1
|
Swain J, Askenasy I, Rudland Nazeer R, Ho PM, Labrini E, Mancini L, Xu Q, Hollendung F, Sheldon I, Dickson C, Welch A, Agbamu A, Godlee C, Welch M. Pathogenicity and virulence of Pseudomonas aeruginosa: Recent advances and under-investigated topics. Virulence 2025; 16:2503430. [PMID: 40353451 PMCID: PMC12087490 DOI: 10.1080/21505594.2025.2503430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/23/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025] Open
Abstract
Pseudomonas aeruginosa is a model for the study of quorum sensing, protein secretion, and biofilm formation. Consequently, it has become one of the most intensely reviewed pathogens, with many excellent articles in the current literature focusing on these aspects of the organism's biology. Here, though, we aim to take a slightly different approach and consider some less well appreciated (but nonetheless important) factors that affect P. aeruginosa virulence. We start by reminding the reader of the global importance of P. aeruginosa infection and that the "virulome" is very niche-specific. Overlooked but obvious questions such as "what prevents secreted protein products from being digested by co-secreted proteases?" are discussed, and we suggest how the nutritional preference(s) of the organism might dictate its environmental reservoirs. Recent studies identifying host genes associated with genetic predisposition towards P. aeruginosa infection (and even infection by specific P. aeruginosa strains) and the role(s) of intracellular P. aeruginosa are introduced. We also discuss the fact that virulence is a high-risk strategy and touch on how expression of the two main classes of virulence factors is regulated. A particular focus is on recent findings highlighting how nutritional status and metabolism are as important as quorum sensing in terms of their impact on virulence, and how co-habiting microbial species at the infection site impact on P. aeruginosa virulence (and vice versa). It is our view that investigation of these issues is likely to dominate many aspects of research into this WHO-designated priority pathogen over the next decade.
Collapse
Affiliation(s)
- Jemima Swain
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Isabel Askenasy
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | - Pok-Man Ho
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Edoardo Labrini
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | - Qingqing Xu
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | | | - Camilla Dickson
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Amelie Welch
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Adam Agbamu
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Camilla Godlee
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Martin Welch
- Department of Biochemistry, Cambridge University, Cambridge, UK
| |
Collapse
|
2
|
Habib MB, Shah NA, Amir A, Tariq MH. Molecular and computational insights into algD biofilm genes in multi drug resistant and extensively drug resistant Pseudomonasaeruginosa. Microb Pathog 2025; 205:107634. [PMID: 40306587 DOI: 10.1016/j.micpath.2025.107634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025]
Abstract
Antibiotic-resistance and biofilm formation are the main virulence factors and present a serious treatment challenge in Pseudomonas aeruginosa. This study aimed to investigate antimicrobial resistance, genetic diversity, biofilm-specific algD gene, and computational analysis of clinical isolates. Forty two isolates of P. aeruginosa were examined by PCR, ELISA, sangers sequencing, phylogenetic analysis, MolProbity score, 3D structural modelling, Ramachandran plot, multiple sequence alignment, and protein domain analysis. According to the results, PCR analysis revealed algD gene presence in all isolates. ELISA showed 55 % (n = 23) of the samples produced strong biofilms, 38 % (n = 16) produced moderate biofilms, and 7 % (3) produced weak biofilms. The evolutionary relationships of 8 (S1-S8) P. aeruginosa strains with 81 reference strains were illustrated by the phylogenetic tree. Samples S1-S8 showed excellent MolProbity score (<1.00), low clashed scores (0.67-0.70), most residues in the favored regions (∼96.2-96.5 %), low Ramachandran outliers (0.53-0.56 %), low Rotamer outlier (0.62 %), low bad angles (<2), indicated high-quality models and values preferred percentages showed excellent models with structural refinement. Over all samples S5 and S6 stood out as the top choices for high-confidence modeling and applications. The essential catalytic domain UDP-glucose/GDP-mannose dehydrogenase was identified that could be used as important therapeutic targets. High quality models indicated suitability for downstream applications, such as studying protein-ligand interactions, understanding structural aspects of biofilm-resistant bacteria. This study improved our knowledge of the mechanisms underlying P. aeruginosa biofilm resistance and sets the stage for the development of novel therapeutic and diagnostic strategies to combat multidrug resistant strains.
Collapse
Affiliation(s)
| | - Naseer Ali Shah
- Department of Biosciences, COMSATS University, Islamabad, 44000, Pakistan.
| | - Afreenish Amir
- Department of Microbiology, National Institute of Health Islamabad, 44000, Pakistan.
| | | |
Collapse
|
3
|
Aziz M, Ahmed K, Ravichandran V. PqsR-specific quorum sensing inhibitors targeting Pseudomonas aeruginosa: current advances and future directions. J Chemother 2025:1-18. [PMID: 40432318 DOI: 10.1080/1120009x.2025.2510093] [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: 01/19/2025] [Revised: 03/23/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025]
Abstract
The emergence of multidrug-resistant (MDR) Pseudomonas aeruginosa is driven by diverse resistance mechanisms, including quorum sensing (QS) and biofilm formation. QS regulates virulence, antibiotic tolerance, efflux pumps, and biofilm development, enhancing the pathogen's adaptability. Among QS systems, the Pseudomonas quinolone signal regulator (PqsR) plays a central role in controlling virulence and biofilm-associated genes. This review critically examines the PqsR-regulated network and highlights the potential of PqsR inhibitors to reduce pathogenicity. Disrupting QS instead of targeting bacterial viability can enhance antibiotic efficacy, making this combinatorial approach a promising strategy to combat MDR P. aeruginosa.
Collapse
Affiliation(s)
- Mandsaurwala Aziz
- Amity Institute of Biotechnology, Amity University Maharashtra, Panvel, Maharashtra, India
- Techinvention Lifecare Private Limted, Mumbai, India
| | - Kafil Ahmed
- Resinno Biotech, Navi Mumbai, Maharashtra, India
| | - Vinothkannan Ravichandran
- Amity Institute of Biotechnology, Amity University Maharashtra, Panvel, Maharashtra, India
- Center for Drug Discovery and Development (CD3), Amity Institute of Biotechnology, Amity University Maharashtra, Panvel, Maharashtra, India
| |
Collapse
|
4
|
Hamdy SA, Hatem S, Elosaily H, Hassan AGAE, Elshimy R, Osman AH, El-Shiekh RA. Therapeutic targeting of ocular diseases with emphasis on PI3K/Akt, and OPRL pathways by Hedera helix L. saponins: a new approach for the treatment of Pseudomonas aeruginosa-induced bacterial keratitis. NATURAL PRODUCTS AND BIOPROSPECTING 2025; 15:30. [PMID: 40353954 PMCID: PMC12069210 DOI: 10.1007/s13659-025-00514-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/21/2025] [Indexed: 05/14/2025]
Abstract
Pseudomonas aeruginosa-induced bacterial keratitis is one of the most sight-threatening corneal infections associated with intense ocular inflammatory reactions that may lead to vision loss. Hence, this study investigated the efficacy of three nanocomposite chitosan-coated penetration enhancer vesicles (PEVs) to augment the ocular delivery of saponin(s), α-hederin (PEVI), hederacoside C (PEVII), or both (PEVIII) for treatment of Pseudomonas keratitis and its induced inflammatory response. The three formulations were prepared using the ethanol injection method and comprehensively characterized. In vitro, the antibacterial activity of the three formulations against P. aeruginosa was evaluated using agar well-diffusion method, pyocyanin production inhibition, and swarming and twitching motility inhibition assays. The therapeutic effect of the three formulations has been investigated in P. aeruginosa keratitis by gross lesion monitoring, determination of bacterial bioburden, biochemical markers, histopathological examination, and scoring after 7 days of topical treatment. Data revealed that PEVI, PEVII, and PEVIII nanocomposites showed particle size in the nanometer range, high entrapment efficiency, good stability, and sustained release of the saponins throughout 24 h. Among them, PEVIII exhibited notably strong in vitro antipseudomonal activity. Additionally, animals treated topically with PEVIII showed an appreciable gross lesion reduction, corneal tissue improvement, and formidable bacterial load reduction compared with untreated and gentamicin sulfate eye (GENTAWISE®) ointment-treated groups. Moreover, PEVIII treatment showed the most significant reduction in TNF-α, NF-κB, ROS levels, and OPRL virulence gene expression while enhancing PI3K/Akt activation. Therefore, this study offers PEVIII as a promising treatment for P. aeruginosa keratitis.
Collapse
Affiliation(s)
- Sherif A Hamdy
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt
| | - Shymaa Hatem
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Future University in Egypt, New Cairo, Egypt.
| | - Heba Elosaily
- Biochemistry Department, Faculty of Pharmacy, Ahram Canadian University, 4th Industrial Region, 6th of October City, 12585, Giza, Egypt
| | | | - Rana Elshimy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
- Department of Microbiology and Immunology, Egyptian Drug Authority, Cairo, Egypt
| | - Ahmed H Osman
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Riham A El-Shiekh
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt.
| |
Collapse
|
5
|
Ramesh R, Rekha ND, Gopal S. Pseudomonas aeruginosa biofilm: treatment strategies to combat infection. Arch Microbiol 2025; 207:141. [PMID: 40348909 DOI: 10.1007/s00203-025-04346-8] [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: 03/26/2025] [Revised: 04/25/2025] [Accepted: 04/26/2025] [Indexed: 05/14/2025]
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogenic bacterium that is a common cause of both acute and chronic infections. Multidrug-resistant P. aeruginosa poses a significant challenge to antibiotics and therapeutic approaches due to its pathogenicity, virulence, and biofilm-forming ability mediated by quorum sensing. Understanding the pathogenic mechanisms is essential for developing potential drug targets. In this regard, strategies aimed at combating the targeted inhibition of virulence, quorum sensing pathways, secretion systems, biofilm-associated two-component systems, and signalling system regulators (such as c-di-GMP) associated with biofilm formation are critical. Several new antimicrobial agents have been developed using these strategies, including antimicrobial peptides, bacteriophages, nanoantibiotics, photodynamics, and natural products, which are considered promising therapeutic tools. In this review, we address the concept of biofilms, their regulation, and recent treatment strategies to target P. aeruginosa, a clinically significant pathogen known for biofilm formation.
