1
|
Defoirdt T. Resistance to quorum sensing inhibition spreads more slowly during host infection than antibiotic resistance. Gut Microbes 2025; 17:2476582. [PMID: 40066860 PMCID: PMC11901357 DOI: 10.1080/19490976.2025.2476582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2025] Open
Abstract
Antibiotic resistance is a rising problem and new and sustainable strategies to combat bacterial (intestinal) infections are therefore urgently needed. One promising strategy under intense investigation is the inhibition of quorum sensing, bacterial cell-to-cell communication with small molecules. A key question with respect to the application of quorum sensing inhibition is whether it will impose selective pressure for the spread of resistance. It was recently shown that resistance to quorum sensing inhibition will spread more slowly during infection of a host than resistance to traditional antibiotics.
Collapse
Affiliation(s)
- Tom Defoirdt
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Ghent University, Gent, Belgium
| |
Collapse
|
2
|
Leitão MM, Gonçalves ASC, Sousa SF, Borges F, Simões M, Borges A. Two cinnamic acid derivatives as inhibitors of Pseudomonas aeruginosa las and pqs quorum-sensing systems: Impact on biofilm formation and virulence factors. Biomed Pharmacother 2025; 187:118090. [PMID: 40318447 DOI: 10.1016/j.biopha.2025.118090] [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: 02/22/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025] Open
Abstract
INTRODUCTION Quorum sensing (QS) is a bacterial communication mechanism that regulates gene expression, playing a crucial role in various physiological processes. Interfering with this signalling pathway is a promising strategy to control bacterial pathogenicity and virulence. OBJECTIVES This study evaluated the potential of two cinnamic acid derivatives, ferulic and sinapic acids, to inhibit the las and pqs systems in Pseudomonas aeruginosa. Their effects on biofilm architecture, virulence factor production and bacterial motility were also investigated. METHODS Bioreporter strains and bioluminescence-based assays were used to evaluate the modulation of QS-activity by cinnamic acid-type phenolic acids. In addition, in silico docking analysis was performed to validate the binding interactions of the cinnamic acid derivatives with QS-receptors. The biofilm architecture was analysed by optical coherence tomography, and virulence factors production (pyoverdine, pyocyanin, total proteases, lipases, gelatinases and siderophores) and motility were measured by absorbance measurement and plate agar method. RESULTS Ferulic and sinapic acids at 1000 µg mL-1 inhibited the las and pqs systems by 90 % and 80 %, respectively. The N-3-oxododecanoyl-homoserine lactone production was reduced by 70 % (6.25 µg mL-¹). In silico analysis demonstrated that cinnamic acid derivatives exhibited comparable interactions and higher docking scores than reference ligands and inhibitors. Biofilm thickness decreased from 96 µm to 11 µm, and virulence factors and swarming motility were significantly impaired. The comparable anti-QS activity of cinnamic acid derivatives suggests that the additional methoxy group in sinapic acid does not directly contribute to its anti-QS effect. CONCLUSION Ferulic and sinapic acids compromised the biofilm architecture and virulence of P. aeruginosa through QS inhibition.
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, s/n, Porto 4200-465, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, Porto 4200-465, Portugal; CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, Porto 4169-007, Portugal
| | - Ariana S C Gonçalves
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, Porto 4200-465, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, Porto 4200-465, Portugal; Environmental Health Department, Portuguese National Health Institute Doctor Ricardo Jorge, Porto, Portugal
| | - Sérgio F Sousa
- LAQV/REQUIMTE, BioSIM-Department of Biomedicine, Faculty of Medicine, University of Porto, Rua Alameda Prof. Hernâni Monteiro, Porto 4200-319, Portugal
| | - Fernanda Borges
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, Porto 4169-007, Portugal
| | - Manuel Simões
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, Porto 4200-465, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, Porto 4200-465, Portugal; DEQB-Department of Chemical and Biological Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, Porto 4200-465, Portugal
| | - Anabela Borges
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, Porto 4200-465, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, Porto 4200-465, Portugal; DEQB-Department of Chemical and Biological Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, Porto 4200-465, Portugal.
| |
Collapse
|
3
|
Garcia-Maset R, Chu V, Yuen N, Blumgart D, Yoon J, Murray BO, Joseph AA, Rohn JL. Effect of host microenvironment and bacterial lifestyles on antimicrobial sensitivity and implications for susceptibility testing. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:42. [PMID: 40399473 PMCID: PMC12095824 DOI: 10.1038/s44259-025-00113-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/01/2025] [Indexed: 05/23/2025]
Abstract
Bacterial infections remain a major global health issue, with antimicrobial resistance (AMR) worsening the crisis. However, treatment failure can occur even when bacteria show antibiotic susceptibility in diagnostic tests. We explore factors such as phenotypic resilience, bacterial lifestyles such as biofilms, and differences between laboratory tests and real infection sites, highlighting the need for improved platforms to better predict treatment outcomes, and reviewing emerging technologies aimed at improving susceptibility testing.
Collapse
Affiliation(s)
- Ramon Garcia-Maset
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK.
| | - Victoria Chu
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Nicholas Yuen
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Dalia Blumgart
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Jenny Yoon
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Benjamin O Murray
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Amelia A Joseph
- Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Jennifer L Rohn
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
4
|
Conaway A, Mould DL, Todorovic I, Hogan DA. Loss of LasR function leads to decreased repression of Pseudomonas aeruginosa PhoB activity at physiological phosphate concentrations. J Bacteriol 2025:e0018924. [PMID: 40366151 DOI: 10.1128/jb.00189-24] [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: 04/30/2024] [Accepted: 03/07/2025] [Indexed: 05/15/2025] Open
Abstract
The Pseudomonas aeruginosa LasR transcription factor plays a role in quorum sensing (QS) across phylogenetically distinct lineages. However, isolates with loss-of-function mutations in lasR (LasR- strains) are commonly found in diverse settings, including infections where they are associated with worse clinical outcomes. In LasR- strains, the LasR-regulated transcription factor RhlR can also be stimulated by the activity of the two-component system PhoR-PhoB in low-inorganic phosphate (Pi) conditions. Here, we demonstrate a novel link between LasR and PhoB in which the absence of LasR increases PhoB activity at physiological Pi concentrations and increases the Pi concentration necessary for PhoB inhibition. PhoB activity was also less sensitive to repression by Pi in mutants lacking different QS regulators (RhlR and PqsR) and in mutants lacking genes required for QS-induced phenazine production, suggesting that decreased phenazine production is one reason for increased PhoB activity in LasR- strains. In addition, the CbrA-CbrB two-component system, which can be more active in LasR- strains, was necessary for increased PhoB activity in LasR- strains, and loss of the CbrA-CbrB-controlled translational repressor Crc was sufficient to activate PhoB in LasR+ P. aeruginosa. Phenazines and CbrA-CbrB affected PhoB activity independently. The ∆lasR mutant also had PhoB-dependent growth advantages in the Pi-deplete medium and increased virulence-associated gene expression at physiological Pi, in part through reactivation of QS. This work suggests PhoR-PhoB activity may contribute to the fitness and virulence of LasR- P. aeruginosa and subsequent clinical outcomes.IMPORTANCELoss-of-function mutations in the gene encoding the Pseudomonas aeruginosa quorum sensing (QS) regulator LasR occur frequently and are associated with worse clinical outcomes. We have found that LasR- P. aeruginosa have elevated PhoB activity at physiological concentrations of inorganic phosphate (Pi). PhoB activity promotes Pi acquisition as well as the expression of QS and virulence-associated genes. Previous work has shown that PhoB induces RhlR, another QS regulator, in a LasR- mutant in low-Pi conditions. Here, we demonstrate a novel relationship wherein LasR represses PhoB activity through the production of phenazines and Crc-mediated translational repression. This work suggests PhoB activity may contribute to the increased virulence of LasR- P. aeruginosa.
Collapse
Affiliation(s)
- Amy Conaway
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Dallas L Mould
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Igor Todorovic
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Deborah A Hogan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
5
|
Zeng J, Iizaka Y, Ouchi Y, Otsuki K, Kikuchi T, Li W, Anzai Y. Inhibitory effects of reumycin produced by Streptomyces sp. TPMA0082 on virulence factors of Pseudomonas aeruginosa. J Nat Med 2025; 79:608-620. [PMID: 40195206 DOI: 10.1007/s11418-025-01902-w] [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: 12/18/2024] [Accepted: 03/19/2025] [Indexed: 04/09/2025]
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogen that causes a wide range of infections. The increasing multidrug-resistance of P. aeruginosa poses a critical challenge for medical care. P. aeruginosa employs virulence factors and biofilms to establish infections in humans and protect itself from environmental stress or antibiotics. These factors are regulated by a quorum sensing mechanism involving multiple regulatory systems that act interdependently through signaling molecules. Therefore, interference with quorum sensing systems can suppress the pathogenicity of P. aeruginosa. In this study, quorum sensing inhibitors were explored from secondary metabolites derived from 111 strains of actinomycetes by targeting the las system, which is thought to be upstream of the quorum sensing cascade in P. aeruginosa. As a result, reumycin was isolated from the culture broth of Streptomyces sp. TPMA0082. Reumycin, a molecule containing a pyrimidotriazine ring, inhibited the binding of the autoinducer to the LasR receptor in the las system, thereby suppressing the production of P. aeruginosa virulence factors, including pyocyanin, rhamnolipids, elastase, motility, and biofilms, without affecting bacterial growth. Toxoflavin, a reumycin derivative with a methyl group at the N1 position, exhibited strong antibacterial activity. Fervenulin, a reumycin derivative with a methyl group at the N8 position, had a negative impact on the logarithmic growth phase of the bacteria and exhibited lower inhibitory activity against virulence factor production compared to reumycin. These findings suggest that the position and number of methyl groups attached to the pyrimidotriazine structure significantly influence its biological activity, exerting distinct effects on quorum sensing inhibition and antibacterial activity.
Collapse
Affiliation(s)
- Jiahao Zeng
- Faculty of Pharmaceutical Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan
| | - Yohei Iizaka
- Faculty of Pharmaceutical Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan.
| | - Yasuhiro Ouchi
- Faculty of Pharmaceutical Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan
| | - Kouharu Otsuki
- Faculty of Pharmaceutical Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan
| | - Takashi Kikuchi
- Faculty of Pharmaceutical Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan
| | - Wei Li
- Faculty of Pharmaceutical Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan
| | - Yojiro Anzai
- Faculty of Pharmaceutical Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan
| |
Collapse
|
6
|
Suresh D, Yu TT, Kuppusamy R, Sabir S, Das T, Black DS, Willcox MDP, Kumar N. Novel cationic dihydropyrrol-2-one compounds as antimicrobial agents and quorum sensing inhibitors. Bioorg Med Chem 2025; 122:118137. [PMID: 40058275 DOI: 10.1016/j.bmc.2025.118137] [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: 12/20/2024] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Antimicrobial resistance has grown to become a global crisis consistently participating in the death of millions worldwide and accumulating costs on healthcare. Quorum sensing inhibition is a new alternative antimicrobial strategy that has been gaining attention due to its ability to suppress the resistance of Pseudomonas aeruginosa (PA). This approach shows great potential in overcoming bacterial resistance and could provide a much needed substitute to conventional antibiotics in the future. PA has 3 main quorum sensing systems of which the Las system has been identified to be the most viable therapeutic target. In this study, we report the synthesis of a library of novel broad-spectrum quorum sensing inhibitors from the dihydropyrrol-2-one scaffold to form urea and imidazolium analogues. Molecular docking was performed in parallel to synthesis to aid design. It also confirmed that the molecules comfortably occupy the ligand binding domain in addition to potential key interactions commonly present in LasR inhibitors. As predicted, these compounds displayed low bactericidal effects against P. aeruginosa with most compounds exhibiting MIC of >250 μM, while maintaining moderate activity towards Escherichia coli with the most potent compound having an MIC of 32 μM. The greatest bactericidal effects were present on Staphylococcus aureus with the thiourea based molecule 10c showed the highest antibacterial activity with MIC of 16 µM. Furthermore, several molecules displayed highly potent quorum sensing inhibitory activity with compounds 10g and 9e both demonstrating over 70 % inhibition respectively of the LasR system at 16 µM. These compounds also expressed inhibition of pyocyanin within P. aeruginosa and haemolytic assay indicates a low level of cell lysis and hence low toxicity of the compounds, further demonstrating the potential of these novel compounds.
