1
|
Deo R, Lakra U, Ojha M, Nigam VK, Sharma SR. Exopolysaccharides in microbial interactions: signalling, quorum sensing, and community dynamics. Nat Prod Res 2025; 39:3224-3239. [PMID: 39311333 DOI: 10.1080/14786419.2024.2405867] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/24/2024] [Accepted: 09/12/2024] [Indexed: 05/31/2025]
Abstract
Microbial interactions within diverse ecosystems are intricately governed by the dynamic interplay of exopolysaccharides (EPSs) produced by microorganisms. This review delves into the multifaceted roles of EPS in microbial signalling, quorum sensing (QS), and community dynamics, highlighting their significance in orchestrating cooperative behaviours and shaping community structures. EPSs serve as pivotal signalling molecules, influencing chemical communication and promoting intricate interactions among microorganisms. The integration of EPS into QS mechanisms adds an additional layer of complexity, allowing microorganisms to assess population density and synchronise communal responses. Furthermore, EPSs actively contribute to community dynamics by influencing spatial organisation, adhesion, and resistance to environmental stressors. By providing comprehensive knowledge of EPS dynamics, this review offers valuable insights into microbial ecology, serving as a foundational resource for future research. It will benefit the research community by advancing our understanding of microbial ecosystems, with broad applications in biotechnology, environmental science, and beyond.
Collapse
Affiliation(s)
- Rachna Deo
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Usha Lakra
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Mihir Ojha
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Vinod Kumar Nigam
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Shubha Rani Sharma
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
| |
Collapse
|
2
|
Datta S, Singh V, Nag S, Roy DN. Marine-Derived Cytosine Arabinoside (Ara-C) Inhibits Biofilm Formation by Inhibiting PEL Operon Proteins (Pel A and Pel B) of Pseudomonas aeruginosa: An In Silico Approach. Mol Biotechnol 2025; 67:1924-1938. [PMID: 38739212 DOI: 10.1007/s12033-024-01169-8] [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: 12/05/2023] [Accepted: 04/03/2024] [Indexed: 05/14/2024]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is a gram-negative biofilm-forming opportunistic human pathogen whose vital mechanism is biofilm formation for better survival. PelA and PelB proteins of the PEL operon are essential for bacterial-synthesized pellicle polysaccharide (PEL), which is a vital structural component of the biofilm. It helps in adherence of biofilm on the surface and maintenance of cell-to-cell interactions and with other matrix components. Here, in-silico molecular docking and simulation studies were performed against PelA and PelB using ten natural bioactive compounds, individually [podocarpic acids, ferruginol, scopadulcic acid B, pisiferic acid, metachromin A, Cytarabine (cytosine arabinoside; Ara-C), ursolic acid, oleanolic acid, maslinic acid, and betulinic acid], those have already been established as anti-infectious compounds. The results obtained from AutoDock and Glide-Schordinger stated that a marine-derived cytosine arabinoside (Ara-C) among the ten compounds binds active sites of PelA and PelB, exhibiting strong binding affinity [Trp224 (hydrogen), Ser219 (polar), Val234 (hydrophobic) for PelA; Leu365 and Glu389 (hydrogen), Gln366 (polar) for PelB] with high negative binding energy - 5.518 kcal/mol and - 6.056 kcal/mol, respectively. The molecular dynamic and simulation studies for 100 ns showed the MMGBSA binding energy scores are - 16.4 kcal/mol (Ara-C with PelA), and - 22.25 kcal/mol (Ara-C with PelB). Further, ADME/T studies indicate the IC50 values of AraC are 6.10 mM for PelA and 18.78 mM for PelB, which is a comparatively very low dose. The zero violation of Lipinski's Rule of Five further established that Ara-C is a good candidate for drug development. Thus, Ara-C could be considered a potent anti-biofilm compound against PEL operon-dependent biofilm formation of P. aeruginosa.
Collapse
Affiliation(s)
- Susmita Datta
- Department of Chemical Engineering, National Institute of Technology - Agartala, Agartala, Tripura, India
| | - Vishal Singh
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be university), Bhubaneswar, Odisha, India
| | - Soma Nag
- Department of Chemical Engineering, National Institute of Technology - Agartala, Agartala, Tripura, India.
| | - Dijendra Nath Roy
- Department of Biotechnology, National Institute of Technology - Raipur, Raipur, Chhattisgarh, India.
| |
Collapse
|
3
|
Van Loon JC, Le Mauff F, Vargas MA, Gilbert S, Pfoh R, Morrison ZA, Razvi E, Nitz M, Sheppard DC, Howell PL. Structural and functional analysis of Pseudomonas aeruginosa PelA provides insight into the modification of the Pel exopolysaccharide. J Biol Chem 2025; 301:108432. [PMID: 40120681 PMCID: PMC12022489 DOI: 10.1016/j.jbc.2025.108432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025] Open
Abstract
A major biofilm matrix determinant of Pseudomonas aeruginosa is the partially deacetylated α-1,4 linked N-acetylgalactosamine polymer, Pel. After synthesis and transport of the GalNAc polysaccharide across the inner membrane, PelA partially deacetylates and hydrolyzes Pel before its export out of the cell via PelB. While the Pel modification and export proteins are known to interact in the periplasm, it is unclear how the interaction of PelA and PelB coordinates these processes. To determine how PelA modifies the polymer, we determined its structure to 2.1 Å and found a unique arrangement of four distinct domains. We have shown previously that the hydrolase domain exhibits endo-α-1,4-N-acetylgalactosaminidase activity. Characterization of the deacetylase domain revealed that PelA is the founding member of a new carbohydrate esterase family, CE21. Further, we found that the PelAB interaction enhances the deacetylation of N-acetylgalactosamine oligosaccharides. Using the PelA structure in conjunction with AlphaFold2 modeling of the PelAB complex, we propose a model wherein PelB guides Pel to the deacetylase domain of PelA and subsequently to the porin domain of PelB for export. Perturbation or loss of the PelAB interaction would result in less efficient deacetylation and potentially increased Pel hydrolysis. In PelA homologs across many phyla, the predicted structure and active sites are conserved, suggesting a common modification mechanism in Gram-negative bacterial species containing a functional pel operon.
Collapse
Affiliation(s)
- Jaime C Van Loon
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - François Le Mauff
- Department of Microbiology and Immunology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Infectious Disease and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada; GlycoNET Integrated Services, Microbial Glycomic Node, Montreal, Quebec, Canada
| | - Mario A Vargas
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stephanie Gilbert
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Roland Pfoh
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Zachary A Morrison
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Erum Razvi
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Mark Nitz
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Donald C Sheppard
- Department of Microbiology and Immunology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Infectious Disease and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada
| | - P Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
4
|
Azimzadeh M, Greco G, Farmani A, Nourian A, Pourhajibagher M, Taherkhani A, Alikhani MY, Bahador A. Biofilm inhibition of multidrug-resistant Pseudomonas aeruginosa using green-synthesized silver nanoparticles and colistin. Sci Rep 2025; 15:14993. [PMID: 40301384 PMCID: PMC12041517 DOI: 10.1038/s41598-025-00005-6] [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: 01/06/2025] [Accepted: 04/24/2025] [Indexed: 05/01/2025] Open
Abstract
We aimed to investigate the synergistic effects of colistin and green-synthesized silver nanoparticles on the biofilm formation and expression of Quorum Sensing regulated and related genes in clinical isolates of P. aeruginosa. Ten clinical P. aeruginosa isolates collected from patients with burn wound infections were investigated. The antibiotic sensitivity pattern of the isolates was determined using disk diffusion and microbroth dilution tests. The silver nanoparticles (AgNPs) were synthesized using propolis and characterized. The microtiter plate method and scanning electron microscopy (SEM) were used to evaluate the synergistic effects of colistin and silver nanoparticles combination (AgNPs@CL) on the inhibition of biofilm formation. The effect of AgNPs@CL on the expression of genes controlled by QS was evaluated using RT-PCR. All isolates were strong biofilm formers. Confronting AgNPs@CL, all isolates were either synergistic or additive and effectively decrease the minimum inhibitory concentration (MIC) and minimum biofilm inhibitory concentration (MBIC) values of Carbapenem-Resistant P. aeruginosa (CRPA) isolates. The SEM analysis corroborated the enhanced biofilm inhibition observed with the combined treatment compared to individual AgNPs or colistin treatments. When exposed to AgNPs@CL, the expression levels of lasI, lasR, rhlI, rhlR, pelA, and pslA genes significantly decreased in P. aeruginosa ATCC 27,853 and clinical isolate No. #354, which displayed synergistic activity. In contrast, with additive activity, clinical isolate No. #30 showed no significant decrease. Targeting critical components of QS could effectively inhibit biofilm production. The results of our study suggest AgNPs@CL as an auxiliary to antibiotic therapy.
Collapse
Affiliation(s)
- Masoud Azimzadeh
- Department of Microbiology, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Grazia Greco
- Department of Veterinary Medicine, University of Bari "Aldo Moro", Valenzano, Bari, 70010, Italy
| | - Abbas Farmani
- Dental Implants Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Nourian
- Department of Pathobiology, Bu-Ali Sina University, Hamedan, Iran
| | - Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Taherkhani
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Yousef Alikhani
- Infectious Disease Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Abbas Bahador
- Department of Microbiology, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Reichhardt C, Matwichuk ML, Lewerke LT, Jacobs HM, Yan J, Parsek MR. Non-disruptive matrix turnover is a conserved feature of biofilm aggregate growth in paradigm pathogenic species. mBio 2025; 16:e0393524. [PMID: 39982068 PMCID: PMC11898600 DOI: 10.1128/mbio.03935-24] [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: 12/19/2024] [Accepted: 01/23/2025] [Indexed: 02/22/2025] Open
Abstract
Bacteria form multicellular aggregates called biofilms. A crucial component of these aggregates is a protective matrix that holds the community together. Biofilm matrix composition varies depending upon bacterial species but typically includes exopolysaccharides (EPS), proteins, and extracellular DNA. Pseudomonas aeruginosa is a model organism for the study of biofilms, and in non-mucoid biofilms, it uses the structurally distinct EPS Psl and Pel, the EPS-binding protein CdrA, and eDNA as key matrix components. An interesting phenomenon that we and others have observed is that the periphery of a biofilm aggregate can be EPS-rich and contain very few cells. In this study, we investigated two possible models of assembly and dynamics of this EPS-rich peripheral region: (i) newly synthesized EPS is inserted and incorporated into the existing EPS-rich region at the periphery during biofilm aggregate growth or (ii) EPS is continuously turned over and newly synthesized EPS is deposited at the outermost edge of the aggregate. Our results support the latter model. Specifically, we observed that new EPS is continually deposited at the aggregate periphery, which is necessary for continued aggregate growth but not aggregate stability. We made similar observations in another paradigm biofilm-forming species, Vibrio cholerae. This pattern of deposition raises the question of how EPS is retained. Specifically, for P. aeruginosa biofilms, the matrix adhesin CdrA is thought to retain EPS. However, current thinking is that cell-associated CdrA is responsible for this retention, and it is not clear how CdrA might function in the relatively cell-free aggregate periphery. We observed that CdrA is enzymatically degraded during aggregate growth without negatively impacting biofilm stability and that cell-free CdrA can partially maintain aggregation and Psl retention. Overall, this study shows that the matrix of P. aeruginosa biofilms undergoes both continuous synthesis of matrix material and matrix turnover to accommodate biofilm aggregate growth and that cell-free matrix can at least partially maintain biofilm aggregation and EPS localization. Furthermore, our similar observations for V. cholerae biofilms suggest that our findings may represent basic principles of aggregate assembly in bacteria. IMPORTANCE Here, we show that, to accommodate growing cellular biomass, newly produced Psl is deposited over existing Psl at the periphery of biofilm aggregates. We demonstrated that V. cholerae employs a similar mechanism with its biofilm matrix EPS, VPS. In addition, we found that the protease LasB is present in the biofilm matrix, resulting in degradation of CdrA to lower molecular weight cell-free forms. We then show that the released forms of CdrA are retained in the matrix and remain functional. Together, our findings support that the P. aeruginosa biofilm matrix is dynamic during the course of aggregate growth and that other species may employ similar mechanisms to remodel their matrix.