Collapse
Affiliation(s)
- Rashmi Ramesh
- Department of Studies in Microbiology, University of Mysore, Manasagangotri, Mysuru, Karnataka, India
| | - N D Rekha
- Department of Biotechnology, JSS College of Arts, Commerce and Science (Autonomous), Mysuru, Karnataka, India
| | - Shubha Gopal
- Department of Studies in Microbiology, University of Mysore, Manasagangotri, Mysuru, Karnataka, India.
| |
Collapse
|
6
|
Howard SA, de Dios R, Maslova E, Myridakis A, Miller TH, McCarthy RR. Pseudomonas aeruginosa clinical isolates can encode plastic-degrading enzymes that allow survival on plastic and augment biofilm formation. Cell Rep 2025:115650. [PMID: 40339583 DOI: 10.1016/j.celrep.2025.115650] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/04/2025] [Accepted: 04/11/2025] [Indexed: 05/10/2025] Open
Abstract
Multiple bacteria encoding plastic-degrading enzymes have been isolated from the environment. Given the widespread use of plastic in healthcare, we hypothesized that bacterial clinical isolates may also degrade plastic. This could render plastic-containing medical devices susceptible to degradation and failure and potentially offer these pathogens a growth-sustaining substrate, enabling them to persist in the hospital-built environment. Here, we mined the genomes of prevalent pathogens and identified several species encoding enzymes with homology to known plastic-degrading enzymes. We identify a clinical isolate of Pseudomonas aeruginosa that encodes an enzyme that enables it to degrade a medically relevant plastic, polycaprolactone (PCL), by 78% in 7 days. Furthermore, this degradation enables the bacterium to utilize PCL as its sole carbon source. We also demonstrate that encoding plastic-degrading enzymes can enhance biofilm formation and pathogenicity. Given the central role of plastic in healthcare, screening nosocomial bacteria for plastic-degrading capacity should be an important future consideration.
Collapse
Affiliation(s)
- Sophie A Howard
- Centre for Antimicrobial Innovations, Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University of London, UB8 3PH Uxbridge, UK
| | - Rubén de Dios
- Centre for Antimicrobial Innovations, Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University of London, UB8 3PH Uxbridge, UK
| | - Evgenia Maslova
- Centre for Antimicrobial Innovations, Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University of London, UB8 3PH Uxbridge, UK
| | - Antonis Myridakis
- Centre for Pollution Research and Policy, Division of Environmental Sciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University of London, UB8 3PH Uxbridge, UK
| | - Thomas H Miller
- Centre for Antimicrobial Innovations, Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University of London, UB8 3PH Uxbridge, UK; Centre for Pollution Research and Policy, Division of Environmental Sciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University of London, UB8 3PH Uxbridge, UK
| | - Ronan R McCarthy
- Centre for Antimicrobial Innovations, Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University of London, UB8 3PH Uxbridge, UK.
| |
Collapse
|
7
|
Benaissa A, Bouali W, Ngenge Tamfu A, Ammara B, Kucukaydin S, Latti N, Khadir A, Bendahou M, Anouar EH, Ceylan O. Inhibition of Clinical Multidrug-Resistant Pseudomonas aeruginosa Biofilms by Cinnamaldehyde and Eugenol From Essential Oils: In Vitro and In Silico Analysis. Chem Biodivers 2025; 22:e202402693. [PMID: 39740034 DOI: 10.1002/cbdv.202402693] [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: 10/21/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/02/2025]
Abstract
Pseudomonas aeruginosa causes nosocomial infections and chronic diseases. Cinnamomum cassia and Syzygium aromaticum are used natural antimicrobials. Essential oil (EO) from C. cassia (CCEO) and S. aromaticum (CEO) was characterized using GC-MS analysis. Eugenol (82.31%), eugenol acetate (10.57%), and β-caryophyllene (3.41%) were major constituents in CEO while cinnamaldehyde (88.18%), cinnamyl acetate (2.85%) and 2-methoxy cinnamaldehyde (1.77%) were main components in CCEO. The EOs and major constituents exhibited good antimicrobial activity against clinical strains of P. aeruginosa. Cinnamaldehyde exhibited the best antimicrobial effect with minimal inhibitory concentration (MIC) as low as 0.031% ± 0.07% (v/v) and inhibition zones reaching 30 ± 0.5 mm diameter. Test samples showed antibiofilm activities against two culture types and seven clinical strains of P. aeruginosa at concentrations of 2MIC to MIC/4. CCEO and its major constituent cinnamaldehyde were more active, compared to CEO and its major constituent eugenol. Scanning electron microscopy images showed untreated colonies with well-developed biofilms while there was significant reduction of biofilms with distorted architecture and cell shrinkage upon treatment with test samples. In silico studies indicated great interactions between the major compounds, eugenol and cinnamaldehyde, with the receptor proteins of P. aeruginosa revealed by negative binding energies. Eugenol and cinnamaldehyde exhibited appreciable druglikeness.
Collapse
Affiliation(s)
- Asma Benaissa
- Laboratory of Applied Microbiology in Food, Biomedical, and Environment (LAMAABE), Department of Biology, Faculty of Sciences of Nature, Life, Earth, and Universe, Abou Bekr Belkaïd University of Tlemcen, Tlemcen, Algeria
| | - Wafaa Bouali
- Laboratory Antifungal, Antibiotic, Physico-chemical, Synthesis and Biological Activity, Department of Biology, Faculty of Natural Sciences and Life, Sciences of the Earth and the Universe, University Abou Bekr Belkaid Tlemcen, Tlemcen, Algeria
| | - Alfred Ngenge Tamfu
- Department of Chemical Engineering, School of Chemical Engineering and Mineral Industries, University of Ngaoundere, Ngaoundere, Cameroon
- Food Quality Control and Analysis Program, Ula Ali Kocman Vocational School, Mugla Sitki Kocman University, Mugla, Turkey
| | - Bousselham Ammara
- Microbiology Laboratory, University Hospital Center of Tlemcen, Tlemcen, Algeria
| | - Selcuk Kucukaydin
- Department of Medical Services and Techniques, Koycegiz Vocational School of Health Services, Mugla Sitki Kocman University, Mugla, Turkey
| | - Nawel Latti
- Laboratory of Applied Microbiology in Food, Biomedical, and Environment (LAMAABE), Department of Biology, Faculty of Sciences of Nature, Life, Earth, and Universe, Abou Bekr Belkaïd University of Tlemcen, Tlemcen, Algeria
| | - Abdelmounaim Khadir
- Laboratory of Applied Microbiology in Food, Biomedical, and Environment (LAMAABE), Department of Biology, Faculty of Sciences of Nature, Life, Earth, and Universe, Abou Bekr Belkaïd University of Tlemcen, Tlemcen, Algeria
- Department of Biology, Oran University, Oran, Algeria
| | - Mourad Bendahou
- Laboratory of Applied Microbiology in Food, Biomedical, and Environment (LAMAABE), Department of Biology, Faculty of Sciences of Nature, Life, Earth, and Universe, Abou Bekr Belkaïd University of Tlemcen, Tlemcen, Algeria
| | - El Hassane Anouar
- Department of Chemistry, College of Sciences and Humanities in Al-Kharj, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Ozgur Ceylan
- Food Quality Control and Analysis Program, Ula Ali Kocman Vocational School, Mugla Sitki Kocman University, Mugla, Turkey
| |
Collapse
|
8
|
Serrano I, Mil-Homens D, Pires RF, Bonifácio VDB, Guerreiro JF, Cunha E, Costa SS, Tavares L, Oliveira M. In Vivo Antimicrobial Activity of Nisin Z Against S. aureus and Polyurea Pharmadendrimer PURE G4OEI 48 Against P. aeruginosa from Diabetic Foot Infections. Antibiotics (Basel) 2025; 14:444. [PMID: 40426511 PMCID: PMC12108245 DOI: 10.3390/antibiotics14050444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Diabetic foot infections (DFIs) are commonly associated with frequent hospitalizations, limb amputations, and premature death due to the profile of the bacteria infecting foot ulcers. DFIs are generally colonized by a polymicrobial net of bacteria that grows in biofilms, developing an increased antimicrobial resistance to multiple antibiotics. DFI treatment is a hurdle, and the need to develop new therapies that do not promote resistance is urgent. Therefore, the antibacterial efficacy of Nisin Z (antimicrobial peptide), a core-shell polycationic polyurea pharmadendrimer (PUREG4OEI48) (antimicrobial polymer), and amlodipine (antihypertensive drug) was evaluated against S. aureus and P. aeruginosa isolated from a DFI and previously characterized. METHODS The antibacterial activity was analyzed in vitro by determining the minimal inhibitory concentration (MIC) and in vivo in a Galleria mellonella model by assessing the larvae survival and health index. RESULTS The results indicate that Nisin Z exhibited antibacterial activity against S. aureus in vivo, allowing larvae full survival, and no antibacterial activity against P. aeruginosa. Nisin Z may have reduced the antibacterial effectiveness of both PUREG4OEI48 and amlodipine. PUREG4OEI48 significantly increased the survival of the larvae infected with P. aeruginosa, while amlodipine showed no activity against both bacteria in vivo. CONCLUSIONS These findings suggest that both Nisin Z and PUREG4OEI48 could potentially be used individually as adjunct treatments for mild DFIs. However, further studies are needed to confirm these findings and assess the potential toxicity and efficacy of PUREG4OEI48 in more complex models.
Collapse
Affiliation(s)
- Isa Serrano
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477 Lisbon, Portugal; (J.F.G.); (E.C.); (L.T.); (M.O.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477 Lisbon, Portugal
| | - Dalila Mil-Homens
- iBB—Institute for Bioengineering and Biosciences and i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; (D.M.-H.); (R.F.P.); (V.D.B.B.)
| | - Rita F. Pires
- iBB—Institute for Bioengineering and Biosciences and i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; (D.M.-H.); (R.F.P.); (V.D.B.B.)
| | - Vasco D. B. Bonifácio
- iBB—Institute for Bioengineering and Biosciences and i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; (D.M.-H.); (R.F.P.); (V.D.B.B.)