Collapse
Affiliation(s)
- Dittu Suresh
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Tsz Tin Yu
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Rajesh Kuppusamy
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia; School of Optometry and Vision Science, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Shekh Sabir
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | | | - David StC Black
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Mark D P Willcox
- School of Optometry and Vision Science, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Naresh Kumar
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
7
|
Dal A, Hekimoğlu R, Sümbül B, Balıbey FB, Gökçe ZÖ, Akbulut E, Elbay A. Quorum-Sensing inhibition by furanone compounds and therapeutic effects on Pseudomonas aeruginosa keratitis rabbit model. BMC Ophthalmol 2025; 25:231. [PMID: 40264027 PMCID: PMC12013088 DOI: 10.1186/s12886-025-04061-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND To investigate Quorum-Sensing inhibition by furanone compounds in Pseudomonas aeruginosa keratitis rabbit model. METHODS Thirty adult New Zealand White rabbits were used. Anesthetized rabbits were intrastromally injected with Pseudomonas aeruginosa (P. aeruginosa). The rabbits were divided into six groups: the control group (infected only with P. aeruginosa), group A (50 mg/mL ceftazidime), group B (0.1 mg/mL furanone), group C (0.2 mg/mL furanone), group D (0.3 mg/mL furanone), and group E (20% dimethyl sulfoxide). One drop of the treatment was applied every hour for 3 days, starting 1-h post-inoculation. Rabbits were then sacrificed, and corneas were analysed clinically, microbiologically, histologically, and biochemically. One-way analysis of variance was used for the mean comparison of independent groups. The Least Significant Difference method was used as a post-hoc test for pairwise comparisons. RESULTS In all evaluations, the antibiotic group (group A) showed the best therapeutic response. The slit-lamp examination score of group C was significantly lowered than those compared of to the control (p = 0.009) and E groups (p = 0.014). Histological evaluation showed that inflammation is decreased in groups B, C, and D. Levels of cyclooxygenase-2 (COX-2), superoxide dismutase-1, and reactive oxygen species (ROS) were lowest in the antibiotic-treated group, whereas the highest levels were detected in the control group. Notably, the COX-2 levels in group B and ROS levels in groups B and C were significantly lower than in control group. (p = 0.045, p = 0.039 and p = 0.045, respectively). CONCLUSION Furanone compounds may have minimal therapeutic effects against Pseudomonas keratitis. Its therapeutic effect has not been observed to be sufficient compared with that of antibiotics. Further studies are needed to investigate their protective effects and mechanisms.
Collapse
Affiliation(s)
- Aleyna Dal
- Faculty of Medicine, Bezmialem Vakıf University, Istanbul, Turkey
| | - Rümeysa Hekimoğlu
- Faculty of Medicine, Department of Histology and Embryology, Bezmialem Vakıf University, Istanbul, Turkey
| | - Bilge Sümbül
- Faculty of Medicine, Department of Clinical Microbiology, Bezmialem Vakıf University, Istanbul, Turkey
| | - Fatmanur Babalı Balıbey
- Faculty of Medicine, Department of Medical Biochemistry, Bezmialem Vakıf University, Istanbul, Turkey
| | - Zeynep Özman Gökçe
- Department of Medical Biochemistry, Institute of Health Sciences, Bezmialem Vakıf University, Istanbul, Turkey
| | - Ersin Akbulut
- Faculty of Medicine, Department of Ophthalmology, Bezmialem Vakıf University, Istanbul, 34093, Turkey
| | - Ahmet Elbay
- Faculty of Medicine, Department of Ophthalmology, Bezmialem Vakıf University, Istanbul, 34093, Turkey.
| |
Collapse
|
8
|
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
|
9
|
Portaccio L, Vergine M, De Pascali M, De Bellis L, Luvisi A. Diffusible Signal Factors and Xylella fastidiosa: A Crucial Mechanism Yet to Be Revealed. BIOLOGY 2025; 14:303. [PMID: 40136559 PMCID: PMC11939919 DOI: 10.3390/biology14030303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/14/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025]
Abstract
Xylella fastidiosa (Xf) is a xylem-limited Gram-negative phytopathogen responsible for severe plant diseases globally. Colonization and dissemination on host plants are regulated primarily by diffusible signal factors (DSFs) and quorum sensing (QS) molecules regulating biofilm formation, motility, and virulence factor synthesis. DSFs play a critical role in the transition of bacteria from adhesion to dispersal phases, influencing plant infection and transmission by vector. Because of Xf's host range (over 550 plant species), effective containment strategies are highly demanded. In this review, we discuss the molecular mechanism of DSF-mediated signalling in Xf, especially concerning its role in pathogenicity and adaptation. Moreover, we shed light on innovative approaches to manage Xf, including quorum-quenching (QQ) strategies and transgenic plants targeted to disrupt QS pathways. Improved knowledge of DSF interactions with host plants and bacterial communities could provide an entry point for novel, sustainable disease control strategies to decrease Xf's agricultural and ecological impact.
Collapse
Affiliation(s)
- Letizia Portaccio
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.P.); (M.D.P.); (L.D.B.); (A.L.)
| | - Marzia Vergine
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.P.); (M.D.P.); (L.D.B.); (A.L.)
| | - Mariarosaria De Pascali
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.P.); (M.D.P.); (L.D.B.); (A.L.)
- National Biodiversity Future Center, 90133 Palermo, Italy
| | - Luigi De Bellis
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.P.); (M.D.P.); (L.D.B.); (A.L.)
- National Biodiversity Future Center, 90133 Palermo, Italy
| | - Andrea Luvisi
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.P.); (M.D.P.); (L.D.B.); (A.L.)
| |
Collapse
|
10
|
Tu Y, Li H, Huo J, Gou L, Wen X, Yu X, Zhang X, Zeng J, Li Y. Disrupting the bacterial language: quorum quenching and its applications. Crit Rev Microbiol 2025:1-15. [PMID: 39973173 DOI: 10.1080/1040841x.2025.2466472] [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: 12/28/2023] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/21/2025]
Abstract
Quorum sensing (QS) is a bacterial communication method closely linked with population density and regulates biofilm formation and the secretion of virulence factors through the release, recognition, and prompt response to small molecule signals. At low cell density, each bacterium produces a low concentration of QS signals that diffuse or are actively transported into the external environment. The accumulated QS signals in the external environment reach a threshold concentration when the bacterial population attains a certain density, enabling effective recognition and interaction of bacterial QS signals with their receptors. This leads to coordinated gene expression and various biological activities across the bacterial population. Targeting the QS system presents a promising strategy to hinder biofilm formation and virulence factor secretion, providing a potential approach to control bacterial growth and reproduction. This study aims to analyze the intercellular mechanisms of quorum quenching (QQ), which focuses on disrupting bacterial signal molecules to keep their concentration below the threshold and preventing the expression of specific pathogenic factors. The applications of QQ in different fields are also reviewed, underscoring its potential as a novel treatment for bacterial infections.
Collapse
Affiliation(s)
- Yeting Tu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hanyu Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiachen Huo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lichen Gou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiang Wen
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaomin Yu
- Department of Emergency Medicine, West China Hospital, Sichuan University/Nursing Key Laboratory of Sichuan Province, West China School of Nursing, Sichuan University, Chengdu, China
| | - Xiaorui Zhang
- West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jumei Zeng
- West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuqing Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Cruz RL, Freeman TS, Asfahl KL, Smalley NE, Dandekar AA. RhlR-mediated cooperation in cystic fibrosis-adapted isolates of Pseudomonas aeruginosa. J Bacteriol 2025; 207:e0034424. [PMID: 39670758 PMCID: PMC11784195 DOI: 10.1128/jb.00344-24] [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: 08/20/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024] Open
Abstract
Pseudomonas aeruginosa uses quorum sensing (QS) to regulate the expression of dozens of genes, many of which encode shared products, called "public goods." P. aeruginosa possesses two complete acyl-homoserine lactone (AHL) QS circuits: the LasR-I and RhlR-I systems. Canonically, these systems are hierarchically organized: RhlR-I activity depends on LasR-I activation. However, in contrast to laboratory strains, isolates from people with cystic fibrosis can engage in AHL QS using only the transcription factor RhlR. In these isolates, RhlR regulates AHL QS and the production of secreted public goods, such as the exoprotease elastase, which are accessible to both producing and non-producing cells. When P. aeruginosa strains that use LasR to regulate elastase production are grown on casein as the sole carbon and energy source, LasR-null mutant "cheaters" commonly arise in populations due to a selective growth advantage. We asked if these social dynamics might differ in "RhlR cooperators": populations that use RhlR, not LasR, to regulate public goods. We passaged RhlR cooperators from several genetic backgrounds in casein broth. We found that cheaters emerged among most RhlR cooperators. However, in one isolate background, E90, RhlR-null mutants were dramatically outcompeted by RhlR cooperators. In this background, the mechanism by which RhlR mutants are outcompeted by RhlR cooperators is AHL-dependent and occurs in stationary phase but is not the same as previously described "policing" mechanisms. Our data suggest that cheating, or the lack thereof, does not explain the lack of RhlR mutants observed in most infection environments.IMPORTANCEQuorum sensing (QS) mutants arise in a variety of populations of bacteria, but mutants of the gene encoding the transcription factor RhlR in Pseudomonas aeruginosa appear to be infrequent. Our work provides insight on the mechanisms through which RhlR-mediated cooperation is maintained in a LasR-null population of P. aeruginosa. Characterizing the selective pressure(s) that disfavor mutations from occurring in RhlR may enhance our understanding of P. aeruginosa evolution in chronic infections and potentially guide the development of therapeutics targeting the RhlR-I QS circuit.
Collapse
Affiliation(s)
- Renae L. Cruz
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Tiia S. Freeman
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Kyle L. Asfahl
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Nicole E. Smalley
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Ajai A. Dandekar
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
12
|
Matheus GG, Chamoun MN, Khosrotehrani K, Sivakumaran Y, Wells TJ. Understanding the pathophysiology of Pseudomonas aeruginosa colonization as a guide for future treatment for chronic leg ulcers. BURNS & TRAUMA 2025; 13:tkae083. [PMID: 39830194 PMCID: PMC11741523 DOI: 10.1093/burnst/tkae083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025]
Abstract
Chronic leg wounds represent a major burden of disease worldwide, costing health care systems billions of dollars each year. Aside from the financial implications, they also impose a significant physical and psychosocial burden on the patient, their relatives and/or carers, and the community. Whilst measures such as maintenance of wound hygiene, debridement, dressings and compression are the current standard of care, complete healing is not always achievable and ulcer recurrence is common. Thus, there is still a gap to breach in terms of understanding the intricate pathophysiology of chronic wounds and the role this plays on treatment and management. Pseudomonas aeruginosa has been linked to poor wound healing, with the pathogen being frequently isolated from chronic leg ulcers. Characterized by its multi-drug resistance, targeting P. aeruginosa requires the development of novel therapeutic options. Thus, the aim of this literature review is to describe the pathophysiology of P. aeruginosa in chronic leg ulcers and discuss novel treatment strategies. Here, we describe the key molecular mechanisms driving the observed clinical effect of P. aeruginosa on wounds and discuss novel strategies of molecular targeting of this common bacteria, establishing new approaches that could benefit patients with chronic hard to heal wounds.
Collapse
Affiliation(s)
- Gabriela Gonzalez Matheus
- Frazer Institute, The University of Queensland, Brisbane, Australia
- Department of Dermatology, Princess Alexandra Hospital, Brisbane, Australia
| | | | - Kiarash Khosrotehrani
- Frazer Institute, The University of Queensland, Brisbane, Australia
- Department of Dermatology, Princess Alexandra Hospital, Brisbane, Australia
| | - Yogeesan Sivakumaran
- Department of Vascular Surgery, Princess Alexandra Hospital, Brisbane, Australia
| | - Timothy J Wells
- Frazer Institute, The University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
| |
Collapse
|
13
|
Pleguezuelos-Manzano C, Beenker WAG, van Son GJF, Begthel H, Amatngalim GD, Beekman JM, Clevers H, den Hertog J. Dual RNA sequencing of a co-culture model of Pseudomonas aeruginosa and human 2D upper airway organoids. Sci Rep 2025; 15:2222. [PMID: 39824906 PMCID: PMC11742674 DOI: 10.1038/s41598-024-82500-w] [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: 04/04/2024] [Accepted: 12/05/2024] [Indexed: 01/20/2025] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative bacterium that is notorious for airway infections in cystic fibrosis (CF) subjects. Bacterial quorum sensing (QS) coordinates virulence factor expression and biofilm formation at population level. Better understanding of QS in the bacterium-host interaction is required. Here, we set up a new P. aeruginosa infection model, using 2D upper airway nasal organoids that were derived from 3D organoids. Using dual RNA-sequencing, we dissected the interaction between organoid epithelial cells and WT or QS-mutant P. aeruginosa strains. Since only a single healthy individual and a single CF subject were used as donors for the organoids, conclusions about CF-specific effects could not be deduced. However, P. aeruginosa induced epithelial inflammation, whereas QS signaling did not affect the epithelial airway cells. Conversely, the epithelium influenced infection-related processes of P. aeruginosa, including QS-mediated regulation. Comparison of our model with samples from the airways of CF subjects indicated that our model recapitulates important aspects of infection in vivo. Hence, the 2D airway organoid infection model is relevant and may help to reduce the future burden of P. aeruginosa infections in CF.