Collapse
Affiliation(s)
- Courtney Reichhardt
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | | | - Lincoln T. Lewerke
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Holly M. Jacobs
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Jing Yan
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - Matthew R. Parsek
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
6
|
Carey JN, Lamont S, Wozniak DJ, Dandekar AA, Parsek MR. Quorum sensing regulation of Psl polysaccharide production by Pseudomonas aeruginosa. J Bacteriol 2024; 206:e0031224. [PMID: 39530727 PMCID: PMC11656772 DOI: 10.1128/jb.00312-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Pseudomonas aeruginosa is a common opportunistic pathogen and a model organism for studying bacterial sociality. A social behavior of P. aeruginosa that is critical for its success as a pathogen is its ability to form protective biofilms. Many of P. aeruginosa's social phenotypes are regulated by quorum sensing-a type of cell-cell communication that allows bacteria to respond to population density. Although biofilm formation is known to be affected by quorum sensing, evidence for direct regulation of biofilm production by quorum regulators has remained elusive. In this work, we show that production of the major biofilm matrix polysaccharide Psl in P. aeruginosa PAO1 is regulated by the quorum regulators LasR and RhlR in stationary-phase cultures. Secretion of Psl into the culture medium requires LasR, RhlR, and the quorum signal molecules N-3-oxo-dodecanoyl-homoserine lactone and N-butanoyl homoserine lactone. Psl production in strains unable to synthesize the homoserine lactone signals can be restored by exogenous introduction of the signal molecules. We found that LasR and RhlR perform different roles in the regulation of Psl production: LasR acts at the promoter of the psl operon and activates transcription of the Psl biosynthetic genes, while RhlR activates translation of the psl transcripts. This work contributes to our understanding of the overlapping but distinct functions of the Las and Rhl quorum-sensing systems and implicates both in the direct regulation of biofilm matrix production.IMPORTANCEPseudomonas aeruginosa biofilms are responsible for many treatment-resistant infections in humans. Many cooperative behaviors in P. aeruginosa are controlled by quorum sensing, but evidence for a direct role of quorum sensing in the regulation of biofilm matrix production has been scant. In this work, we show that the Las and Rhl quorum-sensing systems have distinct roles in regulating production of the matrix polysaccharide Psl and that this regulation happens at the level of transcription (Las) and translation (Rhl) of the psl operon. These findings deepen our understanding of overlapping functions of Las and Rhl quorum sensing and the complex regulation of biofilm development in P. aeruginosa.
Collapse
Affiliation(s)
- Jeffrey N. Carey
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Sabrina Lamont
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Daniel J. Wozniak
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Ajai A. Dandekar
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Matthew R. Parsek
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
7
|
Maryam S, Krukiewicz K. Sweeten the pill: Multi-faceted polysaccharide-based carriers for colorectal cancer treatment. Int J Biol Macromol 2024; 282:136696. [PMID: 39437958 DOI: 10.1016/j.ijbiomac.2024.136696] [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: 07/05/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Colorectal cancer (CRC) ranks as the second deadliest cancer globally and the third most common malignant tumor. While surgery remains the primary treatment for CRC, alternative therapies such as chemotherapy, molecular targeted therapy, and immunotherapy are also commonly used. The significant side effects and toxicity of conventional drugs drive the search for novel targeted therapies, including the design of advanced drug delivery systems. Polysaccharide-based biopolymers, with their low toxicity, non-immunogenic behavior, synergistic interactions with other biopolymers, and tissue and cell compatibility, emerge as excellent drug carriers for this application. This review aims to provide an in-depth overview of recent advancements in developing polysaccharide-based biopolymeric carriers for anticancer compounds in the treatment of CRC. We highlight the multifunctional nature of polysaccharides, showcasing their potential as standalone drug carriers or as integral components of intelligent robotic devices for biomedical therapeutic applications. In addition to exploring the opportunities for using carbohydrate polymers in CRC treatment, we address the challenges and failures that may limit their applicability in biomedical research, as well as summarize the recent preclinical and clinical trials, resulting in several commercialization attempts. This comprehensive overview critically summarizes the potential of polysaccharide-based biomaterials in CRC treatment.
Collapse
Affiliation(s)
- Sajida Maryam
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, Gliwice, Poland; Joint Doctoral School, Silesian University of Technology, Gliwice, Poland
| | - Katarzyna Krukiewicz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, Gliwice, Poland; Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Gliwice, Poland.
| |
Collapse
|
8
|
Zhang Y, Zhang N, Li T, Jing L, Wang Y, Ge W. Risk factors for Pseudomonas aeruginosa isolation in chronic obstructive pulmonary disease: a systematic review and meta-analysis. BMC Pulm Med 2024; 24:521. [PMID: 39425103 PMCID: PMC11487921 DOI: 10.1186/s12890-024-03309-x] [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: 12/27/2023] [Accepted: 09/27/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Pseudomonas aeruginosa (PA) isolation in patients with chronic obstructive pulmonary disease (COPD) has been associated with a poor prognosis. This meta-analysis aimed to determine significant risk factors for PA isolation among patients with COPD. METHODS A systematic literature retrieval from PubMed, Embase, Web of Science and China National Knowledge Infrastructure (CNKI) was conducted, including studies from January 2003 to September 2024. Case-control and cohort studies exploring the risk factors for PA isolation in patients with COPD were included in this analysis. A random-effects model was applied to estimate the pooled adjusted odds ratio (paOR) or hazard ratio (paHR) with the corresponding 95% confidence intervals (CI). RESULTS Thirteen eligible studies with a total of 25,802 participants were included in this meta-analysis. Prior systemic steroid therapy (paOR: 2.67; 95% CI: 1.29-5.53; P = 0.008), previous antibiotic treatment (paOR: 2.83; 95% CI: 1.14-6.97; P = 0.02), high "Body mass index, airflow Obstruction, Dyspnea, Exercise capacity" (BODE) index (paOR: 4.13; 95% CI: 1.67-10.23; P = 0.002), 6-min walking distance (6MWD) < 250 m (paOR: 4.27; 95% CI: 2.59-7.01; P < 0.001), COPD assessment test (CAT) score > 20 points (paOR: 2.49; 95% CI: 1.46-4.23; P = 0.001), hypoproteinemia (paOR: 2.62; 95%CI: 1.32-5.19; P = 0.006), hospitalizations in the previous year (paOR: 3.74; 95%CI: 1.22-11.49; P = 0.021), Bronchiectasis (paOR = 4.81; 95% CI: 3.66-6.33; P < 0.001) and prior PA isolation (paOR: 16.39; 95% CI: 7.65-35.10; P < 0.001) were associated with PA isolation in patients with COPD. CONCLUSIONS Our study identified nine risk factors associated with an increased risk of PA isolation in COPD patients. These findings are significant for the early identification of patients at risk for PA isolation, which might contribute to reducing mortality and improving clinical outcomes.
Collapse
Affiliation(s)
- Yuyu Zhang
- Department of General Practice, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi , 710032, China
| | - Nini Zhang
- Department of Pediatrics, The Second Hospital of Yulin, Yulin, Shaanxi, 719000, China
| | - Tingting Li
- Department of General Practice, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi , 710032, China
| | - Lanrui Jing
- Department of General Practice, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi , 710032, China
| | - Yu Wang
- Department of General Practice, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi , 710032, China
| | - Wei Ge
- Department of General Practice, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi , 710032, China.
| |
Collapse
|
9
|
Ragupathi H, Pushparaj MM, Gopi SM, Govindarajan DK, Kandaswamy K. Biofilm matrix: a multifaceted layer of biomolecules and a defensive barrier against antimicrobials. Arch Microbiol 2024; 206:432. [PMID: 39402397 DOI: 10.1007/s00203-024-04157-3] [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: 08/09/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 11/10/2024]
Abstract
Bacterial cells often exist in the form of sessile aggregates known as biofilms, which are polymicrobial in nature and can produce slimy Extracellular Polymeric Substances (EPS). EPS is often referred to as a biofilm matrix and is a heterogeneous mixture of various biomolecules such as polysaccharides, proteins, and extracellular DNA/RNA (eDNA/RNA). In addition, bacteriophage (phage) was also found to be an integral component of the matrix and can serve as a protective barrier. In recent years, the roles of proteins, polysaccharides, and phages in the virulence of biofilms have been well studied. However, a mechanistic understanding of the release of such biomolecules and their interactions with antimicrobials requires a thorough review. Therefore, this article critically reviews the various mechanisms of release of matrix polymers. In addition, this article also provides a contemporary understanding of interactions between various biomolecules to protect biofilms against antimicrobials. In summary, this article will provide a thorough understanding of the functions of various biofilm matrix molecules.
Collapse
Affiliation(s)
- Harini Ragupathi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Mahamahima Muthuswamy Pushparaj
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Sarves Mani Gopi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Deenadayalan Karaiyagowder Govindarajan
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Drive, 637371, Singapore, Singapore
| | - Kumaravel Kandaswamy
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India.
| |
Collapse
|
10
|
da Cruz Nizer WS, Allison KN, Adams ME, Vargas MA, Ahmed D, Beaulieu C, Raju D, Cassol E, Howell PL, Overhage J. The role of exopolysaccharides Psl and Pel in resistance of Pseudomonas aeruginosa to the oxidative stressors sodium hypochlorite and hydrogen peroxide. Microbiol Spectr 2024; 12:e0092224. [PMID: 39194290 PMCID: PMC11448232 DOI: 10.1128/spectrum.00922-24] [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: 04/11/2024] [Accepted: 07/01/2024] [Indexed: 08/29/2024] Open
Abstract
Pseudomonas aeruginosa is well-known for its antimicrobial resistance and the ability to survive in harsh environmental conditions due to an abundance of resistance mechanisms, including the formation of biofilms and the production of exopolysaccharides. Exopolysaccharides are among the major components of the extracellular matrix in biofilms and aggregates of P. aeruginosa. Although their contribution to antibiotic resistance has been previously shown, their roles in resistance to oxidative stressors remain largely elusive. Here, we studied the function of the exopolysaccharides Psl and Pel in the resistance of P. aeruginosa to the commonly used disinfectants and strong oxidizing agents NaOCl and H2O2. We observed that the simultaneous inactivation of Psl and Pel in P. aeruginosa PAO1 mutant strain ∆pslA pelF resulted in a significant increase in susceptibility to both NaOCl and H2O2. Further analyses revealed that Pel is more important for oxidative stress resistance in P. aeruginosa and that the form of Pel (i.e., cell-associated or cell-free) did not affect NaOCl susceptibility. Additionally, we show that Psl/Pel-negative strains are protected against oxidative stress in co-culture biofilms with P. aeruginosa PAO1 WT. Taken together, our results demonstrate that the EPS matrix and, more specifically, Pel exhibit protective functions against oxidative stressors such as NaOCl and H2O2 in P. aeruginosa. IMPORTANCE Biofilms are microbial communities of cells embedded in a self-produced polymeric matrix composed of polysaccharides, proteins, lipids, and extracellular DNA. Biofilm bacteria have been shown to possess unique characteristics, including increased stress resistance and higher antimicrobial tolerance, leading to failures in bacterial eradication during chronic infections or in technical settings, including drinking and wastewater industries. Previous studies have shown that in addition to conferring structure and stability to biofilms, the polysaccharides Psl and Pel are also involved in antibiotic resistance. This work provides evidence that these biofilm matrix components also contribute to the resistance of Pseudomonas aeruginosa to oxidative stressors including the widely used disinfectant NaOCl. Understanding the mechanisms by which bacteria escape antimicrobial agents, including strong oxidants, is urgently needed in the fight against antimicrobial resistance and will help in developing new strategies to eliminate resistant strains in any environmental, industrial, and clinical setting.
Collapse
Affiliation(s)
| | - Kira N. Allison
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Madison E. Adams
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Mario A. Vargas
- Program in Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Duale Ahmed
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Carole Beaulieu
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Deepa Raju
- Program in Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - P. Lynne Howell
- Program in Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Joerg Overhage
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
11
|
Gong F, Xin S, Liu X, He C, Yu X, Pan L, Zhang S, Gao H, Xu J. Multiple biological characteristics and functions of intestinal biofilm extracellular polymers: friend or foe? Front Microbiol 2024; 15:1445630. [PMID: 39224216 PMCID: PMC11367570 DOI: 10.3389/fmicb.2024.1445630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The gut microbiota is vital to human health, and their biofilms significantly impact intestinal immunity and the maintenance of microbial balance. Certain pathogens, however, can employ biofilms to elude identification by the immune system and medical therapy, resulting in intestinal diseases. The biofilm is formed by extracellular polymorphic substances (EPS), which shield microbial pathogens from the host immune system and enhance its antimicrobial resistance. Therefore, investigating the impact of extracellular polysaccharides released by pathogens that form biofilms on virulence and defence mechanisms is crucial. In this review, we provide a comprehensive overview of current pathogenic biofilm research, deal with the role of extracellular polymers in the formation and maintenance of pathogenic biofilm, and elaborate different prevention and treatment strategies to provide an innovative approach to the treatment of intestinal pathogen-based diseases.