- Bioengineering Department, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Joana F. Guerreiro
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477 Lisbon, Portugal; (J.F.G.); (E.C.); (L.T.); (M.O.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477 Lisbon, Portugal
| | - Eva Cunha
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477 Lisbon, Portugal; (J.F.G.); (E.C.); (L.T.); (M.O.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477 Lisbon, Portugal
| | - Sofia S. Costa
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation Towards Global Health (LA-REAL), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), R. da Junqueira 100, 1349-008 Lisbon, Portugal;
| | - Luís Tavares
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477 Lisbon, Portugal; (J.F.G.); (E.C.); (L.T.); (M.O.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477 Lisbon, Portugal
| | - Manuela Oliveira
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477 Lisbon, Portugal; (J.F.G.); (E.C.); (L.T.); (M.O.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculty of Veterinary Medicine, University of Lisbon, Av. Universidade Técnica, 1300-477 Lisbon, Portugal
- cE3c—Centre for Ecology, Evolution and Environmental Changes & CHANGE—Global Change and Sustainability Institute, Faculty of Sciences, University of Lisbon, Campo Grande 016, 1749-016 Lisbon, Portugal
| |
Collapse
|
9
|
Grace A, Sahu R, Owen DR, Dennis VA. Host-mimicking conditions promote Pseudomonas aeruginosa PA14 virulence gene expression. Front Microbiol 2025; 16:1557664. [PMID: 40351318 PMCID: PMC12062898 DOI: 10.3389/fmicb.2025.1557664] [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: 01/09/2025] [Accepted: 03/25/2025] [Indexed: 05/14/2025] Open
Abstract
Background Pseudomonas aeruginosa is a ubiquitous, opportunistic bacterium whose highly plastic genome and adaptable phenotype have yielded serious treatment challenges for immunocompromised patients. Antibiotic alternatives, such as anti-virulence therapeutics, have gained interest because they disable bacterial virulence mechanisms, thereby restoring the killing efficacy of host immunity or traditional antibiotics. Identifying successful anti-virulence therapeutics may require a paradigm shift from the decades-old antimicrobial susceptibility testing (AST) in Mueller Hinton broth to media that foster optimal virulence expression. Methods This study evaluates the virulence gene expression and activity of P. aeruginosa PA14 in host-mimicking conditions, represented by Dulbecco's Modified Eagle's Medium (DMEM) without serum, with fetal bovine serum (FBS), or with human serum (HuS) in comparison to standard antimicrobial susceptibility testing conditions, represented by Cation-adjusted Mueller Hinton broth (CAMHB). PA14 twitching motility and pyoverdine production were evaluated under these conditions. Results For the first time, our study reveals that culturing the highly virulent P. aeruginosa PA14 in host-mimicking media enhances the expression of multiple virulence therapeutic targets that are critical to host colonization and infection. RNA sequencing showed that multiple Type III Secretion (T3SS), Type I Secretion (T1SS), pyoverdine biosynthesis, uptake and efflux, and Type IV pili (T4P) initiation genes were promoted when PA14 was transitioned into host-mimicking conditions but remained unchanged when transitioned into standard AST conditions. Moreover, qPCR results disclosed that HuS and FBS delivered differential effects on the expression of membrane-associated virulence genes involved in host colonization. Our macroscopic PA14 twitching motility results aligned more closely with PA14 growth patterns than with virulence gene expression patterns. Our microtiter biofilm assay, however, revealed earlier biofilm formation in DMEM 0 than in AST conditions and both showed inhibited twitching motility in serum conditions. UV-Vis spectra showed that pyoverdine production aligned with our gene expression data, revealing higher pyoverdine production in serum conditions for planktonic PA14. Discussion Overall, our findings support using host-mimicking conditions to improve the expression of candidate targets for anti-virulence therapeutics against P. aeruginosa PA14 in a planktonic state. These recommendations may be broadly applicable for antivirulence therapeutic screening against multiple bacterial species at large.
Collapse
Affiliation(s)
- Amber Grace
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
| | - Rajnish Sahu
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
| | | | - Vida A. Dennis
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
| |
Collapse
|
10
|
Spaggiari C, Yamukujije C, Pieroni M, Annunziato G. Quorum sensing inhibitors (QSIs): a patent review (2019-2023). Expert Opin Ther Pat 2025:1-17. [PMID: 40219759 DOI: 10.1080/13543776.2025.2491382] [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: 07/11/2024] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
INTRODUCTION The collective behavior of bacteria is regulated by Quorum Sensing (QS), in which bacteria release chemical signals and express virulence genes in a cell density-dependent manner. Quorum Sensing inhibitors (QSIs) are a large class of natural and synthetic compounds that have the potential to competitively inhibit the Quorum Sensing (QS) systems of several pathogens blocking their virulence mechanisms. They are considered promising compounds to deal with antimicrobial resistance, providing an opportunity to develop new drugs against these targets. AREAS COVERED The present review represents a comprehensive analysis of patents and patent applications available on Espacenet and Google Patent, from 2019 to 2023 referring to the therapeutic use of Quorum Sensing inhibitors. EXPERT OPINION Unlike classical antibiotics, which target the basic cellular metabolic processes, QSIs provide a promising alternative to attenuating virulence and pathogenicity without putting selective pressure on bacteria. The general belief is that QSIs pose no or little selective pressure on bacteria since these do not affect their growth. To date, QSIs are seen as the most promising alternative to traditional antibiotics. The next big step in this area of research is its succession to the clinical stage.
Collapse
Affiliation(s)
| | | | - Marco Pieroni
- Department of Food and Drugs, University of Parma, Parma, Italy
| | | |
Collapse
|
11
|
Elkady FM, Badr BM, Saied E, Hashem AH, Abdel-Maksoud MA, Fatima S, Malik A, Aufy M, Hussein AM, Abdulrahman MS, Hashem HR. Green Biosynthesis of Bimetallic Copper Oxide-Selenium Nanoparticles Using Leaf Extract of Lagenaria Siceraria: Antibacterial, Anti-Virulence Activities Against Multidrug-Resistant Pseudomonas Aeruginosa. Int J Nanomedicine 2025; 20:4705-4727. [PMID: 40255676 PMCID: PMC12007613 DOI: 10.2147/ijn.s497494] [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: 10/16/2024] [Accepted: 03/20/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction Clinical isolates of Pseudomonas aeruginosa (P. aeruginosa) are among the most recovered bacteria with phenotypic antimicrobial resistance. Bimetallic nanoparticles (BNPs) have received much attention for antimicrobial activity in the last decade. This research aimed to biosynthesize bimetallic copper oxide-selenium nanoparticles (CuO-Se BNPs) and to assess its bioactivity on various P. aeruginosa clinical isolates. Methodology Based on the possible synergistic effects, CuO-Se BNPs were selected and biosynthesized using leaf extract of Lagenaria siceraria (L. siceraria) for the first time. The obtained BNPs were characterized using UV-vis spectroscopy, X-ray diffraction (XRD), energy dispersive X-ray spectroscopy (EDX), and transmission and scanning electron microscopes. The capability of Cu-Se BNPs to cease the growth of P. aeruginosa isolates and to reduce their virulence characters was evaluated. Also, different cell lines were used to assess its cytotoxicity and anticancer activity. Results The elemental composition of CuO and Se was revealed by the UV, XRD, and EDX data, indicating the synthesis of CuO-Se core shell BNPs with a size of 50 nm. In well diffusion assay, CuO-Se BNPs P. aeruginosa growth with 10-21 mm inhibition zone diameter and 38-95% inhibition. Also, the minimum inhibitory concentration and minimum bactericidal concentration were in a relatively wide range of 7.8-250 μg/mL and 31.2-500 μg/mL, respectively, with tolerance level range of 2-16. Additionally, CuO-Se BNPs shown anti-pyocyanin activity of 4.35-63.21% inhibition while the anti-proteolytic activity was in a range of 4.96-12.59% and anti-pyoverdine effect was in a range of 0.24-83.41%. The IC50 against Wi-38 normal cells was 267.2 µg/mL while the IC50 were 31.1 and 83.4 µg/mL against MCF-7 and Hep-G2, respectively, indicating promising anticancer activity. Conclusion This research demonstrates the promising antibacterial, anti-virulence, and antitumor properties with safe low concentrations of CuO-Se NPs, synthesized via an eco-friendly green synthesis method without the use of toxic chemicals, offering a sustainable and cost-effective alternative.
Collapse
Affiliation(s)
- Fathy M Elkady
- Microbiology and Immunology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Bahaa M Badr
- Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, Zarqa, Jordan
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Al-Azhar University (Assiut Branch), Assiut, Egypt
| | - Ebrahim Saied
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Amr H Hashem
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Mostafa A Abdel-Maksoud
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Sabiha Fatima
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Abdul Malik
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Mohammed Aufy
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Ahmed M Hussein
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Mohammed S Abdulrahman
- Microbiology and Immunology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Hany R Hashem
- Department of Microbiology and Immunology, Faculty of Pharmacy, Fayoum University, Al-Fayoum, Egypt
| |
Collapse
|
12
|
Mukherjee S, Chakravarty S, Haldar J. Revitalizing Antibiotics with Macromolecular Engineering: Tackling Gram-Negative Superbugs and Mixed Species Bacterial Biofilm Infections In Vivo. Biomacromolecules 2025; 26:2211-2226. [PMID: 40040432 DOI: 10.1021/acs.biomac.4c01520] [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: 03/06/2025]
Abstract
The escalating prevalence of multidrug-resistant Gram-negative pathogens, coupled with dwindling antibiotic development, has created a critical void in the clinical pipeline. This alarming issue is exacerbated by the formation of biofilms by these superbugs and their frequent coexistence in mixed-species biofilms, conferring extreme antibiotic tolerance. Herein, we present an amphiphilic cationic macromolecule, ACM-AHex, as an innovative antibiotic adjuvant to rejuvenate and repurpose resistant antibiotics, for instance, rifampicin, fusidic acid, erythromycin, and chloramphenicol. ACM-AHex mildly perturbs the bacterial membrane, enhancing antibiotic permeability, hampers efflux machinery, and produces reactive oxygen species, resulting in a remarkable 64-1024-fold potentiation in antibacterial activity. The macromolecule reduces bacterial virulence and macromolecule-drug cocktail significantly eradicate both mono- and multispecies bacterial biofilms, achieving >99.9% bacterial reduction in the murine biofilm infection model. Demonstrating potent biocompatibility across multiple administration routes, ACM-AHex offers a promising strategy to restore obsolete antibiotics and combat recalcitrant Gram-negative biofilm-associated infections, advocating for further clinical evaluation as a next-generation macromolecular antibiotic adjuvant.