Collapse
Affiliation(s)
- Cayetano Pleguezuelos-Manzano
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center, Utrecht, The Netherlands
| | - Wouter A G Beenker
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gijs J F van Son
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center, Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Harry Begthel
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center, Utrecht, The Netherlands
| | - Gimano D Amatngalim
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Member of ERN-LUNG, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jeffrey M Beekman
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Member of ERN-LUNG, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Centre for Living Technologies, Eindhoven-Wageningen-Utrecht Alliance, Utrecht, The Netherlands
| | - Hans Clevers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands.
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center, Utrecht, The Netherlands.
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, 4070, Basel, Switzerland.
| | - Jeroen den Hertog
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands.
- Institute Biology Leiden, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
14
|
Lima EMF, de Almeida FA, Pinto UM. Exploring the antivirulence potential of phenolic compounds to inhibit quorum sensing in Pseudomonas aeruginosa. World J Microbiol Biotechnol 2025; 41:32. [PMID: 39794611 DOI: 10.1007/s11274-025-04255-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
Bacteria coordinate gene expression in a cell density-dependent manner in a communication process called quorum sensing (QS). The expression of virulence factors, biofilm formation and enzyme production are QS-regulated phenotypes that can interfere in human health. Due to this importance, there is great interest in inhibiting QS, comprising an anti-virulence strategy. This work aimed to evaluate the effect of selected phenolic compounds on the inhibition of QS-regulated phenotypes in Pseudomonas aeruginosa PAO1, using concentrations that do not interfere in bacterial growth. This is one of the main premises for studying the effect of compounds on QS. Firstly, an in-silico study with the LasR and RhlR proteins of P. aeruginosa by molecular docking of 82 phenolic compounds was performed. Then, a screening with 13 selected phenolic compounds was performed, using biosensor strains P. aeruginosa lasB-gfp and P. aeruginosa rhlA-gfp, which emit fluorescence when the QS system is activated. From this assay, eight compounds were selected and evaluated for inhibition of pyocyanin, rhamnolipids, proteases, elastase, and motility. The compounds variably inhibited the evaluated virulence factors. The greatest inhibitions were observed for swarming motility, achieving inhibition rates of up to 50% for baicalein (500 µM) and curcumin (50 µM). Notably, curcumin showed satisfactory inhibition for all phenotypes even at lower concentrations (12.5 to 50 µM) compared to the other compounds (125 to 500 µM). Four compounds - rosmarinic acid, baicalein, curcumin, and resveratrol - were finally tested against biofilm formation observed by optical microscopy. This study demonstrated that phenolic compounds exhibit strong in silico binding to P. aeruginosa LasR and RhlR proteins and variably inhibit QS-regulated phenotypes in vitro. Although no biofilm inhibition was observed, future studies combining compounds and exploring molecular mechanisms are recommended. These findings highlight the biotechnological potential of phenolic compounds for future applications in the food, clinical, and pharmaceutical fields.
Collapse
Affiliation(s)
- Emília Maria França Lima
- Food Research Center (FoRC), Laboratory of Food Microbiology, Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Felipe Alves de Almeida
- Department of Microbiology, Institute of Biotechnology Applied to Agriculture (BIOAGRO), Federal University of Viçosa (UFV), Viçosa, MG, Brazil
| | - Uelinton Manoel Pinto
- Food Research Center (FoRC), Laboratory of Food Microbiology, Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil.
| |
Collapse
|
15
|
Colautti J, Kelly SD, Whitney JC. Specialized killing across the domains of life by the type VI secretion systems of Pseudomonas aeruginosa. Biochem J 2025; 482:1-15. [PMID: 39774785 DOI: 10.1042/bcj20230240] [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: 08/09/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025]
Abstract
Type VI secretion systems (T6SSs) are widespread bacterial protein secretion machines that inject toxic effector proteins into nearby cells, thus facilitating both bacterial competition and virulence. Pseudomonas aeruginosa encodes three evolutionarily distinct T6SSs that each export a unique repertoire of effectors. Owing to its genetic tractability, P. aeruginosa has served as a model organism for molecular studies of the T6SS. However, P. aeruginosa is also an opportunistic pathogen and ubiquitous environmental organism that thrives in a wide range of habitats. Consequently, studies of its T6SSs have provided insight into the role these systems play in the diverse lifestyles of this species. In this review, we discuss recent advances in understanding the regulation and toxin repertoire of each of the three P. aeruginosa T6SSs. We argue that these T6SSs serve distinct physiological functions; whereas one system is a dedicated defensive weapon for interbacterial antagonism, the other two T6SSs appear to function primarily during infection. We find support for this model in examining the signalling pathways that control the expression of each T6SS and co-ordinate the activity of these systems with other P. aeruginosa behaviours. Furthermore, we discuss the effector repertoires of each T6SS and connect the mechanisms by which these effectors kill target cells to the ecological conditions under which their respective systems are activated. Understanding the T6SSs of P. aeruginosa in the context of this organism's diverse lifestyles will provide insight into the physiological roles these secretion systems play in this remarkably adaptable bacterium.
Collapse
Affiliation(s)
- Jake Colautti
- Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, L8S 4K1, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Steven D Kelly
- Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, L8S 4K1, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - John C Whitney
- Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, L8S 4K1, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8S 4K1, Canada
- David Braley Center for Antibiotic Discovery, McMaster University, Hamilton, ON, L8S 4K1, Canada
| |
Collapse
|
16
|
Bayatli N, Malkawi AS, Malkawi A, Khaled K, Alrabadi N, Ovenseri AC, Alhajj L, Al Sarayrih L, Elnefaily SE. Impact of biofilms on healthcare settings and management strategies. REVIEWS AND RESEARCH IN MEDICAL MICROBIOLOGY 2024. [DOI: 10.1097/mrm.0000000000000425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/12/2024] [Indexed: 04/02/2025]
Abstract
The formation of biofilms on the surfaces of biomaterials in clinical settings is becoming more of a concern, especially with the rise of multidrug-resistant bacteria. They have contributed to high mortality and a major cost burden on healthcare systems. Obstacles related to biofilms have been complicated with the presence of very resistant bacterial strains to antimicrobial drugs, necessitating the development of alternative pathways to treat biofilm-related infections in addition to traditional antibiotics. So far, inhibitors that combat the formation of biofilms and the development of modified biomaterials for the manufacture of medical devices have been proposed as approaches to prevent biofilm formation in clinical practice settings. The self-produced extracellular polymeric substances that function as a protective shield, inhibiting antimicrobial penetration, are a key component of biofilms. Biofilms’ impact on medical settings, healthcare, and the economy as well as a brief description of stages involved in their development are discussed here. Furthermore, this review elucidates the two primary categories of biofilm management: preventing the formation of biofilms by inhibiting bacterial initial attachment and removing biofilms that have already formed. Preventive antibiofilm methods discussed in this review involve modifying the physical and chemical characteristics of biomaterials. In addition, removing biofilms using efficient physical and biomedical approaches and by interfering with the quorum-sensing system, which is essential for biofilm formation, are covered here. Moreover, several relevant examples of each method indicated for biofilm management are highlighted. Lastly, the ongoing progress in the field of biofilm research may reveal additional strategies for future biofilm management.
Collapse
Affiliation(s)
- Nur Bayatli
- Faculty of Pharmacy, Cyprus Health and Social Sciences University, Kutlu Adali Blv, Morphou (Guzelyurt)
| | - Ahmad Saleh Malkawi
- Faculty of Pharmacy, Cyprus International University, Nicosia, Cyprus
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Isra University, Queen Alya Airport Street, Amman
| | - Azhar Malkawi
- Department of Pharmacology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Khaled Khaled
- Faculty of Pharmacy, Cyprus Health and Social Sciences University, Kutlu Adali Blv, Morphou (Guzelyurt)
- Faculty of Pharmacy, Cyprus International University, Nicosia, Cyprus
| | - Nasr Alrabadi
- Department of Pharmacology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | | | - Lara Alhajj
- Faculty of Pharmacy, Cyprus International University, Nicosia, Cyprus
| | - Lina Al Sarayrih
- Faculty of Pharmacy, Cyprus International University, Nicosia, Cyprus
| | | |
Collapse
|
17
|
Ziegert Z, Dietz M, Hill M, McBride M, Painter E, Elias MH, Staley C. Targeting quorum sensing for manipulation of commensal microbiota. BMC Biotechnol 2024; 24:106. [PMID: 39696328 PMCID: PMC11653937 DOI: 10.1186/s12896-024-00937-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024] Open
Abstract
Bacteria communicate through the accumulation of autoinducer (AI) molecules that regulate gene expression at critical densities in a process called quorum sensing (QS). Extensive work using simple systems and single strains of bacteria have revealed a role for QS in the regulation of virulence factors and biofilm formation; however, less is known about QS dynamics among communities, especially in vivo. In this review, we summarize the diversity of QS signals as well as their ability to influence "non-target" behaviors among species that have receptors but not synthases for those signals. We highlight host-microbe interactions facilitated by QS and describe cross-talk between QS and the mammalian endocrine and immune systems, as well as host surveillance of QS. Further, we describe emerging evidence for the role of QS in non-infectious, chronic, microbially associated diseases including inflammatory bowel diseases and cancers. Finally, we describe potential therapeutic approaches that involve leveraging QS signals as well as quorum quenching approaches to block signaling in vivo to mitigate deleterious consequences to the host. Ultimately, QS offers a previously underexplored target that may be leveraged for precision modification of the microbiota without deleterious bactericidal consequences.
Collapse
Affiliation(s)
- Zachary Ziegert
- Division of Basic & Translational Research, Department of Surgery, University of Minnesota Medical School, 420 Delaware St, SE MMC 195, Minneapolis, MN, 55455, USA
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA
| | - Matthew Dietz
- Division of Basic & Translational Research, Department of Surgery, University of Minnesota Medical School, 420 Delaware St, SE MMC 195, Minneapolis, MN, 55455, USA
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA
| | - Max Hill
- Division of Basic & Translational Research, Department of Surgery, University of Minnesota Medical School, 420 Delaware St, SE MMC 195, Minneapolis, MN, 55455, USA
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA
| | - Marjais McBride
- Division of Basic & Translational Research, Department of Surgery, University of Minnesota Medical School, 420 Delaware St, SE MMC 195, Minneapolis, MN, 55455, USA
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA
| | - Elizabeth Painter
- Division of Basic & Translational Research, Department of Surgery, University of Minnesota Medical School, 420 Delaware St, SE MMC 195, Minneapolis, MN, 55455, USA
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA
| | - Mikael H Elias
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, St. Paul, MN, 55108, USA
| | - Christopher Staley
- Division of Basic & Translational Research, Department of Surgery, University of Minnesota Medical School, 420 Delaware St, SE MMC 195, Minneapolis, MN, 55455, USA.
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA.
| |
Collapse
|
18
|
Chu X, Yang Q. Regulatory Mechanisms and Physiological Impacts of Quorum Sensing in Gram-Negative Bacteria. Infect Drug Resist 2024; 17:5395-5410. [PMID: 39654694 PMCID: PMC11626961 DOI: 10.2147/idr.s485388] [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: 08/09/2024] [Accepted: 11/26/2024] [Indexed: 12/12/2024] Open
Abstract
The Quorum sensing (QS) system is a widely existing communication mechanism, which regulates bacterial community behaviors and the expression of specific genes. The most common pathogenic bacteria in clinical infections are gram-negative bacteria, and QS plays an important regulatory role in the production of virulence factors and development of antibiotic resistance. This article reviews the QS systems of gram-negative bacteria and provides an overview of how they regulate their physiological functions.
Collapse
Affiliation(s)
- Xiaobing Chu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Qiwen Yang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
- Key Laboratory of Pathogen Infection Prevention and Control, Peking Union Medical College, Ministry of Education, Beijing, People’s Republic of China
| |
Collapse
|
19
|
Mohammed EZ, El-Dydamony NM, Taha EA, Taha MN, Mehany ABM, Abdel Aziz HA, Abd El-Aleam RH. Design, synthesis, and molecular dynamic simulations of some novel benzo[d]thiazoles with anti-virulence activity against Pseudomonas aeruginosa. Eur J Med Chem 2024; 279:116880. [PMID: 39303517 DOI: 10.1016/j.ejmech.2024.116880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Inhibition of quorum sensing (QS) is an impending approach for targeting bacterial infection. Fourteen benzo[d]thiazole and 2-pyrazolo[1,5-a]pyrimidin-3-yl)benzo[d]thiazoles analogues were designed and synthesized as promising LasR antagonists with QS inhibition activity. Among the investigated compounds, compounds 3c, 3e, and 8d exhibited the highest percentage inhibition in biofilm formation (77 %, 63.9 %, 69.4 %), pyocyanin production (74.6 %, 64.9, 69.4 %), and rhamnolipids production (58.5 %, 51 %, 54.3 %) in P. aeruginosa, respectively. Additionally, compounds 3c, 3e and 8d achieved IC50 values against Las R equal 1.37 ± 0.35, 1.55 ± 0.24, 1.1 ± 0.15 μM respectively. Also, molecular docking of the target compounds into the LasR binding site co-crystalized "odDHL" revealed their binding with the essential residues for protein inhibition. Additionally, molecular dynamics simulation (MDS) experiments over 200 ns of compound 3c showed its ability to interact with the LasR binding site with dissociation of the protein's dimer confirming its action as a LasR antagonist. The obtained findings inspire further investigation for benzo[d]thiazole and 2-pyrazolo[1,5-a]pyrimidin-3-yl)benzo[d]thiazoles aiming to design and synthesize more potential QS inhibitors.