Collapse
Affiliation(s)
- Fengrong Gong
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaohui Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinyi Yu
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Luming Pan
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Sitian Zhang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Han Gao
- Department of Clinical Laboratory, Aerospace Center Hospital, Beijing, China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
David A, Tahrioui A, Tareau AS, Forge A, Gonzalez M, Bouffartigues E, Lesouhaitier O, Chevalier S. Pseudomonas aeruginosa Biofilm Lifecycle: Involvement of Mechanical Constraints and Timeline of Matrix Production. Antibiotics (Basel) 2024; 13:688. [PMID: 39199987 PMCID: PMC11350761 DOI: 10.3390/antibiotics13080688] [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: 07/01/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen causing acute and chronic infections, especially in immunocompromised patients. Its remarkable adaptability and resistance to various antimicrobial treatments make it difficult to eradicate. Its persistence is enabled by its ability to form a biofilm. Biofilm is a community of sessile micro-organisms in a self-produced extracellular matrix, which forms a scaffold facilitating cohesion, cell attachment, and micro- and macro-colony formation. This lifestyle provides protection against environmental stresses, the immune system, and antimicrobial treatments, and confers the capacity for colonization and long-term persistence, often characterizing chronic infections. In this review, we retrace the events of the life cycle of P. aeruginosa biofilm, from surface perception/contact to cell spreading. We focus on the importance of extracellular appendages, mechanical constraints, and the kinetics of matrix component production in each step of the biofilm life cycle.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sylvie Chevalier
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, CBSA UR 4312, F-76000 Rouen, France
| |
Collapse
|
13
|
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
|
14
|
Beaussart A, Paiva TO, Geiger CJ, Baker AE, O'Toole GA, Dufrêne YF. Atomic force microscopy analysis of Pel polysaccharide- and type IV pili-mediated adhesion of Pseudomonas aeruginosa PA14 to an abiotic surface. NANOSCALE 2024; 16:12134-12141. [PMID: 38832761 DOI: 10.1039/d4nr01415d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Type IV pili (TFP) contribute to the ability of microbes such as Pseudomonas aeruginosa to engage with and move across surfaces. We reported previously that P. aeruginosa TFP generate retractive forces of ∼30 pN and provided indirect evidence that TFP-mediated surface attachment was enhanced in the presence of the Pel polysaccharide. Here, we use different mutants defective in flagellar, Pel production or TFP production - alone or in combination - to decipher the relative contribution of these biofilm-promoting factors for P. aeruginosa adhesion. By means of atomic force microscopy (AFM), we show that mutating the flagellum (ΔflgK mutant) results in an increase in Pel polysaccharide production, but this increase in Pel does not result in an increase in surface adhesive properties compared to those previously described for the WT strain. By blocking Pel production in the ΔflgK mutant (ΔflgKΔpel), we directly show that TFP play a major role in the adhesion of the bacteria to hydrophobic AFM tips, but that the adhesion force is only slightly impaired by the absence of Pel. Inversely, performing single-cell force spectroscopy measurements with the mutant lacking TFP (ΔflgKΔpilA) reveals that the Pel can modulate the attachment of the bacteria to a hydrophobic substrate in a time-dependent manner. Finally, little adhesion was detected for the ΔflgKΔpilAΔpelA triple mutant, suggesting that both TFP and Pel polysaccharide make a substantial contribution to bacteria-substratum interaction events. Altogether, our data allow us to decipher the relative contribution of Pel and TFP in the early attachment by P. aeruginosa.
Collapse
Affiliation(s)
- Audrey Beaussart
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, L7.07.07, B-1348 Louvain-la-Neuve, Belgium.
| | - Telmo O Paiva
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, L7.07.07, B-1348 Louvain-la-Neuve, Belgium.
| | - Christopher J Geiger
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, USA.
| | - Amy E Baker
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, USA.
| | - George A O'Toole
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, USA.
| | - Yves F Dufrêne
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, L7.07.07, B-1348 Louvain-la-Neuve, Belgium.
| |
Collapse
|
15
|
Sarangi A, Singh SP, Das BS, Rajput S, Fatima S, Bhattacharya D. Mycobacterial biofilms: A therapeutic target against bacterial persistence and generation of antibiotic resistance. Heliyon 2024; 10:e32003. [PMID: 38882302 PMCID: PMC11176842 DOI: 10.1016/j.heliyon.2024.e32003] [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: 01/22/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/18/2024] Open
Abstract
Mycobacterium tuberculosis (M. tb) is the causative agent of Tuberculosis, one of the deadliest infectious diseases. According to the WHO Report 2023, in 2022, approximately 10.6 million people got infected with TB, and 1.6 million died. It has multiple antibiotics for treatment, but the major drawback of anti-tuberculosis therapy (ATT) is, its prolonged treatment duration. The major contributors to the lengthy treatment period are mycobacterial persistence and drug tolerance. Persistent M. tb is phenotypically drug tolerant and metabolically slow down which makes it difficult to be eliminated during ATT. These persisting bacteria are a huge reservoir of impending disease, waiting to get reactivated upon the onset of an immune compromising state. Directly Observed Treatment Short-course, although effective against replicating bacteria; fails to eliminate the drug-tolerant persisters making TB still the second-highest killer globally. There are different mechanisms for the development of drug-tolerant mycobacterial populations being investigated. Recently, the role of biofilms in the survival and host-evasion mechanism of persisters has come to light. Therefore, it is crucial to understand the mechanism of adaptation, survival and attainment of drug tolerance by persisting M. tb-populations, in order to design better immune responses and therapeutics for the effective elimination of these bacteria by reducing the duration of treatment and also circumvent the generation of drug-resistance to achieve the goal of global eradication of TB. This review summarizes the drug-tolerance mechanism and biofilms' role in providing a niche to dormant-M.tb. We also discuss methods of targeting biofilms to achieve sterile eradication of the mycobacteria and prevent its reactivation by achieving adequate immune responses.
Collapse
Affiliation(s)
- Ashirbad Sarangi
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Shashi Prakash Singh
- Vaccine and Gene Therapy Institute (VGTI) Oregon National Primate Research Centre (ONPRC) Oregon Health and Science University (OHSU) Beaverton, Oregon, USA
| | - Bhabani Shankar Das
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Sristi Rajput
- Departmental of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal, Madhya Pradesh, India
| | - Samreen Fatima
- UMass Chan Medical School, University of Massachusetts, Worcester, MA, USA
| | - Debapriya Bhattacharya
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
- Departmental of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal, Madhya Pradesh, India
| |
Collapse
|
16
|
Kabir RB, Ahsan T, Rahman MF, Jobayer M, Shamsuzzaman SM. Biofilm-producing and specific antibiotic resistance genes in Pseudomonas aeruginosa isolated from patients admitted to a tertiary care hospital, Bangladesh. IJID REGIONS 2024; 11:100369. [PMID: 38799795 PMCID: PMC11126856 DOI: 10.1016/j.ijregi.2024.100369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024]
Abstract
Objectives Biofilms are responsible for persistent infections and antimicrobial resistance. Pseudomonas aeruginosa was investigated with its ability to form biofilm by detecting genes responsible for producing biofilms and biofilm-specific antimicrobial resistance. The association between antibiotic resistance and biofilm was investigated. Methods This cross-sectional study was conducted from July 2017 to December 2018. A total of 446 samples (infected burn, surgical wounds, and endotracheal aspirate) were collected from admitted patients of Dhaka Medical College and Hospital, Bangladesh. P. aeruginosa was isolated and identified by biochemical tests and polymerase chain reaction. Biofilm production by tissue culture plate method followed by detection of biofilm-producing genes (pqsA, pslA, pslD, pslH, pelA, lasR) and biofilm-specific antibiotic resistance genes (ndvB, PA1874, PA1876, PA1877) by polymerase chain reaction were done. Antibiotic susceptibility test was carried out by disk diffusion method; for colistin agar dilution method of minimal inhibitory concentration was followed. Results Among 232 (52.02%) positive strains of P. aeruginosa, 24 (10.30%) produced biofilms in tissue culture plate. Among biofilm-producing genes, pqsA was the highest (79.17%). pslA and pelA were 70.83%, pslD 45.83%, pslH and lasR 37.5%. Among biofilm-specific antibiotic resistance genes, 16.67% were ndvB, and 8.33% were PA1874 and PA1877. Biofilm-forming strains were significantly resistant to colistin. Conclusions Detection of biofilm-forming genes may be a good tool for the evaluation of biofilm production, which will help in prompt and better management of chronic or device-associated infections.
Collapse
Affiliation(s)
| | - Tasnim Ahsan
- Department of Microbiology, Ibn Sina Medical College, Dhaka, Bangladesh
| | - Md. Faizur Rahman
- Department of Microbiology, Dhaka Medical College, Dhaka, Bangladesh
| | - Mohammad Jobayer
- Department of Microbiology, Dhaka Medical College, Dhaka, Bangladesh
| | - SM Shamsuzzaman
- Department of Microbiology, Dhaka Medical College, Dhaka, Bangladesh
| |
Collapse
|
17
|
Okurowska K, Monk PN, Karunakaran E. Increased tolerance to commonly used antibiotics in a Pseudomonas aeruginosa ex vivo porcine keratitis model. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001459. [PMID: 38739119 PMCID: PMC11165664 DOI: 10.1099/mic.0.001459] [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: 11/22/2023] [Accepted: 04/26/2024] [Indexed: 05/14/2024]
Abstract
Introduction. Bacterial keratitis, particularly caused by Pseudomonas aeruginosa, is challenging to treat because of multi-drug tolerance, often associated with the formation of biofilms. Antibiotics in development are typically evaluated against planktonic bacteria in a culture medium, which may not accurately represent the complexity of infections in vivo.Hypothesis/Gap Statement. Developing a reliable, economic ex vivo keratitis model that replicates some complexity of tissue infections could facilitate a deeper understanding of antibiotic efficacy, thus aiding in the optimization of treatment strategies for bacterial keratitis.Methodology. Here we investigated the efficacy of three commonly used antibiotics (gentamicin, ciprofloxacin and meropenem) against Pseudomonas aeruginosa cytotoxic strain PA14 and invasive strain PA01 using an ex vivo porcine keratitis model.Results. Both strains of P. aeruginosa were susceptible to the MIC of the three tested antibiotics. However, significantly higher concentrations were necessary to inhibit bacterial growth in the minimum biofilm eradication concentration (MBEC) assay, with both strains tolerating concentrations greater than 512 mg l-1 of meropenem. When MIC and higher concentrations than MBEC (1024 mg l-1) of antibiotics were applied, ciprofloxacin exhibited the highest potency against both P. aeruginosa strains, followed by meropenem, while gentamicin showed the least potency. Despite this, none of the antibiotic concentrations used effectively cleared the infection, even after 18 h of continuous exposure.Conclusions. Further exploration of antibiotic concentrations and aligning dosing with clinical studies to validate the model is needed. Nonetheless, our ex vivo porcine keratitis model could be a valuable tool for assessing antibiotic efficacy.
Collapse
Affiliation(s)
- Katarzyna Okurowska
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S10 2TN, UK
- National Institute for Health and Care Research, University of Leeds, Leeds LS2 9JT, UK
| | - Peter N. Monk
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Esther Karunakaran
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
18
|
Granton E, Brown L, Defaye M, Moazen P, Almblad H, Randall TE, Rich JD, Geppert A, Abdullah NS, Hassanabad MF, Hiroki CH, Farias R, Nguyen AP, Schubert C, Lou Y, Andonegui G, Iftinca M, Raju D, Vargas MA, Howell PL, Füzesi T, Bains J, Kurrasch D, Harrison JJ, Altier C, Yipp BG. Biofilm exopolysaccharides alter sensory-neuron-mediated sickness during lung infection. Cell 2024; 187:1874-1888.e14. [PMID: 38518773 DOI: 10.1016/j.cell.2024.03.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 01/04/2024] [Accepted: 03/01/2024] [Indexed: 03/24/2024]
Abstract
Infections of the lung cause observable sickness thought to be secondary to inflammation. Signs of sickness are crucial to alert others via behavioral-immune responses to limit contact with contagious individuals. Gram-negative bacteria produce exopolysaccharide (EPS) that provides microbial protection; however, the impact of EPS on sickness remains uncertain. Using genome-engineered Pseudomonas aeruginosa (P. aeruginosa) strains, we compared EPS-producers versus non-producers and a virulent Escherichia coli (E. coli) lung infection model in male and female mice. EPS-negative P. aeruginosa and virulent E. coli infection caused severe sickness, behavioral alterations, inflammation, and hypothermia mediated by TLR4 detection of the exposed lipopolysaccharide (LPS) in lung TRPV1+ sensory neurons. However, inflammation did not account for sickness. Stimulation of lung nociceptors induced acute stress responses in the paraventricular hypothalamic nuclei by activating corticotropin-releasing hormone neurons responsible for sickness behavior and hypothermia. Thus, EPS-producing biofilm pathogens evade initiating a lung-brain sensory neuronal response that results in sickness.
Collapse
Affiliation(s)
- Elise Granton
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Luke Brown
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Manon Defaye
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Inflammation Research Network, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Parisa Moazen
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Henrik Almblad
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Trevor E Randall
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Jacquelyn D Rich
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Andrew Geppert
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Nasser S Abdullah
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Inflammation Research Network, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Mortaza F Hassanabad
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Carlos H Hiroki
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Raquel Farias
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Angela P Nguyen
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Courtney Schubert
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Yuefei Lou
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Graciela Andonegui
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mircea Iftinca
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Inflammation Research Network, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Deepa Raju
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Mario A Vargas
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - P Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Tamás Füzesi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Cumming School of Medicine Optogenetics Core Facility, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jaideep Bains
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Krembil Research Institute, University Health Network, Toronto, ON, Canada.
| | - Deborah Kurrasch
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Joe Jonathan Harrison
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Biological Sciences, University of Calgary, Calgary, AB, Canada.
| | - Christophe Altier
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Inflammation Research Network, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| | - Bryan G Yipp
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
19
|
Subsomwong P, Teng W, Ishiai T, Narita K, Sukchawalit R, Nakane A, Asano K. Extracellular vesicles from Staphylococcus aureus promote the pathogenicity of Pseudomonas aeruginosa. Microbiol Res 2024; 281:127612. [PMID: 38244256 DOI: 10.1016/j.micres.2024.127612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/22/2024]
Abstract
Co-infections with Staphylococcus aureus and Pseudomonas aeruginosa are common in patients with chronic wounds, but little is known about their synergistic effect mediated by extracellular vesicles (EVs). In this study, we investigated the effect of EVs derived from S. aureus (SaEVs) on the pathogenicity of P. aeruginosa. By using lipophilic dye, we could confirm the fusion between SaEV and P. aeruginosa membranes. However, SaEVs did not alter the growth and antibiotic susceptible pattern of P. aeruginosa. Differential proteomic analysis between SaEV-treated and non-treated P. aeruginosa was performed, and the results revealed that lipopolysaccharide (LPS) biosynthesis protein in P. aeruginosa significantly increased after SaEV-treatment. Regarding this result, we also found that SaEVs promoted LPS production, biofilm formation, and expression of polysaccharide polymerization-related genes in P. aeruginosa. Furthermore, invasion of epithelial cells by SaEV-pretreated P. aeruginosa was enhanced. On the other hand, uptake of P. aeruginosa by RAW 264.7 macrophages was impaired after pretreatment P. aeruginosa with SaEVs. Proteomic analysis SaEVs revealed that SaEVs contain the proteins involving in host cell colonization, inhibition of host immune response, anti-phagocytosis of the macrophages, and protein translocation and iron uptake of S. aureus. In conclusion, SaEVs serve as a mediator that promote P. aeruginosa pathogenicity by enhancing LPS biosynthesis, biofilm formation, epithelial cell invasion, and macrophage uptake impairment.