Collapse
Affiliation(s)
- Sudip Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Sayan Chakravarty
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
- School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, Karnataka 560064, India
| |
Collapse
|
13
|
Pei Y, Hamar P, Pei DS. Deciphering Multidrug-Resistant Pseudomonas aeruginosa: Mechanistic Insights and Environmental Risks. TOXICS 2025; 13:303. [PMID: 40278619 PMCID: PMC12030990 DOI: 10.3390/toxics13040303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
The rise of multidrug-resistant (MDR) Pseudomonas aeruginosa (P. aeruginosa) presents a significant challenge to clinical treatment and environmental risks. This review delves into the complex mechanisms underlying MDR development in P. aeruginosa, such as genetic mutations, horizontal gene transfer (HGT), and the interaction between virulence factors and resistance genes. It evaluates current detection methods, from traditional bacteriology to advanced molecular techniques, emphasizing the need for rapid and accurate diagnostics. This review also examines therapeutic strategies, including broad-spectrum antibiotics, novel drug candidates, combination therapies, and innovative approaches like RNA interference, CRISPR-Cas9 gene editing, and bridge RNA-guided gene editing. Importantly, this review highlights the distribution, migration, and environmental risks of MDR P. aeruginosa, underscoring its adaptability to diverse environments. It concludes by stressing the necessity for continued research and development in antimicrobial resistance, advocating for an integrated approach that combines genomics, clinical practice, and environmental considerations to devise innovative solutions and preserve antibiotic efficacy.
Collapse
Affiliation(s)
- Yang Pei
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
- Chongqing No.11 Middle School, Chongqing 400061, China
- Chongqing Miankai Biotechnology Research Institute Co., Ltd., Chongqing 400025, China
| | - Péter Hamar
- Institute of Translational Medicine, Semmelweis University, Tűzoltó Utca 37-47, 1094 Budapest, Hungary
| | - De-Sheng Pei
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
14
|
Bellotti G, Cortimiglia C, Antinori ME, Cocconcelli PS, Puglisi E. Comprehensive genome-wide analysis for the safety assessment of microbial biostimulants in agricultural applications. Microb Genom 2025; 11:001391. [PMID: 40294085 PMCID: PMC12038027 DOI: 10.1099/mgen.0.001391] [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: 11/22/2024] [Accepted: 03/04/2025] [Indexed: 04/30/2025] Open
Abstract
Microbial biostimulants (MBs) offer a sustainable approach to agriculture by helping to reduce reliance on synthetic fertilizers. However, as MBs are intentionally released into the environment, their safety should be rigorously assessed. While taxa with qualified presumption of safety (QPS) benefit from established safety indications, non-QPS taxa lack such guidance. To address this gap, we propose a pipeline combining whole genome sequencing (WGS) and extensive literature search (ELS) data to evaluate microbial safety. We analysed public genomes of three QPS species (Rhodopseudomonas palustris, Bacillus velezensis, Priestia megaterium) and four non-QPS genera (Arthrobacter, Azotobacter, Azospirillum, Herbaspirillum), screening them for virulence factors (VFs), antimicrobial resistance (AMR) genes and mobile genetic elements (MGEs). Results confirmed the safety of QPS taxa, revealing no VFs and only a few intrinsic and non-clinically relevant AMRs. Among non-QPS taxa, VF hits were more prevalent in Azotobacter and Azospirillum spp., though they were mostly related to beneficial plant interactions rather than pathogenicity. AMR genes in non-QPS taxa were primarily associated with efflux pumps or were sporadically distributed. Notably, the only genus-wide pattern observed was that most Azospirillum and Herbaspirillum genomes harboured chromosomally encoded β-lactamases sharing similar genetic structures; however, the detected β-lactamase (bla) genes were distantly related to clinically relevant bla variants, and the absence of MGEs suggests a low risk of horizontal gene transfer, indicating the overall safety of these genera. In general, this WGS-ELS framework provides a robust tool for assessing the safety of non-QPS MBs, supporting regulatory decision-making and ensuring their safe use in sustainable agriculture while safeguarding public health.
Collapse
Affiliation(s)
- Gabriele Bellotti
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Claudia Cortimiglia
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Maria Elena Antinori
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Pier Sandro Cocconcelli
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Edoardo Puglisi
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza, Italy
| |
Collapse
|
15
|
Delgado-Nungaray JA, Figueroa-Yáñez LJ, Reynaga-Delgado E, García-Ramírez MA, Aguilar-Corona KE, Gonzalez-Reynoso O. Influence of Amino Acids on Quorum Sensing-Related Pathways in Pseudomonas aeruginosa PAO1: Insights from the GEM iJD1249. Metabolites 2025; 15:236. [PMID: 40278365 PMCID: PMC12029727 DOI: 10.3390/metabo15040236] [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: 03/07/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Amino acids (AAs) play a critical role in diseases such as cystic fibrosis where Pseudomonas aeruginosa PAO1 adapts its metabolism in response to host-derived nutrients. The adaptation influences virulence and complicates antibiotic treatment mainly for the antimicrobial resistance context. D- and L-AAs have been analyzed for their impact on quorum sensing (QS), a mechanism that regulates virulence factors. This research aimed to reconstruct the genome-scale metabolic model (GEM) of P. aeruginosa PAO1 to investigate the metabolic roles of D- and L-AAs in QS-related pathways. METHODS The updated GEM, iJD1249, was reconstructed by using protocols to integrate data from previous models and refined with well-standardized in silico media (LB, M9, and SCFM) to improve flux balance analysis accuracy. The model was used to explore the metabolic impact of D-Met, D-Ala, D-Glu, D-Ser, L-His, L-Glu, L-Arg, and L-Ornithine (L-Orn) at 5 and 50 mM in QS-related pathways, focusing on the effects on bacterial growth and carbon flux distributions. RESULTS Among the tested AAs, D-Met was the only one that did not enhance the growth rate of P. aeruginosa PAO1, while L-Arg and L-Orn increased fluxes in the L-methionine biosynthesis pathway, influencing the metH gene. These findings suggest a differential metabolic role for D-and L-AAs in QS-related pathways. CONCLUSIONS Our results shed some light on the metabolic impact of AAs on QS-related pathways and their potential role in P. aeruginosa virulence. Future studies should assess D-Met as a potential adjuvant in antimicrobial strategies, optimizing the concentration in combination with antibiotics to maximize its therapeutic effectiveness.
Collapse
Affiliation(s)
- Javier Alejandro Delgado-Nungaray
- Chemical Engineering Department, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico;
| | - Luis Joel Figueroa-Yáñez
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Zapopan 45019, Mexico;
| | - Eire Reynaga-Delgado
- Pharmacobiology Department, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico;
| | - Mario Alberto García-Ramírez
- Electronics Department, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico;
| | - Karla Esperanza Aguilar-Corona
- Food Engineering and Biotechnology, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico;
| | - Orfil Gonzalez-Reynoso
- Chemical Engineering Department, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico;
| |
Collapse
|
16
|
Khose GM, Vagolu SK, Aesoy R, Stefánsson ÍM, Ríkharðsson SG, Ísleifsdóttir D, Xu M, Homberset H, Tønjum T, Rongved P, Herfindal L, Viktorsson EÖ. Functionalized regioisomers of the natural product phenazines myxin and iodinin as potent inhibitors of Mycobacterium tuberculosis and human acute myeloid leukemia cells. Eur J Med Chem 2025; 285:117244. [PMID: 39788066 DOI: 10.1016/j.ejmech.2025.117244] [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: 11/17/2024] [Revised: 12/20/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
The natural bioactive products myxin and iodinin are phenazine 5,10-dioxides possessing potent anti-bacterial and anti-cancer activity in vitro. This work describes the synthesis and derivatization of new myxin and iodinin regioisomers, developed from 1,3-dihydroxyphenazine 5,10-dioxide. Compounds were evaluated for activity towards M. tuberculosis (Mtb) strains, a human AML cell line (MOLM-13), and two non-cancerous mammalian cell lines (NRK and H9c2). Highly potent analogs were developed having IC50 values against MTB down to 20 nM and 1.4 μM for human AML cells. 1-OH-3-O-alkyl substituted derivatives demonstrated high efficacy against Mtb and low toxicity in normal cells. 2,3-substituted regioisomers of myxin and iodinin were shown to be inactive, highlighting the importance of oxygen substituent in position 1 of the scaffold. A strong positive correlation between anti-MTB and anti-AML activity was revealed, suggesting a common mechanism of action in bacteria and cancer cells. These findings demonstrate the therapeutic potential of 1,3-O-functionalized phenazine 5,10-dioxides in chemotherapy for Mtb and AML and contribute to the structure-activity understanding of phenazine 5,10-dioxides with respect to their biological activity.