Collapse
Affiliation(s)
- Esraa Z Mohammed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, October 6 University, Giza, 12585, Egypt.
| | - Nehad M El-Dydamony
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Egypt
| | - Enas A Taha
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, October 6 University, Giza, 12585, Egypt
| | - Mostafa N Taha
- Microbiology and Immunology Department, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| | - Ahmed B M Mehany
- Zoology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Hatem A Abdel Aziz
- Applied Organic Chemistry Department, National Research Centre, Dokki, Giza, P.O.Box 12622, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria, 21648, Egypt.
| | - Rehab H Abd El-Aleam
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information MTI, Cairo, 11571, Egypt
| |
Collapse
|
20
|
Schlichter Kadosh Y, Muthuraman S, Nisaa K, Ben-Zvi A, Karsagi Byron DL, Shagan M, Brandis A, Mehlman T, Gopas J, Saravana Kumar R, Kushmaro A. Pseudomonas aeruginosa quorum sensing and biofilm attenuation by a di-hydroxy derivative of piperlongumine (PL-18). Biofilm 2024; 8:100215. [PMID: 39148892 PMCID: PMC11326495 DOI: 10.1016/j.bioflm.2024.100215] [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: 01/10/2024] [Revised: 07/01/2024] [Accepted: 07/13/2024] [Indexed: 08/17/2024] Open
Abstract
Bacterial communication, Quorum Sensing (QS), is a target against virulence and prevention of antibiotic-resistant infections. 16 derivatives of Piperlongumine (PL), an amide alkaloid from Piper longum L., were screened for QS inhibition. PL-18 had the best QSI activity. PL-18 inhibited the lasR-lasI, rhlR-rhlI, and pqs QS systems of Pseudomonas aeruginosa. PL-18 inhibited pyocyanin and rhamnolipids that are QS-controlled virulence elements. Iron is an essential element for pathogenicity, biofilm formation and resilience in harsh environments, its uptake was inhibited by PL-18. Pl-18 significantly reduced the biofilm biovolume including in established biofilms. PL-18-coated silicon tubes significantly inhibited biofilm formation. The transcriptome study of treated P. aeruginosa showed that PL-18 indeed reduced the expression of QS and iron homeostasis related genes, and up regulated sulfur metabolism related genes. Altogether, PL-18 inhibits QS, virulence, iron uptake, and biofilm formation. Thus, PL-18 should be further developed against bacterial infection, antibiotic resistance, and biofilm formation.
Collapse
Affiliation(s)
- Yael Schlichter Kadosh
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben Gurion University of the Negev, Beer Sheva, Israel
| | | | - Khairun Nisaa
- Department of Life Science, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Anat Ben-Zvi
- Department of Life Science, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Danit Lisa Karsagi Byron
- Department of Civil and Environmental Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Marilou Shagan
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Alexander Brandis
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Tevie Mehlman
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Jacob Gopas
- Department of Microbiology, Immunology and Genetics Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | | | - Ariel Kushmaro
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben Gurion University of the Negev, Beer Sheva, Israel
- The Ilse Katz Center for Nanoscale Science and Technology, Ben Gurion University of the Negev, Beer Sheva, Israel
- School of Sustainability and Climate Change, Ben Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
21
|
Medaglia S, Morellá-Aucejo Á, Ruiz-Rico M, Sancenón F, Villaescusa LA, Martínez-Máñez R, Marcos MD, Bernardos A. Antimicrobial Surfaces: Stainless Steel Functionalized with the Essential Oil Component Vanillin. Int J Mol Sci 2024; 25:12146. [PMID: 39596213 PMCID: PMC11595243 DOI: 10.3390/ijms252212146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/28/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
Pathogenic microorganisms can adhere to solid surfaces, leading to the formation of biofilms, thus building a physical barrier hindering the penetration and diffusion of antimicrobial compounds. In this context, the use of natural antimicrobial compounds, such as essential oil components, as substitutes for common synthetic antimicrobials in the fight to prevent antimicrobial resistance is explored. As stainless steel is one of the most widely used surfaces in different industries, we have developed an innovative antimicrobial treatment for stainless steel surfaces based on a multi-step functionalization process, in which the stainless steel surface is coated with a silica layer to which a vanillin derivative is covalently attached. The surface was analyzed by microscopy studies, indicating the correct immobilization on the surfaces. Antimicrobial studies (viability and bacterial adhesion assays) were performed against the bacteria Staphylococcus epidermidis, which is one of the most frequent causes of nosocomial infections. The results of the microbiological studies showed that vanillin-functionalized stainless steel surfaces reduce the bacteria viability by 100% and the biofilm formation on the stainless steel surface by 75% compared with non-functionalized surfaces, highlighting the contact-killing and adhesion resistance properties of the developed surface. Additional cycles using the functionalized surfaces showed good maintenance of the antimicrobial coating efficacy. Moreover, the surfaces coated with an intermediate silica layer demonstrated much greater antimicrobial activity than surfaces in which the active molecule was directly functionalized on the stainless steel surface.
Collapse
Affiliation(s)
- Serena Medaglia
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Camino de Vera s/n, 46022 Valencia, Spain; (S.M.); (Á.M.-A.); (F.S.); (L.A.V.); (R.M.-M.)
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ángela Morellá-Aucejo
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Camino de Vera s/n, 46022 Valencia, Spain; (S.M.); (Á.M.-A.); (F.S.); (L.A.V.); (R.M.-M.)
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, 46100 Valencia, Spain
| | - María Ruiz-Rico
- Instituto Universitario de Ingeniería de Alimentos (FoodUPV), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain;
| | - Félix Sancenón
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Camino de Vera s/n, 46022 Valencia, Spain; (S.M.); (Á.M.-A.); (F.S.); (L.A.V.); (R.M.-M.)
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, 46100 Valencia, Spain
- Departamento de Química, Universidad Politécnica de Valencia, Camino de Vera s/n, 46022 Valencia, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, Instituto de Investigación Sanitaria La Fe, Av Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - Luis A. Villaescusa
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Camino de Vera s/n, 46022 Valencia, Spain; (S.M.); (Á.M.-A.); (F.S.); (L.A.V.); (R.M.-M.)
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Química, Universidad Politécnica de Valencia, Camino de Vera s/n, 46022 Valencia, Spain
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Camino de Vera s/n, 46022 Valencia, Spain; (S.M.); (Á.M.-A.); (F.S.); (L.A.V.); (R.M.-M.)
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, 46100 Valencia, Spain
- Departamento de Química, Universidad Politécnica de Valencia, Camino de Vera s/n, 46022 Valencia, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, Instituto de Investigación Sanitaria La Fe, Av Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - M. Dolores Marcos
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Camino de Vera s/n, 46022 Valencia, Spain; (S.M.); (Á.M.-A.); (F.S.); (L.A.V.); (R.M.-M.)
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, 46100 Valencia, Spain
- Departamento de Química, Universidad Politécnica de Valencia, Camino de Vera s/n, 46022 Valencia, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, Instituto de Investigación Sanitaria La Fe, Av Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - Andrea Bernardos
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de Valencia, Universitat de València, Camino de Vera s/n, 46022 Valencia, Spain; (S.M.); (Á.M.-A.); (F.S.); (L.A.V.); (R.M.-M.)
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, 46100 Valencia, Spain
- Departamento de Química, Universidad Politécnica de Valencia, Camino de Vera s/n, 46022 Valencia, Spain
| |
Collapse
|
22
|
Cho KH. Molecular Targets in Streptococcus pyogenes for the Development of Anti-Virulence Agents. Genes (Basel) 2024; 15:1166. [PMID: 39336757 PMCID: PMC11430994 DOI: 10.3390/genes15091166] [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: 08/07/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
Streptococcus pyogenes, commonly known as Group A Streptococcus (GAS), is a significant human pathogen responsible for a wide range of diseases, from mild pharyngitis to severe conditions such as necrotizing fasciitis and toxic shock syndrome. The increasing antibiotic resistance, especially against macrolide antibiotics, poses a challenge to the effective treatment of these infections. This paper reviews the current state and mechanisms of antibiotic resistance in S. pyogenes. Furthermore, molecular targets for developing anti-virulence agents, which aim to attenuate virulence rather than killing it outright, are explored. This review specifically focuses on virulence regulators, proteins that coordinate the expression of multiple virulence factors in response to environmental signals, playing a crucial role in the pathogen's ability to cause disease. Key regulatory systems, such as RopB, Mga, CovRS, and the c-di-AMP signaling system, are discussed for their roles in modulating virulence gene expression. Additionally, potential molecular target sites for the development of anti-virulence agents are suggested. By concentrating on these regulatory pathways, it is proposed that anti-virulence strategies could enhance the effectiveness of existing antibiotics and reduce the selective pressure that drives the development of resistance.
Collapse
Affiliation(s)
- Kyu Hong Cho
- Department of Biology, Indiana State University, 600 Chestnut St. S224, Terre Haute, IN 47809, USA
| |
Collapse
|
23
|
Zolpirani FH, Ghaemi EA, Yasaghi M, Nikokar I, Ardebili A. Effect of phenylalanine arginyl β-naphthylamide on the imipenem resistance, elastase production, and the expression of quorum sensing and virulence factor genes in Pseudomonas aeruginosa clinical isolates. Braz J Microbiol 2024; 55:2715-2726. [PMID: 38926315 PMCID: PMC11405361 DOI: 10.1007/s42770-024-01426-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Pseudomonas aeruginosa is one of the most important nosocomial pathogens that possess the ability to produce multiple antibiotic resistance and virulence factors. Elastase B (LasB) is the major factor implicated in tissue invasion and damage during P. aeruginosa infections, whose synthesis is regulated by the quorum sensing (QS) system. Anti-virulence approach is now considered as potential therapeutic alternative and/or adjuvant to current antibiotics' failure. The aim of this study is primarily to find out the impact of the efflux pump inhibitor (EPI) phenylalanine arginyl β-naphthylamide (PAβN) on the production of elastase B and the gene expression of lasI quorum sensing and lasB virulence factor in clinical isolates of P. aeruginosa. Five P. aeruginosa isolates recovered from patients with respiratory tract infections were examined in this study. Antimicrobial susceptibility of isolates was performed by the disk agar diffusion method. Effect of the PAβN on imipenem susceptibility, bacterial viability, and elastase production was evaluated. The expression of lasB and lasI genes was measured by quantitative real-time PCR in the presence of PAβN. All isolates were identified as multidrug-resistant (MDR) and showed resistance to carbapenem (MIC = 64-256 µg/mL). Susceptibility of isolates to imipenem was highly increased in the presence of efflux inhibitor. PAβN significantly reduced elastase activity in three isolates tested without affecting bacterial growth. In addition, the relative expression of both lasB and lasI genes was diminished in all isolates in the presence of inhibitor. Efflux inhibition by using the EPI PAβN could be a potential target for controlling the P. aeruginosa virulence and pathogenesis. Furthermore, impairment of drug efflux by PAβN indicates its capability to be used as antimicrobial adjuvant that can decrease the resistance and lower the effective doses of current drugs.
Collapse
Affiliation(s)
- Fatemeh Hojjati Zolpirani
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ezat Allah Ghaemi
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohammad Yasaghi
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Iraj Nikokar
- Department of Laboratory Sciences, Langroud School of Allied Medical Sciences, Guilan University of Medical Sciences, Guilan, Iran
| | - Abdollah Ardebili
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
- Medical Bacteriology, Golestan University of Medical Sciences, 49341-74515, Gorgan, Iran.
| |
Collapse
|
24
|
Wang J, Yang J, Durairaj P, Wang W, Wei D, Tang S, Liu H, Wang D, Jia AQ. Discovery of β-nitrostyrene derivatives as potential quorum sensing inhibitors for biofilm inhibition and antivirulence factor therapeutics against Serratia marcescens. MLIFE 2024; 3:445-458. [PMID: 39359676 PMCID: PMC11442132 DOI: 10.1002/mlf2.12135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/14/2024] [Accepted: 04/25/2024] [Indexed: 10/04/2024]
Abstract
Quorum sensing (QS) inhibition has emerged as a promising target for directed drug design, providing an appealing strategy for developing antimicrobials, particularly against infections caused by drug-resistant pathogens. In this study, we designed and synthesized a total of 33 β-nitrostyrene derivatives using 1-nitro-2-phenylethane (NPe) as the lead compound, to target the facultative anaerobic bacterial pathogen Serratia marcescens. The QS-inhibitory effects of these compounds were evaluated using S. marcescens NJ01 and the reporter strain Chromobacterium violaceum CV026. Among the 33 new β-nitrostyrene derivatives, (E)-1-methyl-4-(2-nitrovinyl)benzene (m-NPe, compound 28) was proven to be a potent inhibitor that reduced biofilm formation of S. marcescens NJ01 by 79%. Scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM) results revealed that treatment with m-NPe (50 μg/ml) not only enhanced the susceptibility of the formed biofilms but also disrupted the architecture of biofilms by 84%. m-NPe (50 μg/ml) decreased virulence factors in S. marcescens NJ01, reducing the activity of protease, prodigiosin, and extracellular polysaccharide (EPS) by 36%, 72%, and 52%, respectively. In S. marcescens 4547, the activities of hemolysin and EPS were reduced by 28% and 40%, respectively, outperforming the positive control, vanillic acid (VAN). The study also found that the expression levels of QS- and biofilm-related genes (flhD, fimA, fimC, sodB, bsmB, pigA, pigC, and shlA) were downregulated by 1.21- to 2.32-fold. Molecular dynamics analysis showed that m-NPe could bind stably to SmaR, RhlI, RhlR, LasR, and CviR proteins in a 0.1 M sodium chloride solution. Importantly, a microscale thermophoresis (MST) test revealed that SmaR could be a target protein for the screening of a quorum sensing inhibitor (QSI) against S. marcescens. Overall, this study highlights the efficacy of m-NPe in suppressing the virulence factors of S. marcescens, identifying it as a new potential QSI and antibiofilm agent capable of restoring or improving antimicrobial drug sensitivity.