Collapse
Affiliation(s)
- Phawinee Subsomwong
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Wei Teng
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Takahito Ishiai
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Kouji Narita
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan; Institute for Animal Experimentation, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Rojana Sukchawalit
- Laboratory of Biotechnology, Chulabhorn Research Institute, Lak Si, Bangkok, Thailand
| | - Akio Nakane
- Department of Biopolymer and Health Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Krisana Asano
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan; Department of Biopolymer and Health Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan.
| |
Collapse
|
20
|
Wang J, Wu X, Chen J, Gao T, Zhang Y, Yu N. Traditional Chinese medicine polysaccharide in nano-drug delivery systems: Current progress and future perspectives. Biomed Pharmacother 2024; 173:116330. [PMID: 38422656 DOI: 10.1016/j.biopha.2024.116330] [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: 11/16/2023] [Revised: 01/19/2024] [Accepted: 02/22/2024] [Indexed: 03/02/2024] Open
Abstract
Traditional Chinese medicine polysaccharides (TCMPs) have gained increasing attention in the field of nanomedicine due to their diverse biological activities and favorable characteristics as drug carriers, including biocompatibility, biodegradability, safety, and ease of modification. TCMPs-based nano-drug delivery systems (NDDSs) offer several advantages, such as evasion of reticuloendothelial system (RES) phagocytosis, protection against biomolecule degradation, enhanced drug bioavailability, and potent therapeutic effects. Therefore, a comprehensive review of the latest developments in TCMPs-based NDDSs and their applications in disease therapy is of great significance. This review provides an overview of the structural characteristics and biological activities of TCMPs relevant to carrier design, the strategies employed for constructing TCMPs-based NDDSs, and the versatile role of TCMPs in these systems. Additionally, current challenges and future prospects of TCMPs in NDDSs are discussed, aiming to provide valuable insights for future research and clinical translation.
Collapse
Affiliation(s)
- Juan Wang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xia Wu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jing Chen
- Department of Pharmaceutical Preparation, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ting Gao
- Department of Pharmaceutical Preparation, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yumei Zhang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China; Department of Chemistry, School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Na Yu
- Department of Pharmaceutical Preparation, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China; Department of Clinical Pharmacology, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
21
|
Deiss-Yehiely E, Dzordzorme AE, Loiselle ME, Yonker LM, Hammond PT. Carboxylated Nanoparticle Surfaces Enhance Association with Mucoid Pseudomonas aeruginosa Biofilms. ACS APPLIED MATERIALS & INTERFACES 2024; 16:14573-14582. [PMID: 38484043 PMCID: PMC10982939 DOI: 10.1021/acsami.3c18656] [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: 12/29/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 04/04/2024]
Abstract
Pseudomonas aeruginosa biofilms comprise three main polysaccharides: alginate, psl, and pel, which all imbue tolerance against exogenous antimicrobials. Nanoparticles (NPs) are an exciting new strategy to overcome the biofilm matrix for therapeutic delivery applications; however, zero existing FDA approvals for biofilm-specific NP formulations can be attributed to the complex interplay of physiochemical forces at the biofilm-NP interface. Here, we leverage a set of inducible, polysaccharide-specific, expressing isogenic P. aeruginosa mutants coupled with an assembled layer-by-layer NP (LbL NP) panel to characterize biofilm-NP interactions. When investigating these interactions using confocal microscopy, alginate-layered NPs associated more than dextran-sulfate-layered NPs with biofilms that had increased alginate production, including biofilms produced by mucoid P. aeruginosa isolates from people with cystic fibrosis. These differences were further confirmed in LbL NPs layered with polysaccharide- or hydrocarbon-based polymers with pendent carboxylate or sulfate functional groups. These data suggest carboxylated NP surfaces have enhanced interactions specifically with mucoid biofilms as compared to sulfated surfaces and lay the foundation for their inclusion as a design element for increasing biofilm-NP interactions and efficacious drug delivery.
Collapse
Affiliation(s)
- Elad Deiss-Yehiely
- Department
of Materials Science and Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Koch
Institute for Integrative Cancer Research, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Abigail E. Dzordzorme
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Maggie Elizabeth Loiselle
- Mucosal
Immunology and Biology Research Center, Division of Infectious Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department
of Pediatrics, Division of Infectious Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Lael M. Yonker
- Mucosal
Immunology and Biology Research Center, Division of Infectious Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department
of Pediatrics, Division of Infectious Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Harvard
Medical School, Boston, Massachusetts 02115, United States
| | - Paula T. Hammond
- Koch
Institute for Integrative Cancer Research, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
- Institute
for Soldier Nanotechnologies, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
22
|
Yaeger LN, Ranieri MRM, Chee J, Karabelas-Pittman S, Rudolph M, Giovannoni AM, Harvey H, Burrows LL. A genetic screen identifies a role for oprF in Pseudomonas aeruginosa biofilm stimulation by subinhibitory antibiotics. NPJ Biofilms Microbiomes 2024; 10:30. [PMID: 38521769 PMCID: PMC10960818 DOI: 10.1038/s41522-024-00496-7] [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: 09/06/2023] [Accepted: 03/05/2024] [Indexed: 03/25/2024] Open
Abstract
Biofilms are surface-associated communities of bacteria that grow in a self-produced matrix of polysaccharides, proteins, and extracellular DNA (eDNA). Sub-minimal inhibitory concentrations (sub-MIC) of antibiotics induce biofilm formation, potentially as a defensive response to antibiotic stress. However, the mechanisms behind sub-MIC antibiotic-induced biofilm formation are unclear. We show that treatment of Pseudomonas aeruginosa with multiple classes of sub-MIC antibiotics with distinct targets induces biofilm formation. Further, addition of exogenous eDNA or cell lysate failed to increase biofilm formation to the same extent as antibiotics, suggesting that the release of cellular contents by antibiotic-driven bacteriolysis is insufficient. Using a genetic screen for stimulation-deficient mutants, we identified the outer membrane porin OprF and the ECF sigma factor SigX as important. Similarly, loss of OmpA - the Escherichia coli OprF homolog - prevented sub-MIC antibiotic stimulation of E. coli biofilms. Our screen also identified the periplasmic disulfide bond-forming enzyme DsbA and a predicted cyclic-di-GMP phosphodiesterase encoded by PA2200 as essential for biofilm stimulation. The phosphodiesterase activity of PA2200 is likely controlled by a disulfide bond in its regulatory domain, and folding of OprF is influenced by disulfide bond formation, connecting the mutant phenotypes. Addition of reducing agent dithiothreitol prevented sub-MIC antibiotic biofilm stimulation. Finally, activation of a c-di-GMP-responsive promoter follows treatment with sub-MIC antibiotics in the wild-type but not an oprF mutant. Together, these results show that antibiotic-induced biofilm formation is likely driven by a signaling pathway that translates changes in periplasmic redox state into elevated biofilm formation through increases in c-di-GMP.
Collapse
Affiliation(s)
- Luke N Yaeger
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Michael R M Ranieri
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Jessica Chee
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Sawyer Karabelas-Pittman
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Madeleine Rudolph
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Alessio M Giovannoni
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Hanjeong Harvey
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Lori L Burrows
- Biochemistry and Biomedical Sciences and the Michael G. DeGroote Centre for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
23
|
Oladosu VI, Park S, Sauer K. Flip the switch: the role of FleQ in modulating the transition between the free-living and sessile mode of growth in Pseudomonas aeruginosa. J Bacteriol 2024; 206:e0036523. [PMID: 38436566 PMCID: PMC10955856 DOI: 10.1128/jb.00365-23] [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] [Indexed: 03/05/2024] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative, opportunistic pathogen causing chronic infections that are associated with the sessile/biofilm mode of growth rather than the free-living/planktonic mode of growth. The transcriptional regulator FleQ contributes to both modes of growth by functioning both as an activator and repressor and inversely regulating flagella genes associated with the planktonic mode of growth and genes contributing to the biofilm mode of growth. Here, we review findings that enhance our understanding of the molecular mechanism by which FleQ enables the transition between the two modes of growth. We also explore recent advances in the mechanism of action of FleQ to both activate and repress gene expression from a single promoter. Emphasis will be on the role of sigma factors, cyclic di-GMP, and the transcriptional regulator AmrZ in inversely regulating flagella and biofilm-associated genes and converting FleQ from a repressor to an activator.
Collapse
Affiliation(s)
- Victoria I. Oladosu
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
| | - Soyoung Park
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | - Karin Sauer
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
24
|
Dicks LMT, Vermeulen W. Bacteriophage-Host Interactions and the Therapeutic Potential of Bacteriophages. Viruses 2024; 16:478. [PMID: 38543843 PMCID: PMC10975011 DOI: 10.3390/v16030478] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 05/23/2024] Open
Abstract
Healthcare faces a major problem with the increased emergence of antimicrobial resistance due to over-prescribing antibiotics. Bacteriophages may provide a solution to the treatment of bacterial infections given their specificity. Enzymes such as endolysins, exolysins, endopeptidases, endosialidases, and depolymerases produced by phages interact with bacterial surfaces, cell wall components, and exopolysaccharides, and may even destroy biofilms. Enzymatic cleavage of the host cell envelope components exposes specific receptors required for phage adhesion. Gram-positive bacteria are susceptible to phage infiltration through their peptidoglycan, cell wall teichoic acid (WTA), lipoteichoic acids (LTAs), and flagella. In Gram-negative bacteria, lipopolysaccharides (LPSs), pili, and capsules serve as targets. Defense mechanisms used by bacteria differ and include physical barriers (e.g., capsules) or endogenous mechanisms such as clustered regularly interspaced palindromic repeat (CRISPR)-associated protein (Cas) systems. Phage proteins stimulate immune responses against specific pathogens and improve antibiotic susceptibility. This review discusses the attachment of phages to bacterial cells, the penetration of bacterial cells, the use of phages in the treatment of bacterial infections, and the limitations of phage therapy. The therapeutic potential of phage-derived proteins and the impact that genomically engineered phages may have in the treatment of infections are summarized.
Collapse
Affiliation(s)
- Leon M. T. Dicks
- Department of Microbiology, Stellenbosch University, Stellenbosch 7600, South Africa;
| | | |
Collapse
|
25
|
Sousa AM, Ferreira D, Rodrigues LR, Pereira MO. Aptamer-based therapy for fighting biofilm-associated infections. J Control Release 2024; 367:522-539. [PMID: 38295992 DOI: 10.1016/j.jconrel.2024.01.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/06/2024] [Accepted: 01/27/2024] [Indexed: 02/06/2024]
Abstract
Biofilms are key players in the pathogenesis of most of chronic infections associated with host tissue or fluids and indwelling medical devices. These chronic infections are hard to be treated due to the increased biofilms tolerance towards antibiotics in comparison to planktonic (or free living) cells. Despite the advanced understanding of their formation and physiology, biofilms continue to be a challenge and there is no standardized therapeutic approach in clinical practice to eradicate them. Aptamers offer distinctive properties, including excellent affinity, selectivity, stability, making them valuable tools for therapeutic purposes. This review explores the flexibility and designability of aptamers as antibiofilm drugs but, importantly, as targeting tools for diverse drug and delivery systems. It highlights specific examples of application of aptamers in biofilms of diverse species according to different modes of action including inhibition of motility and adhesion, blocking of quorum sensing molecules, and dispersal of biofilm-cells to planktonic state. Moreover, it discusses the limitations and challenges that impaired an increased success of the use of aptamers on biofilm management, as well as the opportunities related to aptamers modifications that can significantly expand their applicability on the biofilm field.
