Chronic polymicrobial wound infections are often characterized by the presence of bacterial biofilms. They show considerable structural and functional heterogeneity, which influences the choice of antimicrobial therapy and wound healing dynamics. The hallmarks of biofilm-associated bacterial infections include elevated antibiotic resistance and extreme pathogenicity. Biofilm helps bacteria to evade the host defense mechanisms and persist longer in the host. Quorum-sensing (QS)-mediated cell signaling primarily regulates biofilm formation in chronic infections and plays a major role in eliciting virulence. This review focuses on the QS mechanisms of two major bacterial pathogens, Staphylococcus aureus and Pseudomonas aeruginosa and explains how they interact in the wound microenvironment to regulate biofilm development and virulence. The review also provides an insight into the treatment modalities aimed at eradicating polymicrobial biofilms. This information will help us develop better diagnostic modalities and devise effective treatment regimens to successfully manage and overcome severe life-threatening bacterial infections.
Collapse
Affiliation(s)
- Anjali Warrier
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kapaettu Satyamoorthy
- Department of Cell & Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Thokur Sreepathy Murali
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India.,Manipal Center for Infectious Diseases (MAC ID), Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|