Collapse
Affiliation(s)
- Jiang Wang
- Hainan Affiliated Hospital of Hainan Medical University Hainan General Hospital Haikou China
- Center for Translational Research Shenzhen Bay Laboratory Shenzhen China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences Hainan University Haikou China
| | - Jingyi Yang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences Hainan University Haikou China
- Hainan Branch, Shanghai Children's Medical Center, School of Medicine Shanghai Jiao Tong University Sanya China
| | - Pradeepraj Durairaj
- Center for Translational Research Shenzhen Bay Laboratory Shenzhen China
- Present address: National High Magnetic Field Laboratory, FAMU-FSU College of Engineering Florida State University Tallahassee Florida USA
| | - Wei Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences Hainan University Haikou China
| | - Dongyan Wei
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences Hainan University Haikou China
| | - Shi Tang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences Hainan University Haikou China
| | - Haiqing Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences Hainan University Haikou China
| | - Dayong Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences Hainan University Haikou China
| | - Ai-Qun Jia
- Hainan Affiliated Hospital of Hainan Medical University Hainan General Hospital Haikou China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences Hainan University Haikou China
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Çevikbaş H, Ulusoy S, Kaya Kinaytürk N. Exploring rose absolute and phenylethyl alcohol as novel quorum sensing inhibitors in Pseudomonas aeruginosa and Chromobacterium violaceum. Sci Rep 2024; 14:15666. [PMID: 38977845 PMCID: PMC11231148 DOI: 10.1038/s41598-024-66888-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024] Open
Abstract
Inter-cellular signaling, referred to as quorum sensing (QS), regulates the production of virulence factors in numerous gram-negative bacteria, such as the human pathogens Pseudomonas aeruginosa and Chromobacterium violaceum. QS inhibition may provide an opportunity for the treatment of bacterial infections. This represents the initial study to examine the antibiofilm and antivirulence capabilities of rose absolute and its primary component, phenylethyl alcohol. QS inhibition was assessed by examining extracellular exopolysaccharide synthesis, biofilm development, and swarming motility in P. aeruginosa PAO1, along with violacein production in C. violaceum ATCC 12472. Molecular docking analysis was conducted to explore the mechanism by which PEA inhibits QS. Our results indicate that rose absolute and PEA caused decrease in EPS production (60.5-33.5%), swarming motility (94.7-64.5%), and biofilm formation (98.53-55.5%) in the human pathogen P. aeruginosa PAO1. Violacein production decreased by 98.1% and 62.5% with an absolute (0.5 v/v %) and PEA (2 mM). Moreover, the molecular docking analysis revealed a promising competitive interaction between PEA and AHLs. Consequently, this study offers valuable insights into the potential of rose absolute and PEA as inhibitors of QS in P. aeruginosa and C. violaceum.
Collapse
Affiliation(s)
- Halime Çevikbaş
- Faculty of Engineering and Natural Sciences, Biology Department, Süleyman Demirel University, Isparta, 32260, Turkey
| | - Seyhan Ulusoy
- Faculty of Engineering and Natural Sciences, Biology Department, Süleyman Demirel University, Isparta, 32260, Turkey.
| | - Neslihan Kaya Kinaytürk
- Faculty of Arts and Science, Nanoscience and Nanotechnology Department, Mehmet Akif Ersoy University, Burdur, 15100, Turkey
| |
Collapse
|
28
|
Edvardsen PKT, Askarian F, Zurich R, Nizet V, Vaaje-Kolstad G. Exploring roles of the chitinase ChiC in modulating Pseudomonas aeruginosa virulence phenotypes. Microbiol Spectr 2024; 12:e0054624. [PMID: 38819151 PMCID: PMC11218509 DOI: 10.1128/spectrum.00546-24] [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: 02/29/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
Chitinases are ubiquitous enzymes involved in biomass degradation and chitin turnover in nature. Pseudomonas aeruginosa (PA), an opportunistic human pathogen, expresses ChiC, a secreted glycoside hydrolase 18 family chitinase. Despite speculation about ChiC's role in PA disease pathogenesis, there is scant evidence supporting this hypothesis. Since PA cannot catabolize chitin, we investigated the potential function(s) of ChiC in PA pathophysiology. Our findings show that ChiC exhibits activity against both insoluble (α- and β-chitin) and soluble chitooligosaccharides. Enzyme kinetics toward (GlcNAc)4 revealed a kcat of 6.50 s-1 and a KM of 1.38 mM, the latter remarkably high for a canonical chitinase. In our label-free proteomics investigation, ChiC was among the most abundant proteins in the Pel biofilm, suggesting a potential contribution to PA biofilm formation. Using an intratracheal challenge model of PA pneumonia, the chiC::ISphoA/hah transposon insertion mutant paradoxically showed slightly increased virulence compared to the wild-type parent strain. Our results indicate that ChiC is a genuine chitinase that contributes to a PA pathoadaptive pathway.IMPORTANCEIn addition to performing chitin degradation, chitinases from the glycoside hydrolase 18 family have been found to play important roles during pathogenic bacterial infection. Pseudomonas aeruginosa is an opportunistic pathogen capable of causing pneumonia in immunocompromised individuals. Despite not being able to grow on chitin, the bacterium produces a chitinase (ChiC) with hitherto unknown function. This study describes an in-depth characterization of ChiC, focusing on its potential contribution to the bacterium's disease-causing ability. We demonstrate that ChiC can degrade both polymeric chitin and chitooligosaccharides, and proteomic analysis of Pseudomonas aeruginosa biofilm revealed an abundance of ChiC, hinting at a potential role in biofilm formation. Surprisingly, a mutant strain incapable of ChiC production showed higher virulence than the wild-type strain. While ChiC appears to be a genuine chitinase, further investigation is required to fully elucidate its contribution to Pseudomonas aeruginosa virulence, an important task given the evident health risk posed by this bacterium.
Collapse
Affiliation(s)
| | - Fatemeh Askarian
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego School of Medicine, La Jolla, California, USA
| | - Raymond Zurich
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego School of Medicine, La Jolla, California, USA
| | - Victor Nizet
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego School of Medicine, La Jolla, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
| | - Gustav Vaaje-Kolstad
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
29
|
Suzuki S, Morita Y, Ishige S, Kai K, Kawasaki K, Matsushita K, Ogura K, Miyoshi-Akiyama† T, Shimizu T. Effects of quorum sensing-interfering agents, including macrolides and furanone C-30, and an efflux pump inhibitor on nitrosative stress sensitivity in Pseudomonas aeruginosa. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001464. [PMID: 38900549 PMCID: PMC11263931 DOI: 10.1099/mic.0.001464] [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: 03/08/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024]
Abstract
Long-term administration of certain macrolides is efficacious in patients with persistent pulmonary Pseudomonas aeruginosa infection, despite how limited the clinically achievable concentrations are, being far below their MICs. An increase in the sub-MIC of macrolide exposure-dependent sensitivity to nitrosative stress is a typical characteristic of P. aeruginosa. However, a few P. aeruginosa clinical isolates do not respond to sub-MIC of macrolide treatment. Therefore, we examined the effects of sub-MIC of erythromycin (EM) on the sensitivity to nitrosative stress together with an efflux pump inhibitor (EPI) phenylalanine arginyl β-naphthylamide (PAβN). The sensitivity to nitrosative stress increased, suggesting that the efflux pump was involved in inhibiting the sub-MIC of macrolide effect. Analysis using efflux pump-mutant P. aeruginosa revealed that MexAB-OprM, MexXY-OprM, and MexCD-OprJ are factors in reducing the sub-MIC of macrolide effect. Since macrolides interfere with quorum sensing (QS), we demonstrated that the QS-interfering agent furanone C-30 (C-30) producing greater sensitivity to nitric oxide (NO) stress than EM. The effect of C-30 was decreased by overproduction of MexAB-OprM. To investigate whether the increase in the QS-interfering agent exposure-dependent sensitivity to nitrosative stress is characteristic of P. aeruginosa clinical isolates, we examined the viability of P. aeruginosa treated with NO. Although treatment with EM could reduce cell viability, a high variability in EM effects was observed. Conversely, C-30 was highly effective at reducing cell viability. Treatment with both C-30 and PAβN was sufficiently effective against the remaining isolates. Therefore, the combination of a QS-interfering agent and an EPI could be effective in treating P. aeruginosa infections.
Collapse
Affiliation(s)
- Shin Suzuki
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan
- Division of Laboratory Medicine, Chiba University Hospital, 1-8-1 Inohana, Chiba, 260-8677, Japan
| | - Yuji Morita
- Department of Infection Control Science, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Shota Ishige
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan
| | - Kiyohiro Kai
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan
| | - Kenji Kawasaki
- Division of Laboratory Medicine, Chiba University Hospital, 1-8-1 Inohana, Chiba, 260-8677, Japan
| | - Kazuyuki Matsushita
- Division of Laboratory Medicine, Chiba University Hospital, 1-8-1 Inohana, Chiba, 260-8677, Japan
| | - Kohei Ogura
- Laboratory of Basic and Applied Molecular Biotechnology, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Gokasho, Uji, Kyoto, 6110011, Japan
| | - Tohru Miyoshi-Akiyama†
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Takeshi Shimizu
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan
| |
Collapse
|
30
|
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
|
31
|
Manisha Y, Srinivasan M, Jobichen C, Rosenshine I, Sivaraman J. Sensing for survival: specialised regulatory mechanisms of Type III secretion systems in Gram-negative pathogens. Biol Rev Camb Philos Soc 2024; 99:837-863. [PMID: 38217090 DOI: 10.1111/brv.13047] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/15/2024]
Abstract
For centuries, Gram-negative pathogens have infected the human population and been responsible for numerous diseases in animals and plants. Despite advancements in therapeutics, Gram-negative pathogens continue to evolve, with some having developed multi-drug resistant phenotypes. For the successful control of infections caused by these bacteria, we need to widen our understanding of the mechanisms of host-pathogen interactions. Gram-negative pathogens utilise an array of effector proteins to hijack the host system to survive within the host environment. These proteins are secreted into the host system via various secretion systems, including the integral Type III secretion system (T3SS). The T3SS spans two bacterial membranes and one host membrane to deliver effector proteins (virulence factors) into the host cell. This multifaceted process has multiple layers of regulation and various checkpoints. In this review, we highlight the multiple strategies adopted by these pathogens to regulate or maintain virulence via the T3SS, encompassing the regulation of small molecules to sense and communicate with the host system, as well as master regulators, gatekeepers, chaperones, and other effectors that recognise successful host contact. Further, we discuss the regulatory links between the T3SS and other systems, like flagella and metabolic pathways including the tricarboxylic acid (TCA) cycle, anaerobic metabolism, and stringent cell response.