Collapse
Affiliation(s)
- Ana Margarida Sousa
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal.
| | - Débora Ferreira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Lígia Raquel Rodrigues
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
26
|
Van Loon JC, Whitfield GB, Wong N, O'Neal L, Henrickson A, Demeler B, O'Toole GA, Parsek MR, Howell PL. Binding of GTP to BifA is required for the production of Pel-dependent biofilms in Pseudomonas aeruginosa. J Bacteriol 2024; 206:e0033123. [PMID: 38197635 PMCID: PMC10882990 DOI: 10.1128/jb.00331-23] [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: 10/06/2023] [Accepted: 12/13/2023] [Indexed: 01/11/2024] Open
Abstract
The Pel exopolysaccharide is one of the most mechanistically conserved and phylogenetically diverse bacterial biofilm matrix determinants. Pel is a major contributor to the structural integrity of Pseudomonas aeruginosa biofilms, and its biosynthesis is regulated by the binding of cyclic-3',5'-dimeric guanosine monophosphate (c-di-GMP) to the PelD receptor. c-di-GMP is synthesized from two molecules of guanosine triphosphate (GTP) by diguanylate cyclases with GGDEF domains and degraded by phosphodiesterases with EAL or HD-GYP domains. As the P. aeruginosa genome encodes 43 c-di-GMP metabolic enzymes, one way signaling specificity can be achieved is through direct interaction between specific enzyme-receptor pairs. Here, we show that the inner membrane hybrid GGDEF-EAL enzyme, BifA, directly interacts with PelD via its cytoplasmic HAMP, GGDEF, and EAL domains. Despite having no catalytic function, the degenerate active site motif of the BifA GGDEF domain (GGDQF) has retained the ability to bind GTP with micromolar affinity. Mutations that abolish GTP binding result in increased biofilm formation but stable global c-di-GMP levels. Our data suggest that BifA forms a dimer in solution and that GTP binding induces conformational changes in dimeric BifA that enhance the BifA-PelD interaction and stimulate its phosphodiesterase activity, thus reducing c-di-GMP levels and downregulating Pel biosynthesis. Structural comparisons between the dimeric AlphaFold2 model of BifA and the structures of other hybrid GGDEF-EAL proteins suggest that the regulation of BifA by GTP may occur through a novel mechanism.IMPORTANCEc-di-GMP is the most common cyclic dinucleotide used by bacteria to regulate phenotypes such as motility, biofilm formation, virulence factor production, cell cycle progression, and cell differentiation. While the identification and initial characterization of c-di-GMP metabolic enzymes are well established, our understanding of how these enzymes are regulated to provide signaling specificity remains understudied. Here we demonstrate that the inactive GGDEF domain of BifA binds GTP and regulates the adjacent phosphodiesterase EAL domain, ultimately downregulating Pel-dependent P. aeruginosa biofilm formation through an interaction with PelD. This discovery adds to the growing body of literature regarding how hybrid GGDEF-EAL enzymes are regulated and provides additional precedence for studying how direct interactions between c-di-GMP metabolic enzymes and effectors result in signaling specificity.
Collapse
Affiliation(s)
- Jaime C. Van Loon
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Gregory B. Whitfield
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Nicole Wong
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Lindsey O'Neal
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Amy Henrickson
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Borries Demeler
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Alberta, Canada
| | - G. A. O'Toole
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Matthew R. Parsek
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - P. Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Wnorowska U, Łysik D, Piktel E, Zakrzewska M, Okła S, Lesiak A, Spałek J, Mystkowska J, Savage PB, Janmey P, Fiedoruk K, Bucki R. Ceragenin-mediated disruption of Pseudomonas aeruginosa biofilms. PLoS One 2024; 19:e0298112. [PMID: 38346040 PMCID: PMC10861078 DOI: 10.1371/journal.pone.0298112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/16/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Microbial biofilms, as a hallmark of cystic fibrosis (CF) lung disease and other chronic infections, remain a desirable target for antimicrobial therapy. These biopolymer-based viscoelastic structures protect pathogenic organisms from immune responses and antibiotics. Consequently, treatments directed at disrupting biofilms represent a promising strategy for combating biofilm-associated infections. In CF patients, the viscoelasticity of biofilms is determined mainly by their polymicrobial nature and species-specific traits, such as Pseudomonas aeruginosa filamentous (Pf) bacteriophages. Therefore, we examined the impact of microbicidal ceragenins (CSAs) supported by mucolytic agents-DNase I and poly-aspartic acid (pASP), on the viability and viscoelasticity of mono- and bispecies biofilms formed by Pf-positive and Pf-negative P. aeruginosa strains co-cultured with Staphylococcus aureus or Candida albicans. METHODS The in vitro antimicrobial activity of ceragenins against P. aeruginosa in mono- and dual-species cultures was assessed by determining minimum inhibitory concentration (MIC) and minimum bactericidal/fungicidal concentration (MBC/MFC). Inhibition of P. aeruginosa mono- and dual-species biofilms formation by ceragenins alone and in combination with DNase I or poly-aspartic acid (pASP) was estimated by the crystal violet assay. Additionally, the viability of the biofilms was measured by colony-forming unit (CFU) counting. Finally, the biofilms' viscoelastic properties characterized by shear storage (G') and loss moduli (G"), were analyzed with a rotational rheometer. RESULTS Our results demonstrated that ceragenin CSA-13 inhibits biofilm formation and increases its fluidity regardless of the Pf-profile and species composition; however, the Pf-positive biofilms are characterized by elevated viscosity and elasticity parameters. CONCLUSION Due to its microbicidal and viscoelasticity-modifying properties, CSA-13 displays therapeutic potential in biofilm-associated infections, especially when combined with mucolytic agents.
Collapse
Affiliation(s)
- Urszula Wnorowska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Dawid Łysik
- Institute of Biomedical Engineering, Bialystok University of Technology, Bialystok, Poland
| | - Ewelina Piktel
- Independent Laboratory of Nanomedicine, Medical University of Białystok, Białystok, Poland
| | - Magdalena Zakrzewska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Sławomir Okła
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, Poland
| | - Agata Lesiak
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, Poland
| | - Jakub Spałek
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, Poland
| | - Joanna Mystkowska
- Institute of Biomedical Engineering, Bialystok University of Technology, Bialystok, Poland
| | - Paul B. Savage
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Paul Janmey
- Department of Physiology and Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Krzysztof Fiedoruk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
28
|
Ma Z, Xiao H, Li H, Lu X, Yan J, Nie H, Yin Q. Prodigiosin as an Antibiofilm Agent against the Bacterial Biofilm-Associated Infection of Pseudomonas aeruginosa. Pathogens 2024; 13:145. [PMID: 38392883 PMCID: PMC10891946 DOI: 10.3390/pathogens13020145] [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: 12/06/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Pseudomonas aeruginosa is known to generate bacterial biofilms that increase antibiotic resistance. With the increase of multi-drug resistance in recent years, the formulation of a new therapeutic strategy has seemed urgent. Preliminary findings show that Prodigiosin (PG), derived from chromium-resistant Serratia marcescens, exhibited efficient anti-biofilm activity against Staphylococcus aureus. However, its anti-biofilm activity against P. aeruginosa remains largely unexplored. The anti-biofilm activity of PG against three clinical single drug-resistant P. aeruginosa was evaluated using crystal violet staining, and the viability of biofilms and planktonic cells were also assessed. A model of chronic lung infection was constructed to test the in vivo antibiofilm activity of PG. The results showed that PG inhibited biofilm formation and effectively inhibited the production of pyocyanin and extracellular polysaccharides in vitro, as well as moderated the expression of interleukins (IL-1β, IL-6, IL-10) and tumor necrosis factor (TNF-α) in vivo, which might be attributed to the downregulation of biofilm-related genes such as algA, pelA, and pslM. These findings suggest that PG could be a potential treatment for drug-resistant P aeruginosa and chronic biofilm infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qi Yin
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, No. 61 Daxuecheng Middle Road, Shapingba District, Chongqing 401334, China
| |
Collapse
|
29
|
Ambreetha S, Zincke D, Balachandar D, Mathee K. Genomic and metabolic versatility of Pseudomonas aeruginosa contributes to its inter-kingdom transmission and survival. J Med Microbiol 2024; 73. [PMID: 38362900 DOI: 10.1099/jmm.0.001791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024] Open
Abstract
Pseudomonas aeruginosa is one of the most versatile bacteria with renowned pathogenicity and extensive drug resistance. The diverse habitats of this bacterium include fresh, saline and drainage waters, soil, moist surfaces, taps, showerheads, pipelines, medical implants, nematodes, insects, plants, animals, birds and humans. The arsenal of virulence factors produced by P. aeruginosa includes pyocyanin, rhamnolipids, siderophores, lytic enzymes, toxins and polysaccharides. All these virulent elements coupled with intrinsic, adaptive and acquired antibiotic resistance facilitate persistent colonization and lethal infections in different hosts. To date, treating pulmonary diseases remains complicated due to the chronic secondary infections triggered by hospital-acquired P. aeruginosa. On the contrary, this bacterium can improve plant growth by suppressing phytopathogens and insects. Notably, P. aeruginosa is one of the very few bacteria capable of trans-kingdom transmission and infection. Transfer of P. aeruginosa strains from plant materials to hospital wards, animals to humans, and humans to their pets occurs relatively often. Recently, we have identified that plant-associated P. aeruginosa strains could be pathologically similar to clinical isolates. In this review, we have highlighted the genomic and metabolic factors that facilitate the dominance of P. aeruginosa across different biological kingdoms and the varying roles of this bacterium in plant and human health.
Collapse
Affiliation(s)
- Sakthivel Ambreetha
- Developmental Biology and Genetics, Division of Biological Sciences, Indian Institute of Science, Bengaluru, Karnataka, 560012, India
| | - Diansy Zincke
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32610, USA
| | - Dananjeyan Balachandar
- Department of Agricultural Microbiology, Tamil Nadu Agricultural University, Coimbatore, 641003, Tamil Nadu, India
| | - Kalai Mathee
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
30
|
Ruijgrok G, Wu DY, Overkleeft HS, Codée JDC. Synthesis and application of bacterial exopolysaccharides. Curr Opin Chem Biol 2024; 78:102418. [PMID: 38134611 DOI: 10.1016/j.cbpa.2023.102418] [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: 11/10/2023] [Revised: 12/02/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023]
Abstract
Exopolysaccharides are produced and excreted by bacteria in the generation of biofilms to provide a protective environment. These polysaccharides are generally generated as heterogeneous polymers of varying length, featuring diverse substitution patterns. To obtain well-defined fragments of these polysaccharides, organic synthesis often is the method of choice, as it allows for full control over chain length and the installation of a pre-determined substitution pattern. This review presents several recent syntheses of exopolysaccharide fragments of Pseudomonas aeruginosa and Staphylococcus aureus and illustrates how these have been used to study biosynthesis enzymes and generate synthetic glycoconjugate vaccines.
Collapse
Affiliation(s)
- Gijs Ruijgrok
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333, CC Leiden, the Netherlands
| | - Dung-Yeh Wu
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333, CC Leiden, the Netherlands
| | - Herman S Overkleeft
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333, CC Leiden, the Netherlands
| | - Jeroen D C Codée
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333, CC Leiden, the Netherlands.
| |
Collapse
|
31
|
Hemmati J, Nazari M, Ahmadi A, Bayati M, Jalili M, Taheri M, Mohammadi Y, Asghari B. In vitro evaluation of biofilm phenotypic and genotypic characteristics among clinical isolates of Pseudomonas aeruginosa in Hamadan, West of Iran. J Appl Genet 2024; 65:213-222. [PMID: 38017355 DOI: 10.1007/s13353-023-00811-6] [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: 05/09/2023] [Revised: 11/03/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023]
Abstract
Due to high antimicrobial resistance and biofilm-forming ability, Pseudomonas aeruginosa is one of the seriously life-threatening agents causing chronic and nosocomial infections. This study was performed to determine the antibiotic resistance pattern, biofilm formation, and frequency of biofilm-related genes in P. aeruginosa strains. In total, 123 P. aeruginosa isolates were collected from different clinical sources. Antimicrobial susceptibility testing (AST) was performed to detect multidrug-resistant P. aeruginosa (MDRPA) isolates. To evaluate the biofilm-forming isolates, the microtiter plate (MTP) method was carried out. Also, the prevalence of biofilm genotype patterns, including pslA, pslD, pelA, pelF, and algD genes, was detected by polymerases chain reaction (PCR). According to our findings, the highest resistance and susceptibility rates were found in ceftazidime with 74.7% (n = 92) and ciprofloxacin with 42.2% (n = 52), respectively. In our study, the highest level of antibiotic resistance belonged to wound isolates which meropenem had the most antibacterial activity against them. In total, 86.1% (n = 106) P. aeruginosa isolates were determined as MDRPA, of which 61.3% (n = 65) were able to form strong biofilm. The highest and lowest frequency of biofilm-related genes among biofilm producer isolates belonged to pelF with 82.1% (n = 101) and algD with 55.2% (n = 68), respectively. The findings of the conducted study indicate a significant relationship between MDRPA and biofilm genotypic/phenotypic patterns, suggesting the necessity of a careful surveillance program in hospital settings.