Collapse
Affiliation(s)
- Goraksha Machhindra Khose
- School of Health Sciences, Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, IS-107, Reykjavik, Iceland
| | - Siva Krishna Vagolu
- Unit for Genome Dynamics, Department of Microbiology, University of Oslo, N-0316, Oslo, Norway
| | - Reidun Aesoy
- Centre for Pharmacy, Department of Clinical Science, University of Bergen, Jonas Lies vei 87, N-5021, Bergen, Norway
| | - Ísak Máni Stefánsson
- School of Health Sciences, Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, IS-107, Reykjavik, Iceland
| | - Snorri Geir Ríkharðsson
- School of Health Sciences, Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, IS-107, Reykjavik, Iceland
| | - Dagmar Ísleifsdóttir
- School of Health Sciences, Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, IS-107, Reykjavik, Iceland
| | - Maonian Xu
- School of Health Sciences, Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, IS-107, Reykjavik, Iceland
| | - Håvard Homberset
- Unit for Genome Dynamics, Department of Microbiology, University of Oslo, N-0316, Oslo, Norway
| | - Tone Tønjum
- Unit for Genome Dynamics, Department of Microbiology, University of Oslo, N-0316, Oslo, Norway; Unit for Genome Dynamics, Department of Microbiology, Oslo University Hospital, N-0424, Oslo, Norway
| | - Pål Rongved
- School of Pharmacy, Department of Pharmaceutical Chemistry, University of Oslo, PO Box 1068 Blindern, N-0316, Oslo, Norway
| | - Lars Herfindal
- Centre for Pharmacy, Department of Clinical Science, University of Bergen, Jonas Lies vei 87, N-5021, Bergen, Norway
| | - Elvar Örn Viktorsson
- School of Health Sciences, Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, IS-107, Reykjavik, Iceland.
| |
Collapse
|
17
|
Wilson RM, Walker JM, Beld J, Yin K. Lactobacillus acidophilus (strain Scav) postbiotic metabolites reduce infection and modulate inflammation in an in vivo model of Pseudomonas aeruginosa wound infection. J Appl Microbiol 2025; 136:lxaf061. [PMID: 40068933 PMCID: PMC11951090 DOI: 10.1093/jambio/lxaf061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 03/30/2025]
Abstract
AIMS This study assessed the antibacterial, antibiofilm, and immunomodulatory activity of Lactobacillus acidophilus (strain Scav) postbiotic (LaP) in a mouse model of Pseudomonas aeruginosa wound infection and evaluated the bioactive components of the LaP. METHODS AND RESULTS LaP was tested for Pseudomonas aeruginosa clearance and immunomodulatory activity during wound infection. We show that LaP applied 1 h after infection reduced tissue bacterial burden within 24 h, and this reduction persisted for 5 days. Ciprofloxacin given once at the exact same time did not reduce bacteria load as compared to vehicle controls. LaP reduced plasma IL-6 and MCP-1 levels after 5 days. Wound tissue IL-6 and MCP-1 levels were increased in infected vehicle mice at 5 days, but tissues from LaP-treated mice were similar to sham controls. LaP increased tissue IL-10 (antiinflammatory cytokine) levels. Ciprofloxacin decreased plasma and tissue IL-6 compared to vehicle controls but did not affect MCP-1 or IL-10 levels. To elucidate antibacterial and antibiofilm metabolite(s) in LaP, fractionation followed by Ps. aeruginosa antagonistic activity assays were performed. This was followed by liquid chromatography coupled to mass spectrometry (LCMS) analysis. Our analyses identified a low molecular weight, polar molecule, which had both antibacterial and antibiofilm activity. CONCLUSIONS Lactobacillus acidophilus secretes an antibacterial and antibiofilm metabolite that reduced pathogen burden and resolved systemic inflammation in a Pseudomonas aeruginosa wound infection model.
Collapse
Affiliation(s)
- Rachael M Wilson
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Medicine and Life Sciences, Stratford, NJ 08084, USA
| | - Jean M Walker
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Medicine and Life Sciences, Stratford, NJ 08084, USA
| | - Joris Beld
- Department of Microbiology and Immunology, College of Medicine, Drexel University, Philadelphia, PA 19104, USA
| | - Kingsley Yin
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Medicine and Life Sciences, Stratford, NJ 08084, USA
| |
Collapse
|
18
|
Ivan FX, Tiew PY, Jaggi TK, Thng KX, Pang PH, Ong TH, Abisheganaden JA, Koh MS, Chotirmall SH. Sputum metagenomics reveals a multidrug resistant Pseudomonas-dominant severe asthma phenotype in an Asian population. Respirology 2025; 30:217-229. [PMID: 39622769 DOI: 10.1111/resp.14863] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 11/20/2024] [Indexed: 03/04/2025]
Abstract
BACKGROUND AND OBJECTIVE While the lung microbiome in severe asthma has been studied, work has employed targeted amplicon-based sequencing approaches without functional assessment with none focused on multi-ethnic Asian populations. Here we investigate the clinical relevance of microbial phenotypes of severe asthma in Asians using metagenomics. METHODS Prospective assessment of clinical, radiological, and immunological measures were performed in a multi-ethnic Asian severe asthma cohort (N = 70) recruited across two centres in Singapore. Sputum was subjected to shotgun metagenomic sequencing and patients followed up for a 2-year period. Metagenomic assessment of sputum microbiomes, resistomes and virulomes were related to clinical outcomes. RESULTS The lung microbiome in a multi-ethnic Asian cohort with severe asthma demonstrates an increased abundance of Pseudomonas species. Unsupervised clustering of sputum metagenomes identified two patient clusters: C1 (n = 52) characterized by upper airway commensals and C2 (n = 18) dominated by established respiratory pathogens including M. catarrhalis, S. aureus and most significantly P. aeruginosa. C2 patients demonstrated a significantly increased exacerbation frequency on 2-year follow up and an antimicrobial resistome characterized by multidrug resistance. Virulomes appear indistinguishable between severe asthmatics with or without co-existing bronchiectasis, and C2 patients exhibit increased gene expression related to biofilm formation, effector delivery systems and microbial motility. Independent comparison of the C2 cluster to a non-asthmatic bronchiectasis cohort demonstrates analogous airway microbial virulence patterns. CONCLUSION Sputum metagenomics demonstrates a multidrug-resistant Pseudomonas-dominant severe asthma phenotype in Asians, characterized by poor clinical outcome including increased exacerbations which is independent of co-existing bronchiectasis.
Collapse
Affiliation(s)
| | - Pei Yee Tiew
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Respiratory and Critical Care Medicine, Singapore General Hospital, Singapore, Singapore
| | - Tavleen Kaur Jaggi
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Kai Xian Thng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Collaborative Institute, Interdisciplinary Graduate Programme, Nanyang Technological University, Singapore, Singapore
| | - Pee Hwee Pang
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Thun How Ong
- Department of Respiratory and Critical Care Medicine, Singapore General Hospital, Singapore, Singapore
| | - John Arputhan Abisheganaden
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Mariko Siyue Koh
- Department of Respiratory and Critical Care Medicine, Singapore General Hospital, Singapore, Singapore
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| |
Collapse
|
19
|
lawal H, Saeed SI, Gaddafi MS, Kamaruzzaman NF. Green Nanotechnology: Naturally Sourced Nanoparticles as Antibiofilm and Antivirulence Agents Against Infectious Diseases. Int J Microbiol 2025; 2025:8746754. [PMID: 40041153 PMCID: PMC11876540 DOI: 10.1155/ijm/8746754] [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: 07/13/2024] [Accepted: 02/06/2025] [Indexed: 03/06/2025] Open
Abstract
The escalating threat of infectious diseases, exacerbated by antimicrobial resistance (AMR) and biofilm formation, necessitates innovative therapeutic strategies. This review presents a comprehensive exploration of the potential of nanoparticles synthesized from natural sources, including plant extracts, microbial products, and marine compounds, as antimicrobial agents. These naturally derived nanoparticles demonstrated significant antibiofilm and antivirulence effects, with specific examples revealing their capacity to reduce biofilm mass by up to 78% and inhibit bacterial quorum sensing by 65%. The integration of bioactive compounds, such as polyphenols and chitosan, facilitates nanoparticle stability and enhances antimicrobial efficacy, while green synthesis protocols reduce environmental risks. Notably, the review identifies the potential of silver nanoparticles synthesized using green tea extracts, achieving 85% inhibition of polymicrobial growth in vitro. Despite these promising results, challenges such as standardization of synthesis protocols and scalability persist. This study underscores the transformative potential of leveraging naturally sourced nanoparticles as sustainable alternatives to conventional antimicrobials, offering quantitative insights for their future application in combating mono- and polymicrobial infections.
Collapse
Affiliation(s)
- Habiba lawal
- Nanotechnology in Veterinary Medicine (NanoVet) Research Group, Faculty of Veterinary Medicine, University Malaysia Kelantan, Pengkalan Chepa, Kelantan, Malaysia
- Department of Public Health, Ministry of Animal Health, Husbandry and Fisheries, Birnin Kebbi, Kebbi State, Nigeria
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Shamsaldeen Ibrahim Saeed
- Nanotechnology in Veterinary Medicine (NanoVet) Research Group, Faculty of Veterinary Medicine, University Malaysia Kelantan, Pengkalan Chepa, Kelantan, Malaysia
- Department of Microbiology, Faculty of Veterinary Science, University of Nyala, Nyala, Sudan
| | - Mohammed Sani Gaddafi
- Department of Public Health, Ministry of Animal Health, Husbandry and Fisheries, Birnin Kebbi, Kebbi State, Nigeria
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Nor Fadhilah Kamaruzzaman
- Nanotechnology in Veterinary Medicine (NanoVet) Research Group, Faculty of Veterinary Medicine, University Malaysia Kelantan, Pengkalan Chepa, Kelantan, Malaysia
| |
Collapse
|
20
|
Badis A, Heleili N, Merradi M, Ayachi A, Martino PA, Meroni G, Soggiu A. Outbreak of Carbapenem-Resistant High-Risk Clone ST244 of Pseudomonas aeruginosa in Dogs and Cats in Algeria. Antibiotics (Basel) 2025; 14:230. [PMID: 40149042 PMCID: PMC11939618 DOI: 10.3390/antibiotics14030230] [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: 02/07/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Pseudomonas aeruginosa causes chronic infections in humans and animals, especially cats and dogs. This bacterium's ability to adapt and acquire antibiotic resistance traits may complicate and exacerbate antibacterial therapy. This study aimed to evaluate the antibiotic resistance patterns, virulence factors and ability to form biofilms of P. aeruginosa strains isolated from Algerian dogs and cats. Methods: Nineteen samples were collected from healthy and diseased dogs and cats. Isolates were studied for their antibiotic-resistance patterns (disc diffusion method) and biofilm formation (Microtiter assay) and were whole-genome sequenced (MinION). Results: Nineteen P. aeruginosa strains (15 from dogs and 4 from cats) were isolated. Antibiotic-resistance phenotypes were observed against amoxicillin-clavulanic acid (100%); meanwhile, resistance towards ticarcillin was 40% (dogs) and 25% (cats), ticarcillin-clavulanic acid was 13.33% and 25% for dogs and cats, respectively, and imipenem was 75% (cats) and 20% (dogs). Moreover, 95% of strains were biofilm-producers. Different antimicrobial resistance genes (ARGs) were found: beta-lactamase genes, mainly PAO, OXA-494, OXA-50 and OXA-396 and an aminoglycoside gene (aph(3')-IIb). The main high-risk sequence types (STs) were ST244, 2788, 388 and 1247. A large panel of virulence genes was detected: exoS, exoT, exoY, lasA, toxA, prpL, algD, rhIA and others. Conclusions: The genetic variety in antibiotic-resistance genes of resistant and virulent P. aeruginosa strains in dogs makes public health protection difficult. Continuous monitoring and research in compliance with the One Health policy are needed to solve this problem.