Collapse
Affiliation(s)
- Yadav Manisha
- Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Mahalashmi Srinivasan
- Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Chacko Jobichen
- Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Ilan Rosenshine
- Department of Microbiology and Molecular Genetics, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem, 91120, Israel
| | - J Sivaraman
- Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| |
Collapse
|
32
|
Quni S, Zhang Y, Liu L, Liu M, Zhang L, You J, Cui J, Liu X, Wang H, Li D, Zhou Y. NF-κB-Signaling-Targeted Immunomodulatory Nanoparticle with Photothermal and Quorum-Sensing Inhibition Effects for Efficient Healing of Biofilm-Infected Wounds. ACS APPLIED MATERIALS & INTERFACES 2024; 16:25757-25772. [PMID: 38738757 DOI: 10.1021/acsami.4c03142] [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: 05/14/2024]
Abstract
The development of therapeutics with high antimicrobial activity and immunomodulatory effects is urgently needed for the treatment of infected wounds due to the increasing danger posed by recalcitrant-infected wounds. In this study, we developed light-controlled antibacterial, photothermal, and immunomodulatory biomimetic N/hPDA@M nanoparticles (NPs). This nanoplatform was developed by loading flavonoid naringenin onto hollow mesoporous polydopamine NPs in a π-π-stacked configuration and encasing them with macrophage membranes. First, our N/hPDA@M NPs efficiently neutralized inflammatory factors present within the wound microenvironment by the integration of macrophage membranes. Afterward, the N/hPDA@M NPs effectively dismantled bacterial biofilms through a combination of the photothermal properties of PDA and the quorum sensing inhibitory effects of naringenin. It is worth noting that N/hPDA@M NPs near-infrared-enhanced release of naringenin exhibited specificity toward the NF-κB-signaling pathway, effectively mitigating the inflammatory response. This innovative design not only conferred remarkable antibacterial properties upon the N/hPDA@M NPs but also endowed them with the capacity to modulate inflammatory responses, curbing excessive inflammation and steering macrophage polarization toward the M2 phenotype. As a result, this multifaceted approach significantly contributes to expediting the healing process of infected skin wounds.
Collapse
Affiliation(s)
- Sezhen Quni
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Jilin 130021, Changchun, China
| | - Yidi Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Jilin 130021, Changchun, China
| | - Lijun Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Jilin 130021, Changchun, China
| | - Manxuan Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Jilin 130021, Changchun, China
| | - Lu Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Jilin 130021, Changchun, China
| | - Jiaqian You
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Jilin 130021, Changchun, China
| | - Jing Cui
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Jilin 130021, Changchun, China
| | - Xiuyu Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Jilin 130021, Changchun, China
| | - Hanchi Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Jilin 130021, Changchun, China
| | - Daowei Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Jilin 130021, Changchun, China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, China
- School of Stomatology, Jilin University, Jilin 130021, Changchun, China
| |
Collapse
|
33
|
Bendary MM, Ali MAM, Abdel Halim AS, Boufahja F, Chaudhary AA, Elkelish A, Soliman RHM, Hegazy WAH. Investigating Sulforaphane's anti-virulence and anti-quorum sensing properties against Pseudomonas aeruginosa. Front Pharmacol 2024; 15:1406653. [PMID: 38835668 PMCID: PMC11148281 DOI: 10.3389/fphar.2024.1406653] [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: 03/25/2024] [Accepted: 04/29/2024] [Indexed: 06/06/2024] Open
Abstract
Background P. aeruginosa, a significant bacterium, can cause severe illness and resistance to antibiotics. Quorum sensing (QS) systems regulate virulence factors production. Targeting QS could reduce bacteria pathogenicity and prevent antibiotic resistance. Cruciferous vegetables contain sulforaphane, known for its anti-inflammatory, antioxidant, anticancer, and antimicrobial properties. Aim We aimed to examine the inhibitory influences of sulforaphane, at a sub-inhibitory concentration (¼ minimum inhibitory concentration, MIC), on virulence and QS in P. aeruginosa. Materials and methods The sulforaphane's anti-virulence actions at sub-inhibitory concentrations were explored in vitro and in vivo. A sub-MIC concentration of sulforaphane was combined with anti-pseudomonal drugs, and the results of this combination were assessed. The virtual affinity of sulforaphane for the receptors of QS was studied, and its effect on the expression of QS genes was quantified. Results Sulforaphane significantly decreased the biofilm formation, motility, ability to withstand oxidative stress, and the synthesis of virulence extracellular enzymes such as proteases, hemolysins, and elastase, as well as other virulence factors like pyocyanin. In addition, sulforaphane lessened the severity of P. aeruginosa infection in mice. Sulforaphane reduced the antipseudomonal antibiotics' MICs when used together, resulting in synergistic effects. The observed anti-virulence impacts were attributed to the ability of sulforaphane to inhibit QS via suppressing the QS genes' expression. Conclusion Sulforaphane shows promise as a potent anti-virulence and anti-QS agent that can be used alongside conventional antimicrobials to manage severe infections effectively. Furthermore, this study paves the way for further investigation of sulforaphane and similar structures as pharmacophores for anti-QS candidates.
Collapse
Affiliation(s)
- Mahmoud M Bendary
- Department of Microbiology and Immunology, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Mohamed A M Ali
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Alyaa S Abdel Halim
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Fehmi Boufahja
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Amr Elkelish
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
- Department of Botany and Microbiology, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Rania H M Soliman
- Department of Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Wael A H Hegazy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
- Pharmacy Program, Department of Pharmaceutical Sciences, Oman College of Health Sciences, Muscat, Oman
| |
Collapse
|
34
|
Conaway A, Todorovic I, Mould DL, Hogan DA. Loss of LasR function leads to decreased repression of Pseudomonas aeruginosa PhoB activity at physiological phosphate concentrations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.586856. [PMID: 38585852 PMCID: PMC10996656 DOI: 10.1101/2024.03.27.586856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
While the Pseudomonas aeruginosa LasR transcription factor plays a role in quorum sensing (QS) across phylogenetically-distinct lineages, isolates with loss-of-function mutations in lasR (LasR- strains) are commonly found in diverse settings including infections where they are associated with worse clinical outcomes. In LasR- strains, the transcription factor RhlR, which is controlled by LasR, can be alternately activated in low inorganic phosphate (Pi) concentrations via the two-component system PhoR-PhoB. Here, we demonstrate a new link between LasR and PhoB in which the absence of LasR increases PhoB activity at physiological Pi concentrations and raises the Pi concentration necessary for PhoB inhibition. PhoB activity was also less repressed by Pi in mutants lacking different QS regulators (RhlR and PqsR) and in mutants lacking genes required for the production of QS-regulated phenazines suggesting that decreased phenazine production was one reason for decreased PhoB repression by Pi in LasR- strains. In addition, the CbrA-CbrB two-component system, which is elevated in LasR- strains, was necessary for reduced PhoB repression by Pi and a Δcrc mutant, which lacks the CbrA-CbrB-controlled translational repressor, activated PhoB at higher Pi concentrations than the wild type. The ΔlasR mutant had a PhoB-dependent growth advantage in a medium with no added Pi and increased virulence-determinant gene expression in a medium with physiological Pi, in part through reactivation of QS. This work suggests PhoB activity may contribute to the virulence of LasR- P. aeruginosa and subsequent clinical outcomes.
Collapse
Affiliation(s)
- Amy Conaway
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH USA
| | - Igor Todorovic
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH USA
| | - Dallas L. Mould
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH USA
| | - Deborah A. Hogan
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH USA
| |
Collapse
|
35
|
Al-Rabia MW, Asfour HZ, Alhakamy NA, Abdulaal WH, Ibrahim TS, Abbas HA, Salem IM, Hegazy WAH, Nazeih SI. Thymoquinone is a natural antibiofilm and pathogenicity attenuating agent in Pseudomonas aeruginosa. Front Cell Infect Microbiol 2024; 14:1382289. [PMID: 38638827 PMCID: PMC11024287 DOI: 10.3389/fcimb.2024.1382289] [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: 02/05/2024] [Accepted: 03/08/2024] [Indexed: 04/20/2024] Open
Abstract
Pseudomonas aeruginosa belongs to the critical pathogens that represent a global public health problem due to their high rate of resistance as listed by WHO. P. aeruginosa can result in many nosocomial infections especially in individuals with compromised immune systems. Attenuating virulence factors by interference with quorum sensing (QS) systems is a promising approach to treat P. aeruginosa-resistant infections. Thymoquinone is a natural compound isolated from Nigella sativa (black seed) essential oil. In this study, the minimum inhibitory concentration of thymoquinone was detected followed by investigating the antibiofilm and antivirulence activities of the subinhibitory concentration of thymoquinone against P. aeruginosa PAO1. The effect of thymoquinone on the expression of QS genes was assessed by quantitative real-time PCR, and the protective effect of thymoquinone against the pathogenesis of PAO1 in mice was detected by the mouse survival test. Thymoquinone significantly inhibited biofilm, pyocyanin, protease activity, and swarming motility. At the molecular level, thymoquinone markedly downregulated QS genes lasI, lasR, rhlI, and rhlR. Moreover, thymoquinone could protect mice from the pathologic effects of P. aeruginosa increasing mouse survival from 20% to 100%. In conclusion, thymoquinone is a promising natural agent that can be used as an adjunct therapeutic agent with antibiotics to attenuate the pathogenicity of P. aeruginosa.
Collapse
Affiliation(s)
- Mohammed W. Al-Rabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hani Z. Asfour
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Wesam H. Abdulaal
- Department of Biochemistry, Faculty of Science, Cancer and Mutagenesis Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tarek S. Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hisham A. Abbas
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Ibrahim M. Salem
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sphinx University, Assiut, Egypt
| | - Wael A. H. Hegazy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
- Pharmacy Program, Department of Pharmaceutical Sciences, Oman College of Health Sciences, Muscat, Oman
| | - Shaimaa I. Nazeih
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
36
|
Kang D, Xu Q, Kirienko NV. In vitro lung epithelial cell model reveals novel roles for Pseudomonas aeruginosa siderophores. Microbiol Spectr 2024; 12:e0369323. [PMID: 38311809 PMCID: PMC10913452 DOI: 10.1128/spectrum.03693-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/21/2023] [Indexed: 02/06/2024] Open
Abstract
The multidrug-resistant pathogen Pseudomonas aeruginosa is a common nosocomial respiratory pathogen that continues to threaten the lives of patients with mechanical ventilation in intensive care units and those with underlying comorbidities such as cystic fibrosis or chronic obstructive pulmonary disease. For over 20 years, studies have repeatedly demonstrated that the major siderophore pyoverdine is an important virulence factor for P. aeruginosa in invertebrate and mammalian hosts in vivo. Despite its physiological significance, an in vitro, mammalian cell culture model that can be used to characterize the impact and molecular mechanisms of pyoverdine-mediated virulence has only been developed very recently. In this study, we adapt a previously-established, murine macrophage-based model to use human bronchial epithelial (16HBE) cells. We demonstrate that conditioned medium from P. aeruginosa induced rapid 16HBE cell death through the pyoverdine-dependent secretion of cytotoxic rhamnolipids. Genetic or chemical disruption of pyoverdine biosynthesis decreased rhamnolipid production and mitigated cell death. Consistent with these observations, chemical depletion of lipids or genetic disruption of rhamnolipid biosynthesis abrogated the toxicity of the conditioned medium. Furthermore, we also examine the effects of exposure to purified pyoverdine on 16HBE cells. While pyoverdine accumulated within cells, it was largely sequestered within early endosomes, resulting in minimal cytotoxicity. More membrane-permeable iron chelators, such as the siderophore pyochelin, decreased epithelial cell viability and upregulated several pro-inflammatory genes. However, pyoverdine potentiated these iron chelators in activating pro-inflammatory pathways. Altogether, these findings suggest that the siderophores pyoverdine and pyochelin play distinct roles in virulence during acute P. aeruginosa lung infection. IMPORTANCE Multidrug-resistant Pseudomonas aeruginosa is a versatile bacterium that frequently causes lung infections. This pathogen is life-threatening to mechanically-ventilated patients in intensive care units and is a debilitating burden for individuals with cystic fibrosis. However, the role of P. aeruginosa virulence factors and their regulation during infection are not fully understood. Previous murine lung infection studies have demonstrated that the production of siderophores (e.g., pyoverdine and pyochelin) is necessary for full P. aeruginosa virulence. In this report, we provide further mechanistic insight into this phenomenon. We characterize distinct and novel ways these siderophores contribute to virulence using an in vitro human lung epithelial cell culture model.
Collapse
Affiliation(s)
- Donghoon Kang
- Department of BioSciences, Rice University, Houston, Texas, USA
| | - Qi Xu
- Department of BioSciences, Rice University, Houston, Texas, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | | |
Collapse
|
37
|
Juszczuk-Kubiak E. Molecular Aspects of the Functioning of Pathogenic Bacteria Biofilm Based on Quorum Sensing (QS) Signal-Response System and Innovative Non-Antibiotic Strategies for Their Elimination. Int J Mol Sci 2024; 25:2655. [PMID: 38473900 DOI: 10.3390/ijms25052655] [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: 12/19/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
One of the key mechanisms enabling bacterial cells to create biofilms and regulate crucial life functions in a global and highly synchronized way is a bacterial communication system called quorum sensing (QS). QS is a bacterial cell-to-cell communication process that depends on the bacterial population density and is mediated by small signalling molecules called autoinducers (AIs). In bacteria, QS controls the biofilm formation through the global regulation of gene expression involved in the extracellular polymeric matrix (EPS) synthesis, virulence factor production, stress tolerance and metabolic adaptation. Forming biofilm is one of the crucial mechanisms of bacterial antimicrobial resistance (AMR). A common feature of human pathogens is the ability to form biofilm, which poses a serious medical issue due to their high susceptibility to traditional antibiotics. Because QS is associated with virulence and biofilm formation, there is a belief that inhibition of QS activity called quorum quenching (QQ) may provide alternative therapeutic methods for treating microbial infections. This review summarises recent progress in biofilm research, focusing on the mechanisms by which biofilms, especially those formed by pathogenic bacteria, become resistant to antibiotic treatment. Subsequently, a potential alternative approach to QS inhibition highlighting innovative non-antibiotic strategies to control AMR and biofilm formation of pathogenic bacteria has been discussed.