Collapse
Affiliation(s)
- Jaber Hemmati
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohsen Nazari
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amjad Ahmadi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maral Bayati
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahsa Jalili
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Taheri
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Younes Mohammadi
- Department of Epidemiology, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Babak Asghari
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
32
|
Wang D, Naqvi STA, Lei F, Zhang Z, Yu H, Ma LZ. Glycosyl hydrolase from Pseudomonas fluorescens inhibits the biofilm formation of Pseudomonads. Biofilm 2023; 6:100155. [PMID: 37928620 PMCID: PMC10622837 DOI: 10.1016/j.bioflm.2023.100155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 11/07/2023] Open
Abstract
Biofilms are complex microbial communities embedded in extracellular matrix. Pathogens within the biofilm become more resistant to the antibiotics than planktonic counterparts. Novel strategies are required to encounter biofilms. Exopolysaccharides are one of the major components of biofilm matrix and play a vital role in biofilm architecture. In previous studies, a glycosyl hydrolase, PslGPA, from Pseudomonas aeruginosa was found to be able to inhibit biofilm formation by disintegrating exopolysaccharide in biofilms. Here, we investigate the potential spectrum of PslG homologous protein with anti-biofilm activity. One glycosyl hydrolase from Pseudomonas fluorescens, PslGPF, exhibits anti-biofilm activities and the key catalytic residues of PslGPF are conserved with those of PslGPA. PslGPF at concentrations as low as 50 nM efficiently inhibits the biofilm formation of P. aeruginosa and disassemble its preformed biofilm. Furthermore, PslGPF exhibits anti-biofilm activity on a series of Pseudomonads, including P. fluorescens, Pseudomonas stutzeri and Pseudomonas syringae pv. phaseolicola. PslGPF stays active under various temperatures. Our findings suggest that P. fluorescens glycosyl hydrolase PslGPF has potential to be a broad spectrum inhibitor on biofilm formation of a wide range of Pseudomonads.
Collapse
Affiliation(s)
- Di Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Syed Tatheer Alam Naqvi
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Fanglin Lei
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Yunnan University, Kunming, 650500, PR China
| | - Zhenyu Zhang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Haiying Yu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Luyan Z. Ma
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
33
|
Guo Y, Mao Z, Ran F, Sun J, Zhang J, Chai G, Wang J. Nanotechnology-Based Drug Delivery Systems to Control Bacterial-Biofilm-Associated Lung Infections. Pharmaceutics 2023; 15:2582. [PMID: 38004561 PMCID: PMC10674810 DOI: 10.3390/pharmaceutics15112582] [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: 08/31/2023] [Revised: 10/09/2023] [Accepted: 10/17/2023] [Indexed: 11/26/2023] Open
Abstract
Airway mucus dysfunction and impaired immunological defenses are hallmarks of several lung diseases, including asthma, cystic fibrosis, and chronic obstructive pulmonary diseases, and are mostly causative factors in bacterial-biofilm-associated respiratory tract infections. Bacteria residing within the biofilm architecture pose a complex challenge in clinical settings due to their increased tolerance to currently available antibiotics and host immune responses, resulting in chronic infections with high recalcitrance and high rates of morbidity and mortality. To address these unmet clinical needs, potential anti-biofilm therapeutic strategies are being developed to effectively control bacterial biofilm. This review focuses on recent advances in the development and application of nanoparticulate drug delivery systems for the treatment of biofilm-associated respiratory tract infections, especially addressing the respiratory barriers of concern for biofilm accessibility and the various types of nanoparticles used to combat biofilms. Understanding the obstacles facing pulmonary drug delivery to bacterial biofilms and nanoparticle-based approaches to combatting biofilm may encourage researchers to explore promising treatment modalities for bacterial-biofilm-associated chronic lung infections.
Collapse
Affiliation(s)
- Yutong Guo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zeyuan Mao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Fang Ran
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Jingfeng Zhang
- The Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo 315000, China
| | - Guihong Chai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510180, China
| |
Collapse
|
34
|
Gheorghita AA, Wozniak DJ, Parsek MR, Howell PL. Pseudomonas aeruginosa biofilm exopolysaccharides: assembly, function, and degradation. FEMS Microbiol Rev 2023; 47:fuad060. [PMID: 37884397 PMCID: PMC10644985 DOI: 10.1093/femsre/fuad060] [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: 04/21/2023] [Revised: 10/04/2023] [Accepted: 10/25/2023] [Indexed: 10/28/2023] Open
Abstract
The biofilm matrix is a fortress; sheltering bacteria in a protective and nourishing barrier that allows for growth and adaptation to various surroundings. A variety of different components are found within the matrix including water, lipids, proteins, extracellular DNA, RNA, membrane vesicles, phages, and exopolysaccharides. As part of its biofilm matrix, Pseudomonas aeruginosa is genetically capable of producing three chemically distinct exopolysaccharides - alginate, Pel, and Psl - each of which has a distinct role in biofilm formation and immune evasion during infection. The polymers are produced by highly conserved mechanisms of secretion, involving many proteins that span both the inner and outer bacterial membranes. Experimentally determined structures, predictive modelling of proteins whose structures are yet to be solved, and structural homology comparisons give us insight into the molecular mechanisms of these secretion systems, from polymer synthesis to modification and export. Here, we review recent advances that enhance our understanding of P. aeruginosa multiprotein exopolysaccharide biosynthetic complexes, and how the glycoside hydrolases/lyases within these systems have been commandeered for antimicrobial applications.
Collapse
Affiliation(s)
- Andreea A Gheorghita
- Program in Molecular Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Medical Science Building, 1 King's College Cir, Toronto, ON M5S 1A8, Canada
| | - Daniel J Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, 776 Biomedical Research Tower, 460 W 12th Ave, Columbus, OH 43210, United States
- Department of Microbiology, The Ohio State University College, Biological Sciences Bldg, 105, 484 W 12th Ave, Columbus, OH 43210, United States
| | - Matthew R Parsek
- Department of Microbiology, University of Washington, Health Sciences Bldg, 1705 NE Pacific St, Seattle, WA 98195-7735, United States
| | - P Lynne Howell
- Program in Molecular Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay St, Toronto, ON M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Medical Science Building, 1 King's College Cir, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
35
|
Wiesmann CL, Wang NR, Zhang Y, Liu Z, Haney CH. Origins of symbiosis: shared mechanisms underlying microbial pathogenesis, commensalism and mutualism of plants and animals. FEMS Microbiol Rev 2023; 47:fuac048. [PMID: 36521845 PMCID: PMC10719066 DOI: 10.1093/femsre/fuac048] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/17/2023] Open
Abstract
Regardless of the outcome of symbiosis, whether it is pathogenic, mutualistic or commensal, bacteria must first colonize their hosts. Intriguingly, closely related bacteria that colonize diverse hosts with diverse outcomes of symbiosis have conserved host-association and virulence factors. This review describes commonalities in the process of becoming host associated amongst bacteria with diverse lifestyles. Whether a pathogen, commensal or mutualist, bacteria must sense the presence of and migrate towards a host, compete for space and nutrients with other microbes, evade the host immune system, and change their physiology to enable long-term host association. We primarily focus on well-studied taxa, such as Pseudomonas, that associate with diverse model plant and animal hosts, with far-ranging symbiotic outcomes. Given the importance of opportunistic pathogens and chronic infections in both human health and agriculture, understanding the mechanisms that facilitate symbiotic relationships between bacteria and their hosts will help inform the development of disease treatments for both humans, and the plants we eat.
Collapse
Affiliation(s)
- Christina L Wiesmann
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Nicole R Wang
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Yue Zhang
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Zhexian Liu
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Cara H Haney
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
36
|
Esfahani MB, Khodavandi A, Alizadeh F, Bahador N. Antibacterial and Anti-Biofilm Activities of Microbial Synthesized Silver and Magnetic Iron Oxide Nanoparticles Against Pseudomonas aeruginosa. IEEE Trans Nanobioscience 2023; 22:956-966. [PMID: 37071524 DOI: 10.1109/tnb.2023.3268138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
Pseudomonas aeruginosa is a human bacterial pathogen causing devastating diseases and equipped with various virulence factors like biofilm formation. Common antibiotic treatment has limited efficacy for the P. aeruginosa present in biofilms because of the increased resistance. In this study, we focused our attention on the antibacterial and anti-biofilm activities of various microbial synthesized silver (nano-Ag) and magnetic iron oxide (nano-Fe3O4) nanoparticles against clinical isolates of P. aeruginosa that displayed ceftazidime resistance. The nano-Ag and nano-Fe3O4 represented great antibacterial properties. Nano-Ag and nano-Fe3O4 exhibited a reduction in the biofilm formation by P. aeruginosa reference strain as determined by crystal violet and XTT assays and light microscopy method. Among all, nano-Ag-2 and 7 owing to inherent attributes and mechanisms of resistance in the bacterial biofilm, exhibited anti-biofilm efficacy against ceftazidime resistance clinical isolate of P. aeruginosa. Moreover, nano-Ag and nano-Fe3O4 changed the relative expression of biofilm-associated genes, PELA and PSLA in a concentration dependent manner by P. aeruginosa reference strain. As revealed by qRT-PCR, the expression levels of biofilm-associated genes were downregulated in P. aeruginosa biofilms treated with nano-Ag, while selected biofilm-associated genes were low expressed under treated with nano-Fe3O4. Results of the study demonstrate the potential of microbial synthesized nano-Ag-2 and 7 to act as anti-biofilm agents against ceftazidime resistance clinical isolate of P. aeruginosa. Molecular targeting of biofilm-associated genes by nano-Ag and nano-Fe3O4 may be candidate for new therapeutics against P. aeruginosa diseases.
Collapse
|
37
|
Rodríguez-Suárez JM, Gershenson A, Onuh TU, Butler CS. The Heterogeneous Diffusion of Polystyrene Nanoparticles and the Effect on the Expression of Quorum-Sensing Genes and EPS Production as a Function of Particle Charge and Biofilm Age. ENVIRONMENTAL SCIENCE. NANO 2023; 10:2551-2565. [PMID: 37868332 PMCID: PMC10585598 DOI: 10.1039/d3en00219e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Biofilms are abundantly present in both natural and engineered environmental systems and will likely influence broader particle fate and transport phenomena. While some developed models describe the interactions between nanoparticles and biofilms, studies are only beginning to uncover the complexity of nanoparticle diffusion patterns. With the knowledge of the nanoparticle potential to influence bacterial processes, more systematic studies are needed to uncover the dynamics of bacteria-nanoparticle interactions. This study explored specific microbial responses to nanoparticles and the heterogeneity of nanoparticle diffusion. Pseudomonas aeruginosa biofilms (cultivated for 48 and 96 hours, representing early and late stages of development) were exposed to charged (aminated and carboxylated) polystyrene nanoparticles. With a combination of advanced fluorescence microscopy and real time quantitative PCR, we characterized the diffusion of polystyrene nanoparticles in P. aeruginosa biofilms and evaluated how biofilms respond to the presence of nanoparticles in terms of the expression of key EPS production-associated genes (pelA and rpsL) and quorum-sensing associated (lasR) genes. Our findings show that nanoparticle diffusion coefficients are independent of the particle surface charge only in mature biofilms and that the presence of nanoparticles influences bacterial gene expression. Independent of the particle's charge polystyrene nanoparticles down-regulated pelA in mature biofilms. By contrast, charge-specific responses were identified in lasR and rpsL gene expression. The targeted genes expression analysis and heterogeneous diffusion models demonstrate that particle charge influences nanoparticle mobility and provides significant insight into the intrinsic structural heterogeneity of P. aeruginosa biofilms. These findings suggest that biofilm maturity and particle charge are essential factors to consider when evaluating the transport of nanoparticles within a biofilm matrix.
Collapse
Affiliation(s)
- Joann M. Rodríguez-Suárez
- Department of Civil and Environmental Engineering, University of Massachusetts Amherst, Amherst MA 01003
| | - Anne Gershenson
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst MA 01003
| | - Timothy Umma Onuh
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst MA 01003
| | - Caitlyn S. Butler
- Department of Civil and Environmental Engineering, University of Massachusetts Amherst, Amherst MA 01003
| |
Collapse
|
38
|
Wang C, Li C, Li X, Cai L, Han Z, Du R. RETRACTED ARTICLE: Burn Wounds: Proliferating Site for Biofilm Infection. Appl Biochem Biotechnol 2023; 195:5478. [PMID: 35604533 DOI: 10.1007/s12010-022-03964-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Chao Wang
- Second Department of Burn and Plastic Surgery, Tangshan Worker's Hospital, 063000, Tangshan, Hebei, China
| | - Chungeng Li
- Department of proctology, Tangshan Hospital of Traditional Chinese Medicine, 063000, Tangshan, Hebei, China
| | - Xiaoying Li
- Department of Internal Emergency, Tangshan Worker's Hospital, 063000, Tangshan, Hebei, China
| | - Lanfang Cai
- Second Department of Burn and Plastic Surgery, Tangshan Worker's Hospital, 063000, Tangshan, Hebei, China
| | - Zhenning Han
- Second Department of Burn and Plastic Surgery, Tangshan Worker's Hospital, 063000, Tangshan, Hebei, China
| | - Rui Du
- Department one of Cardiology, Tangshan Worker's Hospital, 063000, Tangshan, Hebei, China.
| |
Collapse
|
39
|
Stewart PS, Owkes M. Simulation of catalase-dependent tolerance of microbial biofilm to hydrogen peroxide with a biofilm computer model. NPJ Biofilms Microbiomes 2023; 9:60. [PMID: 37612330 PMCID: PMC10447567 DOI: 10.1038/s41522-023-00426-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023] Open
Abstract
Hydrogen peroxide (HP) is a common disinfectant and antiseptic. When applied to a biofilm, it may be expected that the top layer of the biofilm would be killed by HP, the HP would penetrate further, and eventually eradicate the entire biofilm. However, using the Biofilm.jl computer model, we demonstrate a mechanism by which the biofilm can persist, and even become thicker, in the indefinite treatment with an HP solution at concentrations that are lethal to planktonic microorganisms. This surprising result is found to be dependent on the neutralization of HP by dead biomass, which provides protection for living biomass deeper within the biofilm. Practically, to control a biofilm, this result leads to the concept of treating with an HP dose exceeding a critical threshold concentration rather than a sustained, lower-concentration treatment.