Collapse
Affiliation(s)
- Amina Badis
- ESPA Laboratory, Department of Veterinary Sciences, Institute of Veterinary Sciences and Agronomic Sciences, University of Batna 1, Batna 05000, Algeria; (N.H.); (M.M.)
| | - Nouzha Heleili
- ESPA Laboratory, Department of Veterinary Sciences, Institute of Veterinary Sciences and Agronomic Sciences, University of Batna 1, Batna 05000, Algeria; (N.H.); (M.M.)
- Department of Veterinary Sciences, Institute of Veterinary Sciences and Agronomic Sciences, University of Batna 1, Batna 05000, Algeria;
| | - Manel Merradi
- ESPA Laboratory, Department of Veterinary Sciences, Institute of Veterinary Sciences and Agronomic Sciences, University of Batna 1, Batna 05000, Algeria; (N.H.); (M.M.)
- Department of Microbiology and Biochemistry, Faculty of Natural and Life Sciences, University of Batna 2, Batna 05078, Algeria
| | - Ammar Ayachi
- Department of Veterinary Sciences, Institute of Veterinary Sciences and Agronomic Sciences, University of Batna 1, Batna 05000, Algeria;
| | - Piera Anna Martino
- One Health Unit, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (P.A.M.); (A.S.)
| | - Gabriele Meroni
- One Health Unit, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (P.A.M.); (A.S.)
| | - Alessio Soggiu
- One Health Unit, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (P.A.M.); (A.S.)
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
21
|
Vascon F, De Felice S, Gasparotto M, Huber ST, Catalano C, Chinellato M, Mezzetti R, Grinzato A, Filippini F, Maso L, Jakobi AJ, Cendron L. Snapshots of Pseudomonas aeruginosa SOS response reveal structural requisites for LexA autoproteolysis. iScience 2025; 28:111726. [PMID: 39898034 PMCID: PMC11787620 DOI: 10.1016/j.isci.2024.111726] [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: 05/28/2024] [Revised: 09/09/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025] Open
Abstract
Antimicrobial resistance poses a severe threat to human health and Pseudomonas aeruginosa stands out among the pathogens responsible for this emergency. The SOS response to DNA damage is crucial in bacterial evolution, influencing resistance development and adaptability in challenging environments, especially under antibiotic exposure. Recombinase A (RecA) and the transcriptional repressor LexA are the key players that orchestrate this process, determining either the silencing or the active transcription of the genes under their control. By integrating state-of-the-art structural approaches with in vitro binding and functional assays, we elucidated the molecular events activating the SOS response in P. aeruginosa, focusing on the RecA-LexA interaction. Our findings identify the conserved determinants and strength of the interactions that allow RecA to trigger LexA autocleavage and inactivation. These results provide the groundwork for designing novel antimicrobial strategies and exploring the potential translation of Escherichia coli-derived approaches, to address the implications of P. aeruginosa infections.
Collapse
Affiliation(s)
- Filippo Vascon
- Department of Biology, University of Padua, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Sofia De Felice
- Department of Biology, University of Padua, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Matteo Gasparotto
- Department of Biology, University of Padua, Via Ugo Bassi 58/b, 35131 Padova, Italy
- Department of Translational Brain Research, Central Institute of Mental Health (ZI), University of Heidelberg/Medical Faculty Mannheim, 68159 Mannheim, Germany
| | - Stefan T. Huber
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, 2628CD Delft, the Netherlands
| | - Claudio Catalano
- NanoImaging Services, 4940 Carroll Canyon Road, Suite 115, San Diego, CA 92121, USA
| | - Monica Chinellato
- Department of Biology, University of Padua, Via Ugo Bassi 58/b, 35131 Padova, Italy
- Department of Medicine, University of Padua, Via Giustiniani 2, 35121 Padova, Italy
| | - Riccardo Mezzetti
- Department of Biology, University of Padua, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Alessandro Grinzato
- Department of Biology, University of Padua, Via Ugo Bassi 58/b, 35131 Padova, Italy
- CM01 Beamline, European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - Francesco Filippini
- Department of Biology, University of Padua, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Lorenzo Maso
- Department of Biology, University of Padua, Via Ugo Bassi 58/b, 35131 Padova, Italy
- Aethon Therapeutics, Long Island City, NY 11101, USA
| | - Arjen J. Jakobi
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, 2628CD Delft, the Netherlands
| | - Laura Cendron
- Department of Biology, University of Padua, Via Ugo Bassi 58/b, 35131 Padova, Italy
| |
Collapse
|
22
|
Sandri A, Boschi F. Exploring Proteases as Alternative Molecular Targets to Tackle Inflammation in Cystic Fibrosis Respiratory Infections. Int J Mol Sci 2025; 26:1871. [PMID: 40076497 PMCID: PMC11899166 DOI: 10.3390/ijms26051871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/07/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Cystic fibrosis (CF) is characterized by chronic respiratory infections and excessive inflammation, driven by both host- and pathogen-derived proteases. The dysregulated activity of proteolytic enzymes such as neutrophil elastase (NE), cathepsin G, and matrix metalloproteases (MMPs) degrades lung tissue, exacerbates airway remodeling, and perpetuates inflammatory cycles. Concurrently, bacterial proteases from pathogens such as Pseudomonas aeruginosa and Staphylococcus aureus contribute to immune evasion and tissue destruction, compounding disease severity. Despite advances in antimicrobial and anti-inflammatory therapies, protease-driven lung damage remains a critical challenge. This review examines the dual role of host and bacterial proteases in CF pathophysiology, highlighting emerging protease-targeted therapies aimed at mitigating lung damage and inflammation. Strategies explored include the inhibition of NE, MMPs, and bacterial proteases, with a focus on innovative therapeutic approaches such as dual-function inhibitors, biologics, and advanced drug delivery systems. By restoring the protease-antiprotease balance, these interventions offer the potential to improve clinical outcomes and quality of life for CF patients.
Collapse
Affiliation(s)
- Angela Sandri
- Department of Diagnostics and Public Health, University of Verona, Strada Le Grazie 8-15, 37134 Verona, Italy;
- General and Upper GI Surgery Division, Azienda Ospedaliera Universitaria Integrata Verona, Piazzale Stefani 1, 37126 Verona, Italy
| | - Federico Boschi
- Department of Engineering for Innovation Medicine, University of Verona, Strada Le Grazie 8-15, 37134 Verona, Italy
| |
Collapse
|
23
|
Wang X, Gao K, Pan B, Wang B, Song Y, Guo W. The virulence trait and genotype distribution amongst the Pseudomonas aeruginosa clinical strains. BMC Microbiol 2025; 25:82. [PMID: 39979804 PMCID: PMC11841163 DOI: 10.1186/s12866-025-03754-6] [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: 09/17/2024] [Accepted: 01/09/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Pseudomonas aeruginosa is notorious for its complex virulence system and rapid adaptive drug resistance. This study aimed to compare the prevalence and genotype distribution of virulence genes in multidrug-sensitive and multidrug-resistant clinical strains of Pseudomonas aeruginosa. It is possible to better understand the genetic characteristics of Pseudomonas aeruginosa and carry out effective treatment and prevention measures. METHODS The genes phzS, aprA, plcH, toxA, pilA and exoU were detected amongst 184 clinical strains, whose cytotoxicity and biofilm formation ability were evaluated as well. Phenotypic screening for drug susceptibility was conducted by standard antimicrobial susceptibility test and interpreted according to standards established by CLSI. RESULTS A total of 94 multidrug-sensitive and 90 multidrug-resistant isolates were included in this study. Statistically significant relationship was observed in the frequency of the toxA (p = 0.002) and plcH (p = 0.001) genes between multidrug-resistant and multidrug-sensitive strains. Moreover, thirteen genotypes were observed in multidrug-sensitive strains, and seven of them were included in multidrug-resistant groups. There was statistically significant correlation found between the presence of genotype IV (p = 0.001) and genotype VII (p = 0.001) in two subgroups. Additionally, It was found that genotype III isolates exhibited most obvious cytotoxicity, and multidrug-resistant isolates of genotype III showed the most significant cytotoxicity. Moreover, the strains of strong biofilm-formation accounted for a relatively high proportion in genotype III and VI groups. CONCLUSION These virulence genes could form abundant genotype varieties, whose overall number is greater in multi-sensitive strains. In addition, particular genotypes were characteristically distributed and exhibited different cytotoxicity and biofilm-formation abilities.
Collapse
Affiliation(s)
- Xiaohuan Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, 111 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Kaijing Gao
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Baishen Pan
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, 111 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Beili Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, 111 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Yuanlin Song
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, 180 Feng lin Road, Shanghai, 200032, China.
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, 111 Yi Xue Yuan Road, Shanghai, 200032, China.
- Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Laboratory Medicine, Wusong Central Hospital, Baoshan District, Shanghai, China.