Collapse
Affiliation(s)
- Edyta Juszczuk-Kubiak
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology-State Research Institute, Rakowiecka 36 Street, 02-532 Warsaw, Poland
| |
Collapse
|
38
|
Kristensen R, Andersen JB, Rybtke M, Jansen CU, Fritz BG, Kiilerich RO, Uhd J, Bjarnsholt T, Qvortrup K, Tolker-Nielsen T, Givskov M, Jakobsen TH. Inhibition of Pseudomonas aeruginosa quorum sensing by chemical induction of the MexEF-oprN efflux pump. Antimicrob Agents Chemother 2024; 68:e0138723. [PMID: 38189278 PMCID: PMC10848761 DOI: 10.1128/aac.01387-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/17/2023] [Indexed: 01/09/2024] Open
Abstract
The cell-to-cell communication system quorum sensing (QS), used by various pathogenic bacteria to synchronize gene expression and increase host invasion potentials, is studied as a potential target for persistent infection control. To search for novel molecules targeting the QS system in the Gram-negative opportunistic pathogen Pseudomonas aeruginosa, a chemical library consisting of 3,280 small compounds from LifeArc was screened. A series of 10 conjugated phenones that have not previously been reported to target bacteria were identified as inhibitors of QS in P. aeruginosa. Two lead compounds (ethylthio enynone and propylthio enynone) were re-synthesized for verification of activity and further elucidation of the mode of action. The isomeric pure Z-ethylthio enynone was used for RNA sequencing, revealing a strong inhibitor of QS-regulated genes, and the QS-regulated virulence factors rhamnolipid and pyocyanin were significantly decreased by treatment with the compounds. A transposon mutagenesis screen performed in a newly constructed lasB-gfp monitor strain identified the target of Z-ethylthio enynone in P. aeruginosa to be the MexEF-OprN efflux pump, which was further established using defined mex knockout mutants. Our data indicate that the QS inhibitory capabilities of Z-ethylthio enynone were caused by the drainage of intracellular signal molecules as a response to chemical-induced stimulation of the MexEF-oprN efflux pump, thereby inhibiting the autogenerated positive feedback and its enhanced signal-molecule synthesis.
Collapse
Affiliation(s)
- Rasmus Kristensen
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bo Andersen
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Morten Rybtke
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | | | - Blaine Gabriel Fritz
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Rikke Overgaard Kiilerich
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Uhd
- Department of Chemistry, Technical University of Denmark, Lyngby, Denmark
| | - Thomas Bjarnsholt
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Katrine Qvortrup
- Department of Chemistry, Technical University of Denmark, Lyngby, Denmark
| | - Tim Tolker-Nielsen
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Michael Givskov
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Tim Holm Jakobsen
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
39
|
Yang Q, Defoirdt T. Weak selection for resistance to quorum sensing inhibition during multiple host infection cycles. THE ISME JOURNAL 2024; 18:wrae251. [PMID: 39689238 DOI: 10.1093/ismejo/wrae251] [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: 09/11/2024] [Revised: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 12/19/2024]
Abstract
Quorum sensing (QS) inhibition is a promising novel approach to control bacterial infections. However, it is not clear whether QS inhibition will impose selective pressure for the spread of resistance against QS inhibition in pathogen populations. Previous research tried to answer this question by using synthetic growth media, and this revealed that whether or not resistance will spread completely depends on the environment in which it is studied. Therefore, the spread of resistance should be studied in the environment where it ultimately matters: in vivo during infection of a host. Here, using QS inhibitor-susceptible and -resistant mimics, we show that resistance to QS inhibition does not spread in host-associated populations of Vibrio campbellii during up to 35 cycles of infection and transmission if the initial frequency of the resistance is low in the pathogen population, whereas it further increases to 100% if it is already prevalent. However, even in the latter case, the resistance spreads at a slower pace than resistance to antibiotics spreads under the same conditions.
Collapse
Affiliation(s)
- Qian Yang
- Department of Biotechnology, Center for Microbial Ecology and Technology (CMET), Ghent University, Frieda Saeysstraat 1, 9052 Gent, Belgium
| | - Tom Defoirdt
- Department of Biotechnology, Center for Microbial Ecology and Technology (CMET), Ghent University, Frieda Saeysstraat 1, 9052 Gent, Belgium
| |
Collapse
|
40
|
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
|
41
|
Ham S, Ryoo HS, Jang Y, Lee SH, Lee JY, Kim HS, Lee JH, Park HD. Isolation of a quorum quenching bacterium effective to various acyl-homoserine lactones: Its quorum quenching mechanism and application to a membrane bioreactor. CHEMOSPHERE 2024; 347:140735. [PMID: 37977541 DOI: 10.1016/j.chemosphere.2023.140735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/27/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Biofouling, caused by microbial biofilm formation on the membrane surface and in pores, is a major operational problem in membrane bioreactors (MBR). Many quorum quenching (QQ) bacteria have been isolated and applied to MBR to reduce biofouling. However, for more effective MBR biofouling control, novel approaches for isolating QQ bacteria and applying them in MBR are needed. Therefore, Listeria grayi (HEMM-2) was isolated using a mixture of different N-acyl homoserine lactones (AHLs). HEMM-2 degraded various AHLs, regardless of the length and oxo group in the carbon chain, with quorum sensing (QS) inhibition ratios of 47-61%. This QQ activity was attributed to extracellular substances in HEMM-2 cell-free supernatant (CFS). Furthermore, the HEMM-2 CFS negatively regulated QS-related gene expression, inhibiting Pseudomonas aeruginosa and activated sludge-biofilm formation by 53-75%. Surprisingly, when the HEMM-2 CFS was directly injected into a laboratory-scale MBR system, biofouling was not significantly affected. Biofouling was only controlled by cell suspension (CS) of HEMM-2, indicating the importance of QQ bacteria in MBR. The HEMM-2 CS increased operation time to reach 0.4 bar, a threshold transmembrane pressure for complete biofouling, from 315 h to 371 h. Taken together, HEMM-2, which is effective in the degradation of various AHLs, and its applicable method to MBR may be considered a potent approach for controlling biofouling and understanding the behavior of QQ bacteria in MBR systems.
Collapse
Affiliation(s)
- Soyoung Ham
- Department of Geosciences, University of Tübingen, Schnarrenbergstraße 94-96, Tübingen, 72076, Germany
| | - Hwa-Soo Ryoo
- Department of Civil, Environmental and Architectural Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yongsun Jang
- Department of Civil, Environmental and Architectural Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Sang-Hoon Lee
- Department of Civil, Environmental and Architectural Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Ji-Yoon Lee
- Department of Civil, Environmental and Architectural Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Han-Shin Kim
- Division of Biotechnology, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea
| | - Jeong-Hoon Lee
- Eco Lab Center, SK Ecoplant Co.,Ltd., 19, Yulgok-ro 2-gil, Jongro-gu, Seoul, 03143, Republic of Korea
| | - Hee-Deung Park
- Department of Civil, Environmental and Architectural Engineering, Korea University, Seoul, 02841, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
42
|
Mellini M, Letizia M, Caruso L, Guiducci A, Meneghini C, Heeb S, Williams P, Cámara M, Visca P, Imperi F, Leoni L, Rampioni G. RsaL-driven negative regulation promotes heterogeneity in Pseudomonas aeruginosa quorum sensing. mBio 2023; 14:e0203923. [PMID: 37843294 PMCID: PMC10746200 DOI: 10.1128/mbio.02039-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/05/2023] [Indexed: 10/17/2023] Open
Abstract
IMPORTANCE Single-cell analyses can reveal that despite experiencing identical physico-chemical conditions, individual bacterial cells within a monoclonal population may exhibit variations in gene expression. Such phenotypic heterogeneity has been described for several aspects of bacterial physiology, including QS activation. This study demonstrates that the transition of non-quorate cells to the quorate state is a graded process that does not occur at a specific cell density and that subpopulations of non-quorate cells also persist at high cell density. Here, we provide a mechanistic explanation for this phenomenon, showing that a negative feedback regulatory loop integrated into the las system has a pivotal role in promoting cell-to-cell variation in the QS activation state and in limiting the transition of non-quorate cells to the quorate state in P. aeruginosa.
Collapse
Affiliation(s)
- Marta Mellini
- Department of Science, University Roma Tre, Rome, Italy
| | | | | | | | | | - Stephan Heeb
- National Biofilms Innovation Centre, Biodiscovery Institute and School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Paul Williams
- National Biofilms Innovation Centre, Biodiscovery Institute and School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Miguel Cámara
- National Biofilms Innovation Centre, Biodiscovery Institute and School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Paolo Visca
- Department of Science, University Roma Tre, Rome, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
- IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Francesco Imperi
- Department of Science, University Roma Tre, Rome, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
- IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Livia Leoni
- Department of Science, University Roma Tre, Rome, Italy
| | - Giordano Rampioni
- Department of Science, University Roma Tre, Rome, Italy
- IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
43
|
Dahshan NA, Abu-Dahab R, Khalil EA, Al-Bakri AG. Bactericidal effect of Iberin combined with photodynamic antimicrobial chemotherapy against Pseudomonas aeruginosa biofilm cultured on ex vivo wound model. Photodiagnosis Photodyn Ther 2023; 44:103841. [PMID: 37832710 DOI: 10.1016/j.pdpdt.2023.103841] [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: 02/19/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
Wounds infected by Pseudomonas aeruginosa (P. aeruginosa) biofilms are characterized by poor healing and by being long lasting. Pyocyanin and pyoverdine are exotoxins that contribute to P. aeruginosa pathogenicity in wound infections and are known as virulence factors. Despite the usefulness of antimicrobial photodynamic therapy (PDT) in the management of wound infections, biofilms are hurdle for microbial photoinactivation. Quorum sensing (QS) is a cell density-dependent chemical signaling system P. aeruginosa uses to regulate biofilm formation and virulence factors production. In the current study, QS attenuation was used in combination with PDT against P. aeruginosa biofilm cultured on skin explant. Iberin is a QS inhibitor that attenuates P. aeruginosa virulence and affects biofilm integrity. The antibiofilm and QS inhibitory activities of iberin in combination with either riboflavin or 5,10,15,20-Tetrakis(1-methyl-4-pyridinio) porphyrin tetra p-toluenesulfonate (TMP) mediated PDT were investigated using viable count method and pyocyanin and pyoverdine assays, respectively. No bactericidal activity was reported when iberin was added to a mature biofilm (24 h) followed by PDT. When added to a growing biofilm at multiple time points (0 h, 24 h and 48 h), iberin inhibited P. aeruginosa biofilm QS signaling system. This inhibitory effect resulted in an observable decrease in the levels of the QS-regulated virulence factors, pyocyanin and pyoverdine, without any effect on the growth of the biofilm cultures. These changes in biofilm virulence were associated with a decrease in biofilm resistance to PDT and caused bactericidal effect upon photosensitizers treatment and irradiation. Iberin-treated-riboflavin-mediated PDT resulted in a significant 1.3 log reduction in biofilm population. Similarly, iberin-treated-TMP-mediated PDT caused a significant 1.8 log reduction in biofilm population. The combination of QS inhibitor with PDT is a promising alternative antimicrobial therapy for the management of biofilms.
Collapse
Affiliation(s)
- Nisreen A Dahshan
- Department of Applied Pharmaceutical Sciences and Clinical Pharmacy, Faculty of Pharmacy, Isra University, Amman, Jordan
| | - Rana Abu-Dahab
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Enam A Khalil
- Department of Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Amal G Al-Bakri
- Department of Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, The University of Jordan, Amman, Jordan.
| |
Collapse
|
44
|
Proctor CR, Taggart MG, O'Hagan BM, McCarron PA, McCarthy RR, Ternan NG. Furanone loaded aerogels are effective antibiofilm therapeutics in a model of chronic Pseudomonas aeruginosa wound infection. Biofilm 2023; 5:100128. [PMID: 37223215 PMCID: PMC10200818 DOI: 10.1016/j.bioflm.2023.100128] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/25/2023] Open
Abstract
Almost 80% of chronic wounds have a bacterial biofilm present. These wound biofilms are caused by a range of organisms and are often polymicrobial. Pseudomonas aeruginosa is one of the most common causative organisms in wound infections and readily forms biofilms in wounds. To coordinate this, P. aeruginosa uses a process known as quorum sensing. Structural homologues of the quorum sensing signalling molecules have been used to disrupt this communication and prevent biofilm formation by Pseudomonas. However, these compounds have not yet reached clinical use. Here, we report the production and characterisation of a lyophilised PVA aerogel for use in delivering furanones to wound biofilms. PVA aerogels successfully release a model antimicrobial and two naturally occurring furanones in an aqueous environment. Furanone loaded aerogels inhibited biofilm formation in P. aeruginosa by up to 98.80%. Further, furanone loaded aerogels successfully reduced total biomass of preformed biofilms. Treatment with a sotolon loaded aerogel yielded a 5.16 log reduction in viable biofilm bound cells in a novel model of chronic wound biofilm, equivalent to the current wound therapy Aquacel AG. These results highlight the potential utility of aerogels in drug delivery to infected wounds and supports the use of biofilm inhibitory compounds as wound therapeutics.