Collapse
Affiliation(s)
- Philip S Stewart
- Chemical & Biological Engineering, Montana State University, Bozeman, 59717, MT, USA.
| | - Mark Owkes
- Mechanical & Industrial Engineering, Montana State University, Bozeman, 59717, MT, USA.
| |
Collapse
|
40
|
Varponi I, Ferro S, Menilli L, Grapputo A, Moret F, Mastrotto F, Marin O, Sandrelli F. Fighting Pseudomonas aeruginosa Infections: Antibacterial and Antibiofilm Activity of D-Q53 CecB, a Synthetic Analog of a Silkworm Natural Cecropin B Variant. Int J Mol Sci 2023; 24:12496. [PMID: 37569868 PMCID: PMC10419416 DOI: 10.3390/ijms241512496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic Gram-negative bacterium responsible for severe nosocomial infections and is considered a critical pulmonary pathogen for both immunocompromised and cystic fibrosis patients. Planktonic cells of P. aeruginosa possess intrinsic and acquired resistances, inactivating several classes of conventional antibiotics. Additionally, this bacterium can grow, forming biofilms, and complex structures, further hampering the action of multiple antibiotics. Here, we report the biological properties of D-Q53 CecB, an all-D enantiomer of the silkworm natural peptide Q53 CecB. Compared to the L-variant, D-Q53 CecB was resistant to in vitro degradation by humans and P. aeruginosa elastases and showed an enhanced bactericidal activity against P. aeruginosa planktonic bacteria. D-Q53 CecB was thermostable and maintained its antimicrobial activity at high salt concentrations and in the presence of divalent cations or fetal-bovine serum, although at reduced levels. Against different types of human cells, D-Q53 CecB showed cytotoxic phenomena at concentrations several folds higher compared to those active against P. aeruginosa. When L- and D-Q53 CecB were compared for their antibiofilm properties, both peptides were active in inhibiting biofilm formation. However, the D-enantiomer was extremely effective in inducing biofilm degradation, suggesting this peptide as a favorable candidate in an anti-Pseudomonas therapy.
Collapse
Affiliation(s)
- Irene Varponi
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35131 Padova, Italy; (I.V.); (L.M.); (A.G.); (F.M.)
| | - Stefania Ferro
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, 35131 Padova, Italy; (S.F.); (O.M.)
| | - Luca Menilli
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35131 Padova, Italy; (I.V.); (L.M.); (A.G.); (F.M.)
| | - Alessandro Grapputo
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35131 Padova, Italy; (I.V.); (L.M.); (A.G.); (F.M.)
- National Biodiversity Future Centre, Piazza Marina 61, 90133 Palermo, Italy
| | - Francesca Moret
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35131 Padova, Italy; (I.V.); (L.M.); (A.G.); (F.M.)
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy;
| | - Oriano Marin
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, 35131 Padova, Italy; (S.F.); (O.M.)
| | - Federica Sandrelli
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35131 Padova, Italy; (I.V.); (L.M.); (A.G.); (F.M.)
| |
Collapse
|
41
|
Melaugh G, Martinez VA, Baker P, Hill PJ, Howell PL, Wozniak DJ, Allen RJ. Distinct types of multicellular aggregates in Pseudomonas aeruginosa liquid cultures. NPJ Biofilms Microbiomes 2023; 9:52. [PMID: 37507436 PMCID: PMC10382557 DOI: 10.1038/s41522-023-00412-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Pseudomonas aeruginosa forms suspended multicellular aggregates when cultured in liquid media. These aggregates may be important in disease, and/or as a pathway to biofilm formation. The polysaccharide Psl and extracellular DNA (eDNA) have both been implicated in aggregation, but previous results depend strongly on the experimental conditions. Here we develop a quantitative microscopy-based method for assessing changes in the size distribution of suspended aggregates over time in growing cultures. For exponentially growing cultures of P. aeruginosa PAO1, we find that aggregation is mediated by cell-associated Psl, rather than by either eDNA or secreted Psl. These aggregates arise de novo within the culture via a growth process that involves both collisions and clonal growth, and Psl non-producing cells do not aggregate with producers. In contrast, we find that stationary phase (overnight) cultures contain a different type of multicellular aggregate, in which both eDNA and Psl mediate cohesion. Our findings suggest that the physical and biological properties of multicellular aggregates may be very different in early-stage vs late-stage bacterial cultures.
Collapse
Affiliation(s)
- Gavin Melaugh
- SUPA, School of Physics and Astronomy, University of Edinburgh, Edinburgh, EH9 3FD, UK.
- School of Engineering, University of Edinburgh, Edinburgh, EH9 3JL, UK.
| | - Vincent A Martinez
- SUPA, School of Physics and Astronomy, University of Edinburgh, Edinburgh, EH9 3FD, UK
| | - Perrin Baker
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, M5G 0A4, ON, Canada
| | - Preston J Hill
- Departments of Microbial Infection and Immunity, Microbiology, Infectious Diseases Institute, Ohio State University, Columbus, OH, 43210, USA
| | - P Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, M5G 0A4, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, ON, Canada
| | - Daniel J Wozniak
- Departments of Microbial Infection and Immunity, Microbiology, Infectious Diseases Institute, Ohio State University, Columbus, OH, 43210, USA
| | - Rosalind J Allen
- SUPA, School of Physics and Astronomy, University of Edinburgh, Edinburgh, EH9 3FD, UK
- Theoretical Microbial Ecology, Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, Jena, 07745, Germany
| |
Collapse
|
42
|
Batoni G, Catelli E, Kaya E, Pompilio A, Bianchi M, Ghelardi E, Di Bonaventura G, Esin S, Maisetta G. Antibacterial and Antibiofilm Effects of Lactobacilli Strains against Clinical Isolates of Pseudomonas aeruginosa under Conditions Relevant to Cystic Fibrosis. Antibiotics (Basel) 2023; 12:1158. [PMID: 37508254 PMCID: PMC10376640 DOI: 10.3390/antibiotics12071158] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Therapy of lung infections sustained by Pseudomonas aeruginosa in cystic fibrosis (CF) patients is challenging due to the presence of a sticky mucus in the airways and the ability of the bacterium to form biofilm, which exhibits increased antibiotic tolerance. A lung-directed bacteriotherapy through the airway administration of probiotics could represent an alternative approach to probiotic diet supplementation to improve the benefits and clinical outcomes of this kind of intervention in CF patients. This study aims to evaluate the ability of probiotic strains to grow in artificial sputum medium (ASM), mimicking the CF lung microenvironment, and to affect the planktonic and biofilm growth of CF clinical strains of P. aeruginosa in the same conditions. The results demonstrate that Lacticaseibacillus rhamnosus and Lactiplantibacillus plantarum (LP) can grow in ASM. LP inhibited the planktonic growth of P. aeruginosa, while both lactobacilli reduced the pre-formed biofilm of P. aeruginosa. Interestingly, LP was demonstrated to reduce the amount of polysaccharides in the extracellular matrix of P. aeruginosa biofilms and to potentiate the antibiofilm effects of tobramycin. Overall, the results indicated that LP is a promising candidate as an adjuvant in the antimicrobial therapy of P. aeruginosa infections in CF patients.
Collapse
Affiliation(s)
- Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Elisa Catelli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Esingül Kaya
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Arianna Pompilio
- Department of Medical, Oral, and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Marta Bianchi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Emilia Ghelardi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Giovanni Di Bonaventura
- Department of Medical, Oral, and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Semih Esin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| | - Giuseppantonio Maisetta
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via S. Zeno 37, 56123 Pisa, Italy
| |
Collapse
|
43
|
Razvi E, DiFrancesco BR, Wasney GA, Morrison ZA, Tam J, Auger A, Baker P, Alnabelseya N, Rich JD, Sivarajah P, Whitfield GB, Harrison JJ, Melnyk RA, Nitz M, Howell PL. Small Molecule Inhibition of an Exopolysaccharide Modification Enzyme is a Viable Strategy To Block Pseudomonas aeruginosa Pel Biofilm Formation. Microbiol Spectr 2023; 11:e0029623. [PMID: 37098898 PMCID: PMC10269871 DOI: 10.1128/spectrum.00296-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/03/2023] [Indexed: 04/27/2023] Open
Abstract
Biosynthesis of the Pel exopolysaccharide in Pseudomonas aeruginosa requires all seven genes of the pelABCDEFG operon. The periplasmic modification enzyme PelA contains a C-terminal deacetylase domain that is necessary for Pel-dependent biofilm formation. Herein, we show that extracellular Pel is not produced by a P. aeruginosa PelA deacetylase mutant. This positions PelA deacetylase activity as an attractive target to prevent Pel-dependent biofilm formation. Using a high-throughput screen (n = 69,360), we identified 56 compounds that potentially inhibit PelA esterase activity, the first enzymatic step in the deacetylase reaction. A secondary biofilm inhibition assay identified methyl 2-(2-pyridinylmethylene) hydrazinecarbodithioate (SK-017154-O) as a specific Pel-dependent biofilm inhibitor. Structure-activity relationship studies identified the thiocarbazate as a necessary functional group and that the pyridyl ring could be replaced with a phenyl substituent (compound 1). Both SK-017154-O and compound 1 inhibit Pel-dependent biofilm formation in Bacillus cereus ATCC 10987, which has a predicted extracellular PelA deacetylase in its pel operon. Michaelis-Menten kinetics determined SK-017154-O to be a noncompetitive inhibitor of PelA, while compound 1 did not directly inhibit PelA esterase activity. Cytotoxicity assays using human lung fibroblast cells showed that compound 1 is less cytotoxic than SK-017154-O. This work provides proof of concept that biofilm exopolysaccharide modification enzymes are important for biofilm formation and can serve as useful antibiofilm targets. IMPORTANCE Present in more than 500 diverse Gram-negative and 900 Gram-positive organisms, the Pel polysaccharide is one of the most phylogenetically widespread biofilm matrix determinants found to date. Partial de-N-acetylation of this α-1,4 linked N-acetylgalactosamine polymer by the carbohydrate modification enzyme PelA is required for Pel-dependent biofilm formation in Pseudomonas aeruginosa and Bacillus cereus. Given this and our observation that extracellular Pel is not produced by a P. aeruginosa PelA deactylase mutant, we developed an enzyme-based high-throughput screen and identified methyl 2-(2-pyridinylmethylene) hydrazinecarbodithioate (SK-017154-O) and its phenyl derivative as specific Pel-dependent biofilm inhibitors. Michaelis-Menten kinetics revealed SK-017154-O is a noncompetitive inhibitor and that its noncytotoxic, phenyl derivative does not directly inhibit P. aeruginosa PelA esterase activity. We provide proof of concept that exopolysaccharide modification enzymes can be targeted with small molecule inhibitors to block Pel-dependent biofilm development in both Gram-negative and Gram-positive bacteria.
Collapse
Affiliation(s)
- Erum Razvi
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | | | - Gregory A. Wasney
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Structural & Biophysical Core Facility, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - John Tam
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Anick Auger
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- SPARC BioCentre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Perrin Baker
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Noor Alnabelseya
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Jacquelyn D. Rich
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Piyanka Sivarajah
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Gregory B. Whitfield
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Joe J. Harrison
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Roman A. Melnyk
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Mark Nitz
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - P. Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
44
|
Liang Z, Nilsson M, Kragh KN, Hedal I, Alcàcer-Almansa J, Kiilerich RO, Andersen JB, Tolker-Nielsen T. The role of individual exopolysaccharides in antibiotic tolerance of Pseudomonas aeruginosa aggregates. Front Microbiol 2023; 14:1187708. [PMID: 37333638 PMCID: PMC10272609 DOI: 10.3389/fmicb.2023.1187708] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
The bacterium Pseudomonas aeruginosa is involved in chronic infections of cystic fibrosis lungs and chronic wounds. In these infections the bacteria are present as aggregates suspended in host secretions. During the course of the infections there is a selection for mutants that overproduce exopolysaccharides, suggesting that the exopolysaccharides play a role in the persistence and antibiotic tolerance of the aggregated bacteria. Here, we investigated the role of individual P. aeruginosa exopolysaccharides in aggregate-associated antibiotic tolerance. We employed an aggregate-based antibiotic tolerance assay on a set of P. aeruginosa strains that were genetically engineered to over-produce a single, none, or all of the three exopolysaccharides Pel, Psl, and alginate. The antibiotic tolerance assays were conducted with the clinically relevant antibiotics tobramycin, ciprofloxacin and meropenem. Our study suggests that alginate plays a role in the tolerance of P. aeruginosa aggregates toward tobramycin and meropenem, but not ciprofloxacin. However, contrary to previous studies we did not observe a role for Psl or Pel in the tolerance of P. aeruginosa aggregates toward tobramycin, ciprofloxacin, and meropenem.