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China.
| |
Collapse
|
24
|
de Sousa T, Machado S, Caniça M, Ramos MJN, Santos D, Ribeiro M, Hébraud M, Igrejas G, Alves O, Costa E, Silva A, Lopes R, Poeta P. Pseudomonas aeruginosa: One Health approach to deciphering hidden relationships in Northern Portugal. J Appl Microbiol 2025; 136:lxaf037. [PMID: 39947205 DOI: 10.1093/jambio/lxaf037] [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] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 03/06/2025]
Abstract
AIMS Antimicrobial resistance in Pseudomonas aeruginosa represents a major global challenge in public and veterinary health, particularly from a One Health perspective. This study aimed to investigate antimicrobial resistance, the presence of virulence genes, and the genetic diversity of P. aeruginosa isolates from diverse sources. METHODS AND RESULTS The study utilized antimicrobial susceptibility testing, genomic analysis for resistance and virulence genes, and multilocus sequence typing to characterize a total of 737 P. aeruginosa isolates that were collected from humans, domestic animals, and aquatic environments in Northern Portugal. Antimicrobial resistance profiles were analyzed, and genomic approaches were employed to detect resistance and virulence genes. The study found a high prevalence of multidrug-resistant isolates, including high-risk clones such as ST244 and ST446, particularly in hospital sources and wastewater treatment plants. Key genes associated with resistance and virulence, including efflux pumps (e.g. MexA and MexB) and secretion systems (T3SS and T6SS), were identified. CONCLUSIONS This work highlights the intricate dynamics of multidrug-resistant P. aeruginosa across interconnected ecosystems in Northern Portugal. It underscores the importance of genomic studies in revealing the mechanisms of resistance and virulence, contributing to the broader understanding of resistance dynamics and informing future mitigation strategies.
Collapse
Affiliation(s)
- Telma de Sousa
- MicroART-Antibiotic Resistance Team, Department of Veterinary Sciences, University of Trás-os Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Associated Laboratory for Green Chemistry, University NOVA of Lisbon, 1099-085 Caparica, Portugal
| | - Sandro Machado
- MicroART-Antibiotic Resistance Team, Department of Veterinary Sciences, University of Trás-os Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Manuela Caniça
- National Reference Laboratory of Antibiotic Resistance and Healthcare Associated Infections, Department of Infectious Diseases, National Institute of Health Dr. Ricardo Jorge, 1649-016 Lisbon, Portugal
- Centre for the Studies of Animal Science (CECA) - Institute of Agrarian and Agri-Food Sciences and Technologies, University of Porto, 4051-401, Portugal
| | - Miguel J N Ramos
- National Reference Laboratory of Antibiotic Resistance and Healthcare Associated Infections, Department of Infectious Diseases, National Institute of Health Dr. Ricardo Jorge, 1649-016 Lisbon, Portugal
| | - Daniela Santos
- National Reference Laboratory of Antibiotic Resistance and Healthcare Associated Infections, Department of Infectious Diseases, National Institute of Health Dr. Ricardo Jorge, 1649-016 Lisbon, Portugal
| | - Miguel Ribeiro
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Chemistry Research Centre-Vila Real (CQ-VR), Food and Wine Chemistry Laboratory, University of Trás-os-Montes and Alto Douro, 5001-801 Vila Real, Portugal
| | - Michel Hébraud
- INRAE, Université Clermont Auvergne, UMR Microbiologie Environnement Digestif Santé (MEDiS), 63122 Saint-Genès-Champanelle, France
| | - Gilberto Igrejas
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Associated Laboratory for Green Chemistry, University NOVA of Lisbon, 1099-085 Caparica, Portugal
| | - Olimpia Alves
- Medical Centre of Trás-os-Montes and Alto Douro, Clinical Pathology Department, 5000-508 Vila Real, Portugal
| | - Eliana Costa
- Medical Centre of Trás-os-Montes and Alto Douro, Clinical Pathology Department, 5000-508 Vila Real, Portugal
| | - Augusto Silva
- INNO Veterinary Laboratories, R. Cândido de Sousa 15, 4710-300 Braga, Portugal
| | - Ricardo Lopes
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Department of Veterinary and Animal Sciences, University Institute of Health Sciences (IUCS), CESPU, 4585-116 Gandra, Portugal
| | - Patrícia Poeta
- MicroART-Antibiotic Resistance Team, Department of Veterinary Sciences, University of Trás-os Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Associated Laboratory for Green Chemistry, University NOVA of Lisbon, 1099-085 Caparica, Portugal
- CECAV - Veterinary and Animal Research Centre, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Veterinary and Animal Research Centre, Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| |
Collapse
|
25
|
Lombardi A, Tamburro M, Sammarco ML, Ripabelli G. Hospital environment as reservoir of Pseudomonas aeruginosa in human cases: a molecular epidemiology investigation in a hospital setting in central Italy. Lett Appl Microbiol 2025; 78:ovaf019. [PMID: 39929183 DOI: 10.1093/lambio/ovaf019] [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: 10/18/2024] [Revised: 01/10/2025] [Accepted: 02/08/2025] [Indexed: 02/22/2025]
Abstract
Pseudomonas aeruginosa is one of the main causes of hospital infections that are difficult to manage because of multidrug resistance (MDR). The aim of this study was to describe a molecular investigation on 19 clinical and 14 isolates from the environment in the Hub hospital of the Molise region, central Italy. Antimicrobial susceptibility was evaluated using BD Phoenix™ Automated Microbiology System. Pulsed-field gel electrophoresis (PFGE) with SpeI, enterobacterial repetitive intergenic consensus (ERIC-PCR), and random amplified polymorphic DNA-PCR were performed for genotyping. All 33 P. aeruginosa showed MDR phenotype. PFGE had 0.99 discriminatory power, underlining high heterogeneity among the strains. The genetic relatedness between two human isolates (H12 and H15) from neonatal intensive care (NICU) and one environmental strain (E1) collected from siphon of sink in the delivery room was noticeable, as well as between one strain from faucet in NICU (E2) and siphon sink (E5) from delivery room. The link between H12, H15, and E1 strains was corroborated by ERIC-PCR showing epidemiological concordance, although with a lower discriminatory power. The study findings strengthened the critical correlation between clinical P. aeruginosa and environment, according to previous molecular surveys on outbreaks occurred in Italy.
Collapse
Affiliation(s)
- Adele Lombardi
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| | - Manuela Tamburro
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| | - Michela Lucia Sammarco
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| | - Giancarlo Ripabelli
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| |
Collapse
|
26
|
Sk MS, Mwangomo R, Daniel L, Gilmore J. Solution Blow Spinning: An Emerging Nanomaterials-Based Wound-Care Technology. J Biomed Mater Res B Appl Biomater 2025; 113:e35513. [PMID: 39854136 DOI: 10.1002/jbm.b.35513] [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: 03/27/2024] [Revised: 09/09/2024] [Accepted: 11/10/2024] [Indexed: 01/26/2025]
Abstract
Application of one-dimensional nanofibers have witnessed exponential growth over the past few decades and are still emerging with their excellent physicochemical and electrical properties. The driving force behind this intriguing transition lies in their unique high surface-to-volume ratio, ubiquitous nanodomains, improved tensile strength, and flexibility to incorporate deliberate functionalities required for specific and advanced applications. Besides numerous benefits, nanomaterials may adversely interact with biological tissues and potentially be cytotoxic and carcinogenic. However, precisely engineered design can outperform the risk with myriad benefits. Wound care technologies are evolving, and products involved in wound care management have a yearly market value of $15-22 billion. Solution blow spinning (SBS) is a facile technique to synthesize biocompatible nanofibers with scalable processing variables for multidirectional biomedical applications. SBS is feasible for a wide range of thermoplastic polymers and nanomaterials to fabricate nanocomposites. This review will focus on the relevance of SBS technology for wound care, including dressings, drug delivery, tissue engineering scaffolds, and sensors.
Collapse
Affiliation(s)
- Md Salauddin Sk
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Ruth Mwangomo
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Luke Daniel
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Jordon Gilmore
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
27
|
Weingard B, Becker SL, Schneitler S, Trudzinski FC, Bals R, Wilkens H, Langer F. Risk factors for survival after lung transplantation in cystic fibrosis: impact of colonization with multidrug-resistant strains of Pseudomonas aeruginosa. Infection 2025:10.1007/s15010-025-02478-z. [PMID: 39883262 DOI: 10.1007/s15010-025-02478-z] [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: 11/15/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Lung transplantation is the ultimate treatment option for patients with advanced cystic fibrosis. Chronic colonization of these recipients with multidrug-resistant (MDR) pathogens may constitute a risk factor for an adverse outcome. We sought to analyze whether colonization with MDR pathogens, as outlined in the German classification of multiresistant Gram-negative bacteria (MRGN), was associated with the success of lung transplantation. METHODS We performed a monocentric retrospective analysis of 361 lung transplantations performed in Homburg, Germany, between 1995 and 2020. All recipients with a main diagnosis of cystic fibrosis (n = 69) were stratified into two groups based on colonization with Pseudomonas aeruginosa in view of MRGN before transplantation: no colonization and colonization without (n = 23) or with (n = 46) resistance to three or four antibiotic groups (3MRGN/4MRGN). Multivariable analyses were performed including various clinical parameters (preoperative data, postoperative data). RESULTS CF patients colonized with multidrug-resistant pathogens (Pseudomonas aeruginosa) classified as 3MRGN/4MRGN had poorer survival (median survival 16 years (without MRGN) versus 8 years (with MRGN), P = 0.048). Extracorporeal support (P = 0.014, HR = 2.929), re-transplantation (P = 0.023, HR = 2.303), female sex (P = 0.019, HR = 2.244) and 3MRGN/4MRGN (P = 0.036, HR = 2.376) were predictors of poor outcomes in the multivariate analysis. Co-colonization with the mold Aspergillus fumigatus was further associated with mortality risk in the 3MRGN/4MRGN group (P = 0.037, HR = 2.150). CONCLUSION Patients with cystic fibrosis and MDR colonization (Pseudomonas aeruginosa) are risk candidates for lung transplantation, targeted diagnostics and tailored anti-infective strategies are essential for survival after surgery. MDR colonization as expressed by MRGN may help to identify patients at increased risk to improve the organ allocation process.