Collapse
Affiliation(s)
- Chris R. Proctor
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Northern Ireland, UK
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Northern Ireland, UK
- Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Megan G. Taggart
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Northern Ireland, UK
| | - Barry M.G. O'Hagan
- Genomic Medicine Research Group, School of Biomedical Sciences, Ulster University, Northern Ireland, UK
| | - Paul A. McCarron
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Northern Ireland, UK
| | - Ronan R. McCarthy
- Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Nigel G. Ternan
- Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, Ulster University, Northern Ireland, UK
| |
Collapse
|
45
|
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
|
46
|
Moiketsi BN, Makale KPP, Rantong G, Rahube TO, Makhzoum A. Potential of Selected African Medicinal Plants as Alternative Therapeutics against Multi-Drug-Resistant Bacteria. Biomedicines 2023; 11:2605. [PMID: 37892979 PMCID: PMC10604549 DOI: 10.3390/biomedicines11102605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/04/2023] [Accepted: 09/18/2023] [Indexed: 10/29/2023] Open
Abstract
Antimicrobial resistance is considered a "One-Health" problem, impacting humans, animals, and the environment. The problem of the rapid development and spread of bacteria resistant to multiple antibiotics is a rising global health threat affecting both rich and poor nations. Low- and middle-income countries are at highest risk, in part due to the lack of innovative research on the surveillance and discovery of novel therapeutic options. Fast and effective drug discovery is crucial towards combatting antimicrobial resistance and reducing the burden of infectious diseases. African medicinal plants have been used for millennia in folk medicine to cure many diseases and ailments. Over 10% of the Southern African vegetation is applied in traditional medicine, with over 15 species being partially or fully commercialized. These include the genera Euclea, Ficus, Aloe, Lippia. And Artemisia, amongst many others. Bioactive compounds from indigenous medicinal plants, alone or in combination with existing antimicrobials, offer promising solutions towards overcoming multi-drug resistance. Secondary metabolites have different mechanisms and modes of action against bacteria, such as the inhibition and disruption of cell wall synthesis; inhibition of DNA replication and ATP synthesis; inhibition of quorum sensing; inhibition of AHL or oligopeptide signal generation, broadcasting, and reception; inhibition of the formation of biofilm; disruption of pathogenicity activities; and generation of reactive oxygen species. The aim of this review is to highlight some promising traditional medicinal plants found in Africa and provide insights into their secondary metabolites as alternative options in antibiotic therapy against multi-drug-resistant bacteria. Additionally, synergism between plant secondary metabolites and antibiotics has been discussed.
Collapse
Affiliation(s)
| | | | | | - Teddie O. Rahube
- Department of Biological Sciences and Biotechnology, Faculty of Science, Botswana International University of Science and Technology (BIUST), Private Bag 16, Palapye, Botswana; (B.N.M.); (K.P.P.M.); (G.R.)
| | - Abdullah Makhzoum
- Department of Biological Sciences and Biotechnology, Faculty of Science, Botswana International University of Science and Technology (BIUST), Private Bag 16, Palapye, Botswana; (B.N.M.); (K.P.P.M.); (G.R.)
| |
Collapse
|
47
|
Ghosh M, Raghav S, Ghosh P, Maity S, Mohela K, Jain D. Structural analysis of novel drug targets for mitigation of Pseudomonas aeruginosa biofilms. FEMS Microbiol Rev 2023; 47:fuad054. [PMID: 37771093 DOI: 10.1093/femsre/fuad054] [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: 05/30/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 09/30/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogen responsible for acute and chronic, hard to treat infections. Persistence of P. aeruginosa is due to its ability to develop into biofilms, which are sessile bacterial communities adhered to substratum and encapsulated in layers of self-produced exopolysaccharides. These biofilms provide enhanced protection from the host immune system and resilience towards antibiotics, which poses a challenge for treatment. Various strategies have been expended for combating biofilms, which involve inhibiting biofilm formation or promoting their dispersal. The current remediation approaches offer some hope for clinical usage, however, treatment and eradication of preformed biofilms is still a challenge. Thus, identifying novel targets and understanding the detailed mechanism of biofilm regulation becomes imperative. Structure-based drug discovery (SBDD) provides a powerful tool that exploits the knowledge of atomic resolution details of the targets to search for high affinity ligands. This review describes the available structural information on the putative target protein structures that can be utilized for high throughput in silico drug discovery against P. aeruginosa biofilms. Integrating available structural information on the target proteins in readily accessible format will accelerate the process of drug discovery.
Collapse
Affiliation(s)
- Moumita Ghosh
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Shikha Raghav
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Puja Ghosh
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Swagatam Maity
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Kavery Mohela
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Deepti Jain
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| |
Collapse
|
48
|
Mosallam FM, Abbas HA, Shaker GH, Gomaa SE. Alleviating the virulence of Pseudomonas aeruginosa and Staphylococcus aureus by ascorbic acid nanoemulsion. Res Microbiol 2023; 174:104084. [PMID: 37247797 DOI: 10.1016/j.resmic.2023.104084] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 05/31/2023]
Abstract
The high incidence of persistent multidrug resistant bacterial infections is a worldwide public health burden. Alternative strategies are required to deal with such issue including the use of drugs with anti-virulence activity. The application of nanotechnology to develop advanced Nano-materials that target quorum sensing regulated virulence factors is an attractive approach. Synthesis of ascorbic acid Nano-emulsion (ASC-NEs) and assessment of its activity in vitro against the virulence factors and its protective ability against pathogenesis as well as the effect against expression of quorum sensing genes of Pseudomonas aeruginosa and Staphylococcus aureus isolates. Ascorbic acid Nano-emulsion was characterized by DLS Zetasizer Technique, Zeta potential; Transmission Electron Microscopy (TEM) and Fourier transform infrared spectroscopy (FT-IR). The antibacterial activity of ASC-NEs was tested by the broth microdilution method and the activity of their sub-MIC against the expression of quorum sensing controlled virulence was investigated using phenotypic experiments and RT-PCR. The protective activity of ASC-NEs against P. aeruginosa as well as S. aureus pathogenesis was tested in vivo. Phenotypically, ASC-NEs had strong virulence inhibitory activity against the tested bacteria. The RT-PCR experiment showed that it exhibited significant QS inhibitory activity. The in vivo results showed that ASC-NEs protected against staphylococcal infection, however, it failed to protect mice against Pseudomonal infection. These results suggest the promising use of nanoformulations against virulence factors in multidrug resistant P. aeruginosa and S. aureus. However, further studies are required concerning the potential toxicity, clearance and phamacokinetics of the nanoformulations.
Collapse
Affiliation(s)
- Farag M Mosallam
- Drug Microbiology Lab., Drug Radiation Research Department, Biotechnology Division, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Hisham A Abbas
- Department of Microbiology and Immunology-Faculty of Pharmacy-Zagazig University, Zagazig, Egypt
| | - Ghada H Shaker
- Department of Microbiology and Immunology-Faculty of Pharmacy-Zagazig University, Zagazig, Egypt
| | - Salwa E Gomaa
- Department of Microbiology and Immunology-Faculty of Pharmacy-Zagazig University, Zagazig, Egypt
| |
Collapse
|
49
|
Shi X, Zhang R, Sand W, Mathivanan K, Zhang Y, Wang N, Duan J, Hou B. Comprehensive Review on the Use of Biocides in Microbiologically Influenced Corrosion. Microorganisms 2023; 11:2194. [PMID: 37764038 PMCID: PMC10535546 DOI: 10.3390/microorganisms11092194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/04/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
A microbiologically influenced corrosion (MIC) causes huge economic losses and serious environmental damage every year. The prevention and control measures for MIC mainly include physical, chemical, and biological methods. Among them, biocide application is the most cost-effective method. Although various biocides have their own advantages in preventing and treating MIC, most biocides have the problem of polluting the environment and increasing microorganism resistance. Therefore, it has stimulated the exploration of continuously developing new environmentally friendly and efficient biocides. In this review, the application advantages and research progress of various biocides used to prevent and control MIC are discussed. Also, this review provides a resource for the research and rational use of biocides regarding MIC mitigation and prevention.
Collapse
Affiliation(s)
- Xin Shi
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (X.S.); (W.S.); (K.M.); (Y.Z.); (N.W.); (J.D.); (B.H.)
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
- Open Studio for Marine Corrosion and Protection, Laoshan Laboratory, Qingdao 266237, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruiyong Zhang
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (X.S.); (W.S.); (K.M.); (Y.Z.); (N.W.); (J.D.); (B.H.)
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
- Open Studio for Marine Corrosion and Protection, Laoshan Laboratory, Qingdao 266237, China
- Institute of Marine Corrosion Protection, Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Sciences, Nanning 530007, China
| | - Wolfgang Sand
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (X.S.); (W.S.); (K.M.); (Y.Z.); (N.W.); (J.D.); (B.H.)
- Aquatic Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
- Institute of Biosciences, University of Mining and Technology, 09599 Freiberg, Germany
| | - Krishnamurthy Mathivanan
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (X.S.); (W.S.); (K.M.); (Y.Z.); (N.W.); (J.D.); (B.H.)
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
| | - Yimeng Zhang
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (X.S.); (W.S.); (K.M.); (Y.Z.); (N.W.); (J.D.); (B.H.)
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
- Open Studio for Marine Corrosion and Protection, Laoshan Laboratory, Qingdao 266237, China
| | - Nan Wang
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (X.S.); (W.S.); (K.M.); (Y.Z.); (N.W.); (J.D.); (B.H.)
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
- Open Studio for Marine Corrosion and Protection, Laoshan Laboratory, Qingdao 266237, China
| | - Jizhou Duan
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (X.S.); (W.S.); (K.M.); (Y.Z.); (N.W.); (J.D.); (B.H.)
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
- Open Studio for Marine Corrosion and Protection, Laoshan Laboratory, Qingdao 266237, China
- Institute of Marine Corrosion Protection, Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Sciences, Nanning 530007, China
| | - Baorong Hou
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; (X.S.); (W.S.); (K.M.); (Y.Z.); (N.W.); (J.D.); (B.H.)
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
- Open Studio for Marine Corrosion and Protection, Laoshan Laboratory, Qingdao 266237, China
- Institute of Marine Corrosion Protection, Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Sciences, Nanning 530007, China
| |
Collapse
|
50
|
Kauser A, Parisini E, Suarato G, Castagna R. Light-Based Anti-Biofilm and Antibacterial Strategies. Pharmaceutics 2023; 15:2106. [PMID: 37631320 PMCID: PMC10457815 DOI: 10.3390/pharmaceutics15082106] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/29/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Biofilm formation and antimicrobial resistance pose significant challenges not only in clinical settings (i.e., implant-associated infections, endocarditis, and urinary tract infections) but also in industrial settings and in the environment, where the spreading of antibiotic-resistant bacteria is on the rise. Indeed, developing effective strategies to prevent biofilm formation and treat infections will be one of the major global challenges in the next few years. As traditional pharmacological treatments are becoming inadequate to curb this problem, a constant commitment to the exploration of novel therapeutic strategies is necessary. Light-triggered therapies have emerged as promising alternatives to traditional approaches due to their non-invasive nature, precise spatial and temporal control, and potential multifunctional properties. Here, we provide a comprehensive overview of the different biofilm formation stages and the molecular mechanism of biofilm disruption, with a major focus on the quorum sensing machinery. Moreover, we highlight the principal guidelines for the development of light-responsive materials and photosensitive compounds. The synergistic effects of combining light-triggered therapies with conventional treatments are also discussed. Through elegant molecular and material design solutions, remarkable results have been achieved in the fight against biofilm formation and antibacterial resistance. However, further research and development in this field are essential to optimize therapeutic strategies and translate them into clinical and industrial applications, ultimately addressing the global challenges posed by biofilm and antimicrobial resistance.
Collapse
Affiliation(s)
- Ambreen Kauser
- Department of Biotechnology, Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia; (A.K.); (E.P.)
- Faculty of Materials Science and Applied Chemistry, Riga Technical University, Paula Valdena 3, LV-1048 Riga, Latvia
| | - Emilio Parisini
- Department of Biotechnology, Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia; (A.K.); (E.P.)
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Giulia Suarato
- Istituto di Elettronica e di Ingegneria dell’Informazione e delle Telecomunicazioni, Consiglio Nazionale delle Ricerche, CNR-IEIIT, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Rossella Castagna
- Department of Biotechnology, Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia; (A.K.); (E.P.)
- Dipartimento di Chimica, Materiali e Ingegneria Chimica “G. Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| |
Collapse
|