Collapse
Affiliation(s)
- Ziwei Liang
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Martin Nilsson
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Kasper Nørskov Kragh
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Ida Hedal
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Júlia Alcàcer-Almansa
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Bacterial Infections: Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Rikke Overgaard Kiilerich
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bo Andersen
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Tim Tolker-Nielsen
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
45
|
Islam OK, Islam I, Saha O, Rahaman MM, Sultana M, Bockmühl DP, Hossain MA. Genomic variability correlates with biofilm phenotypes in multidrug resistant clinical isolates of Pseudomonas aeruginosa. Sci Rep 2023; 13:7867. [PMID: 37188866 DOI: 10.1038/s41598-023-35056-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 05/11/2023] [Indexed: 05/17/2023] Open
Abstract
The multifactorial nature of Pseudomonas aeruginosa biofilm development and genomic variabilities implicates its resistance to conventional antimicrobials and virulence. Therefore, genetic determinants need to be extensively studied to block the early steps of biofilm or already formed biofilms. In this study, a total of 20 multidrug resistant (MDR) clinical P. aeruginosa isolates were evaluated for their biofilm forming abilities and related genes. Of the isolates tested, all of them showed surface attachment tendencies in nutrient limiting conditions, and classified as strong (SBF = 45%), moderate (MBF = 30%) and weak (WBF = 25%) biofilm formers. Complete genome sequencing of representative strong (DMC-27b), moderate (DMC-20c) and weak biofilm former (DMC-30b) isolates was performed. Analysis of biofilm related genes in the sequenced genomes revealed that, 80 of the 88 biofilm related genes possess 98-100% sequence identity to the reference PAO1 strain. Complete and partial sequence data of LecB proteins from tested isolates indicate that isolates containing PA14-like LecB sequences produced strong biofilms. All of the 7 pel operon protein coding genes in weak biofilm former isolate 30b showed significant nucleotide sequence variation with other tested isolates, and their corresponding proteins are 99% identical with the pel operon proteins of PA7. Bioinformatics analyses identified divergent sequence and structural features that separate PA7 like pel operon proteins from reference PAO1-like pel operon. Congo red and pellicle forming assays revealed that the sequence and structure variations may have interfered with the Pel production pathway and resulted in impaired Pel production in isolate 30b that has a PA7 like pel operon. Expression analysis also showed that both pelB and lecB genes were about 5 to 6 folds upregulated after 24 h in SBF 27b in comparison with WBF 30b. Our findings indicate significant genomic divergence in biofilm related genes of P. aeruginosa strains that affect their biofilm phenotypes.
Collapse
Affiliation(s)
- Ovinu Kibria Islam
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
- Department of Microbiology, Jashore University of Science & Technology, Jashore, Bangladesh
| | - Israt Islam
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
- Department of Microbiology, Noakhali University of Science & Technology, Noakhali, Bangladesh
| | - Otun Saha
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
- Department of Microbiology, Noakhali University of Science & Technology, Noakhali, Bangladesh
| | | | - Munawar Sultana
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
| | - Dirk P Bockmühl
- Faculty of Life Science, Rhine-Waal University of Applied Science, Kleve, Germany
| | - M Anwar Hossain
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh.
- Department of Microbiology, Jashore University of Science & Technology, Jashore, Bangladesh.
| |
Collapse
|
46
|
Chung J, Eisha S, Park S, Morris AJ, Martin I. How Three Self-Secreted Biofilm Exopolysaccharides of Pseudomonas aeruginosa, Psl, Pel, and Alginate, Can Each Be Exploited for Antibiotic Adjuvant Effects in Cystic Fibrosis Lung Infection. Int J Mol Sci 2023; 24:ijms24108709. [PMID: 37240055 DOI: 10.3390/ijms24108709] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
In cystic fibrosis (CF), pulmonary infection with Pseudomonas aeruginosa is a cause of increased morbidity and mortality, especially in patients for whom infection becomes chronic and there is reliance on long-term suppressive therapies. Current antimicrobials, though varied mechanistically and by mode of delivery, are inadequate not only due to their failure to eradicate infection but also because they do not halt the progression of lung function decline over time. One of the reasons for this failure is thought to be the biofilm mode of growth of P. aeruginosa, wherein self-secreted exopolysaccharides (EPSs) provide physical protection against antibiotics and an array of niches with resulting metabolic and phenotypic heterogeneity. The three biofilm-associated EPSs secreted by P. aeruginosa (alginate, Psl, and Pel) are each under investigation and are being exploited in ways that potentiate antibiotics. In this review, we describe the development and structure of P. aeruginosa biofilms before examining each EPS as a potential therapeutic target for combating pulmonary infection with P. aeruginosa in CF, with a particular focus on the current evidence for these emerging therapies and barriers to bringing these therapies into clinic.
Collapse
Affiliation(s)
- Jonathan Chung
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Shafinaz Eisha
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Subin Park
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Amanda J Morris
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Isaac Martin
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
47
|
Simsek AN, Koch MD, Sanfilippo JE, Gitai Z, Gompper G, Sabass B. Type-IV pili tune an adhesion-migration trade-off during surface colonization of Pseudomonas aeruginosa. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.09.538458. [PMID: 37215001 PMCID: PMC10197611 DOI: 10.1101/2023.05.09.538458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Bacterial pathogenicity relies on both firm surface adhesion and cell dissemination. How twitching bacteria resolve the fundamental contradiction between adhesion and migration is unknown. To address this question, we employ live-cell imaging of type-IV pili (T4P) and therewith construct a comprehensive mathematical model of Pseudomonas aeruginosa migration. The data show that only 10% to 50% of T4P bind to substrates and contribute to migration through random extension and retraction. Individual T4P do not display a measurable sensory response to surfaces, but their number increases on cellular surface contact. Attachment to surfaces is mediated, besides T4P, by passive adhesive forces acting on the cell body. Passive adhesions slow down cell migration and result in local random motion on short time scales, which is followed by directionally persistent, superdiffusive motion on longer time scales. Moreover, passive adhesions strongly enhance surface attachment under shear flow. Δ pilA mutants, which produce no T4P, robustly stick to surfaces under shear flow. In contrast, rapidly migrating Δ pilH cells, which produce an excessive number of T4P, are easily detached by shear. Wild-type cells sacrifice migration speed for robust surface attachment by maintaining a low number of active pili. The different cell strains pertain to disjunct regimes in a generic adhesion-migration trait space. Depending on the nature of the adhesion structures, adhesion and migration are either compatible or a trade-off is required for efficient bacterial surface colonization under different conditions.
Collapse
|
48
|
Zhou S, An W, Zhao K, Lin L, Yang S, Zhang Y, Xu M. Protection of electroactive biofilms against hypersaline shock by quorum sensing. WATER RESEARCH 2023; 233:119823. [PMID: 36871386 DOI: 10.1016/j.watres.2023.119823] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/22/2023] [Accepted: 02/26/2023] [Indexed: 06/18/2023]
Abstract
Quorum sensing (QS) is an ideal strategy for boosting the operating performance of electroactive biofilms (EABs), but its potential effects on the protection of electroactive biofilms against environmental shocks (e.g., hypersaline shock) have been rarely revealed. In this study, a QS signaling molecule, the N-(3-oxo-dodecanoyl)-L-homoserine lactone, was employed to promote the anti-shock property of the EABs against extreme saline shock. The maximum current density of the QS-regulated biofilm recovered to 0.17 mA/cm2 after 10% salinity exposure, which was much higher than those of its counterparts. The laser scanning confocal microscope confirmed a thicker and more compact biofilm with the presence of the QS signaling molecule. The extracellular polymeric substances (EPS) might play a crucial role in the anti-shocking behaviors, as the polysaccharides in EPS of QS-biofilm had doubled compared to the groups with acylase (the QS quencher). The microbial community analysis indicated that the QS molecule enriched the relative abundance of key species including Pseudomonas sp. and Geobacter sp., which were both beneficial to the stability and electroactivity of the biofilms. The functional genes related to the bacterial community were also up-regulated with the presence of the QS molecule. These results highlight the importance of QS effects in protecting electroactive biofilm under extreme environmental shock, which provides effective and feasible strategies for the future development of microbial electrochemical technologies.
Collapse
Affiliation(s)
- Shaofeng Zhou
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China
| | - Wenwen An
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Kexin Zhao
- Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
| | - Lizhou Lin
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Shan Yang
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Yifeng Zhang
- Department of Environmental and Resource Engineering, Technical University of Denmark, Lyngby DK-2800, Denmark
| | - Meiying Xu
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China.
| |
Collapse
|
49
|
Shetty S, Sekar P, Shetty RM, Abou Neel EA. Antibacterial and Antibiofilm Efficacy of Copper-Doped Phosphate Glass on Pathogenic Bacteria. Molecules 2023; 28:molecules28073179. [PMID: 37049941 PMCID: PMC10096066 DOI: 10.3390/molecules28073179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 04/07/2023] Open
Abstract
This study aimed to investigate the antibacterial [minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC)] and antibiofilm activity [log10 colony forming unit/mL (CFU/mL) and biofilm disruption] of copper-doped phosphate glass (CDPG) against Streptococcus oralis, Enterococcus faecalis, Lactobacillus casei, Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa. Methods: the antibacterial activity was determined using microbroth dilution and time-kill assay. The antibiofilm activity was investigated using crystal violet and confocal laser scanning microscopy. Bacteria growing in absence of CDPG were used as controls. Results: the MIC was ≥125 mg of CPDG/mL; the log10 CFU/mL reduction ranged from 2.66–3.14 to 6.23–9.65 after 4 and 24 h respectively. Generally, no growth was observed after 24 h of treatment with CDPG; the MBC was 250 mg/mL for L. casei and S. oralis while 500 mg/mL for the rest of the bacteria. The highest and lowest antibiofilm activity was observed against S. oralis and E. coli respectively. Three patterns of complete biofilm disruption were seen: (i) large areas with E. fecalis and S. oralis, (ii) medium-size pockets with S. aureus and P. aeruginosa, or (iii) small areas with E. coli and L. casei. Conclusion: CDPG can be potentially used as an antibacterial and an antibiofilm agent against oral biofilm-forming bacteria.
Collapse
Affiliation(s)
- Sunaina Shetty
- Preventive and Restorative Dentistry Department, College of Dental Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Priyadharshini Sekar
- RIMHS, Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Raghavendra M. Shetty
- Department of Clinical Sciences, College of Dentistry, Ajman University, Ajman P.O. Box 346, United Arab Emirates
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
- Department of Pediatric and Preventive Dentistry, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research (Deemed-to-be University), Wardha 442001, India
| | - Ensanya Ali Abou Neel
- Preventive and Restorative Dentistry Department, College of Dental Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- UCL Eastman Dental Institute, Biomaterials & Tissue Engineering, Royal Free Hospital, Rowland Hill Street, London NW3 2QG, UK
| |
Collapse
|
50
|
Yahya AH, Harston SR, Colton WL, Cabeen MT. Distinct Screening Approaches Uncover PA14_36820 and RecA as Negative Regulators of Biofilm Phenotypes in Pseudomonas aeruginosa PA14. Microbiol Spectr 2023; 11:e0377422. [PMID: 36971546 PMCID: PMC10100956 DOI: 10.1128/spectrum.03774-22] [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: 12/21/2022] [Accepted: 02/28/2023] [Indexed: 03/29/2023] Open
Abstract
Pseudomonas aeruginosa commonly infects hospitalized patients and the lungs of individuals with cystic fibrosis. This species is known for forming biofilms, which are communities of bacterial cells held together and encapsulated by a self-produced extracellular matrix. The matrix provides extra protection to the constituent cells, making P. aeruginosa infections challenging to treat. We previously identified a gene, PA14_16550, which encodes a DNA-binding TetR-type repressor and whose deletion reduced biofilm formation. Here, we assessed the transcriptional impact of the 16550 deletion and found six differentially regulated genes. Among them, our results implicated PA14_36820 as a negative regulator of biofilm matrix production, while the remaining 5 had modest effects on swarming motility. We also screened a transposon library in a biofilm-impaired ΔamrZ Δ16550 strain for restoration of matrix production. Surprisingly, we found that disruption or deletion of recA increased biofilm matrix production, both in biofilm-impaired and wild-type strains. Because RecA functions both in recombination and in the DNA damage response, we asked which function of RecA is important with respect to biofilm formation by using point mutations in recA and lexA to specifically disable each function. Our results implied that loss of either function of RecA impacts biofilm formation, suggesting that enhanced biofilm formation may be one physiological response of P. aeruginosa cells to loss of either RecA function. IMPORTANCE Pseudomonas aeruginosa is a notorious human pathogen well known for forming biofilms, communities of bacteria that protect themselves within a self-secreted matrix. Here, we sought to find genetic determinants that impacted biofilm matrix production in P. aeruginosa strains. We identified a largely uncharacterized protein (PA14_36820) and, surprisingly, RecA, a widely conserved bacterial DNA recombination and repair protein, as negatively regulating biofilm matrix production. Because RecA has two main functions, we used specific mutations to isolate each function and found that both functions influenced matrix production. Identifying negative regulators of biofilm production may suggest future strategies to reduce the formation of treatment-resistant biofilms.
Collapse
Affiliation(s)
- Amal H. Yahya
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Sophie R. Harston
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - William L. Colton
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Matthew T. Cabeen
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|