Collapse
Affiliation(s)
- Bettina Weingard
- Internal Medicine V, Saarland University, 66421, Homburg/Saar, Germany
| | - Sören L Becker
- Institute of Medical Microbiology and Hygiene, Saarland University, 66421, Homburg/Saar, Germany
| | - Sophie Schneitler
- Institute of Medical Microbiology and Hygiene, Saarland University, 66421, Homburg/Saar, Germany
| | | | - Robert Bals
- Internal Medicine V, Saarland University, 66421, Homburg/Saar, Germany
| | - Heinrike Wilkens
- Internal Medicine V, Saarland University, 66421, Homburg/Saar, Germany
| | - Frank Langer
- Department of Thoracic Surgery, Saarland University, 66421, Homburg/Saar, Germany.
| |
Collapse
|
28
|
Pajaro-Castro N, Diaz-Morales E, Hoyos K, Ibañez-Bersinger C. Whole-Genome Sequencing of Resistance, Virulence and Regulation Genes in Extremely Resistant Strains of Pseudomonas aeruginosa. Med Sci (Basel) 2025; 13:6. [PMID: 39846701 PMCID: PMC11755660 DOI: 10.3390/medsci13010006] [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: 11/09/2024] [Revised: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pseudomonas aeruginosa is a clinically significant opportunistic pathogen, renowned for its ability to acquire and develop diverse mechanisms of antibiotic resistance. This study examines the resistance, virulence, and regulatory mechanisms in extensively drug-resistant clinical strains of P. aeruginosa. METHODS Antibiotic susceptibility was assessed using the Minimum Inhibitory Concentration (MIC) method, and whole-genome sequencing (WGS) was performed on the Illumina NovaSeq platform. RESULTS The analysis demonstrated a higher prevalence of virulence genes compared to resistance and regulatory genes. Key virulence factors identified included secretion systems, motility, adhesion, and biofilm formation. Resistance mechanisms observed comprised efflux pumps and beta-lactamases, while regulatory systems involved two-component systems, transcriptional regulators, and sigma factors. Additionally, phenotypic profiles were found to correlate with resistance genes identified through genotypic analysis. CONCLUSIONS This study underscores the significant resistance and virulence of the clinical P. aeruginosa strains analyzed, highlighting the urgent need for alternative strategies to address infections caused by extensively drug-resistant bacteria.
Collapse
Affiliation(s)
- Nerlis Pajaro-Castro
- Medical and Pharmaceutical Sciences Group, Faculty of Health Sciences, University of Sucre, Sincelejo 700001, Sucre, Colombia;
- Salud Social Clinic, Sincelejo 700001, Sucre, Colombia; (E.D.-M.); (K.H.)
| | - Erick Diaz-Morales
- Salud Social Clinic, Sincelejo 700001, Sucre, Colombia; (E.D.-M.); (K.H.)
| | - Kenia Hoyos
- Salud Social Clinic, Sincelejo 700001, Sucre, Colombia; (E.D.-M.); (K.H.)
| | - Cristhian Ibañez-Bersinger
- Medical and Pharmaceutical Sciences Group, Faculty of Health Sciences, University of Sucre, Sincelejo 700001, Sucre, Colombia;
| |
Collapse
|
29
|
Kumar S, Singh M. Combating Amoebic Meningoencephalitis in Kerala and other Indian States. RECENT ADVANCES IN ANTI-INFECTIVE DRUG DISCOVERY 2025; 20:2-4. [PMID: 40302547 DOI: 10.2174/0127724344356382241024101557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 05/02/2025]
Affiliation(s)
- Sanjesh Kumar
- Institute of Pharmaceutical Research, GLA University Mathura, Uttar Pradesh, 281406, India
- Rakshpal Bahadur College of Pharmacy, Bareilly, India
| | - Mansi Singh
- Institute of Pharmaceutical Research, GLA University Mathura, Uttar Pradesh, 281406, India
- Rakshpal Bahadur College of Pharmacy, Bareilly, India
| |
Collapse
|
30
|
Elshimy R, El-Shiekh RA, Okba MM, Ashour RMS, Ibrahim MA, Hassanen EI, Aboul-Ella H, Ali ME. Unveiling the antimicrobial, antivirulence, and wound-healing accelerating potentials of resveratrol against carbapenem-resistant Pseudomonas aeruginosa (CRPA)-septic wound in a murine model. Inflammopharmacology 2025; 33:401-416. [PMID: 39508957 PMCID: PMC11799074 DOI: 10.1007/s10787-024-01591-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024]
Abstract
Pseudomonas aeruginosa is a repertoire of several virulence factors that create a frightening high pathogenicity level as well as high antimicrobial resistance toward commercially used antibiotics. Therefore, finding a new alternative to traditional antimicrobials is a must. Resveratrol is a very famous phytochemical that harbors many beneficial health properties by possessing antibacterial, anti-inflammatory, and antioxidant properties. The current study aimed to explore the antimicrobial efficacy of resveratrol against P. aeruginosa and explore its ability to accelerate wound healing in a murine model. The obtained results revealed the potent antimicrobial, antivirulence, and wound-healing accelerating potentials of resveratrol against carbapenem-resistant P. aeruginosa (CRPA)-septic wounds. It significantly lowered the transcript levels of P. aeruginosa virulent genes toxA, pelA, and lasB. Additionally, resveratrol significantly accelerated skin wound healing by shortening the inflammatory phase and promoting re-vascularization, cell proliferation, re-epithelialization, and collagen deposition. Furthermore, it increased the immunoexpression of αSMA along with a reduction of the mRNA levels of VEGF, IL-1β, and TNF-α genes. Resveratrol has high therapeutic potential for the treatment of P. aeruginosa wound infection and is a prospective and promising candidate for this problem.
Collapse
Affiliation(s)
- Rana Elshimy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
- Department of Microbiology and Immunology, Egyptian Drug Authority, Cairo, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Mona M Okba
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Giza, Egypt.
| | - Rehab M S Ashour
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Marwa A Ibrahim
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Eman I Hassanen
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Hassan Aboul-Ella
- Department of Microbiology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Merhan E Ali
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
31
|
Salim SNM, Din NIM, Rashid R, Hitam SAS, Deris ZZ. Risk Factors and Outcomes of Multidrug-resistant Pseudomonas aeruginosa in Kelantan, Malaysia: A Multicenter Case-control Study. SAUDI JOURNAL OF MEDICINE & MEDICAL SCIENCES 2025; 13:18-25. [PMID: 39935999 PMCID: PMC11809758 DOI: 10.4103/sjmms.sjmms_429_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/05/2024] [Accepted: 09/22/2024] [Indexed: 02/13/2025]
Abstract
Background Increasing trend and spread of multidrug-resistant Pseudomonas aeruginosa (MDR-PA) in clinical settings is a great challenge in managing patients with infections caused by this pathogen. Objective To determine the risk factors and outcomes of MDR-PA acquisition in the northeastern state of Malaysia. In addition, this study also reported on the susceptibility pattern and common resistant genes among MDR-PA. Materials and Methods MDR-PA isolates obtained between March 2021 and February 2022 from all four major hospitals in the state of Kelantan, Malaysia, were submitted for susceptibility and resistant genes identification. The clinical data of the patients with MDR-PA were retrospectively reviewed. The risk factors and outcomes of MDR-PA acquired patients were analyzed by comparing with patients who acquired susceptible-PA while admitted to the same hospital during the study time. Results A total of 100 MDR-PA and 100 susceptible-PA cases were included. Ceftolozane-tazobactam was susceptible in 41.3% of MDR-PA compared to only 4%-8% with other β-lactams. About half (46%) of the MDR-PA isolates harbored the bla -NDM-1 gene, but none had the bla -OXA-48 gene. Factors independently associated with MDR-PA acquisitions were age (OR: 1.02; P = 0.028), genitourinary disorder (OR: 6.89; P = 0.001), and central venous catheter (OR: 3.18; P = 0.001). In addition, MDR-PA acquisitions were found to be associated with antimicrobial treatment failure (41.1% vs. 25.0%; P = 0.001) and mortality (40.0% versus 6.0%; P <0.001). Conclusion Most of the MDR-PA strains in Kelantan tertiary hospitals harbored the bla -NDM-1 gene, which is easily transmissible and can lead to an outbreak. Nonetheless, a significant number of the MDR-PA isolates were still susceptible to ceftolozane-tazobactam.
Collapse
Affiliation(s)
- Siti Norfairuz Md. Salim
- Department of Medical Microbiology and Parasitology, Universiti Sains Malaysia Health Campus, Kota Bharu, Malaysia
- Department of Pathology, Hospital Raja Permaisuri Bainun, Jalan Raja Ashman Shah, Ipoh, Perak Darul Ridzuan, Malaysia
| | - Nurul Izzah Md Din
- Unit of Microbiology, Hospital Raja Perempuan Zainab II, Kota Bharu, Malaysia
| | - Rosnita Rashid
- Unit of Microbiology, Hospital Tanah Merah, Tanah Merah, Malaysia
| | - Sharifah Aisyah Sayed Hitam
- Unit of Microbiology, Hospital Raja Perempuan Zainab II, Kota Bharu, Malaysia
- Unit of Microbiology, Hospital Sultan Ismail Petra, Kuala Krai, Kelantan, Malaysia
| | - Zakuan Zainy Deris
- Department of Medical Microbiology and Parasitology, Universiti Sains Malaysia Health Campus, Kota Bharu, Malaysia
- Microbiology Laboratory, Hospital USM, Health Campus, USM, Kubang Kerian, Malaysia
| |
Collapse
|
32
|
Koujalagi T, Ruhal R. Mitigating Health Risks Through Environmental Tracking of Pseudomonas aeruginosa. Curr Microbiol 2024; 82:57. [PMID: 39718600 DOI: 10.1007/s00284-024-04036-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
Pseudomonas aeruginosa is a prevalent nosocomial pathogen and a significant reservoir of antimicrobial resistance genes in residential and built environments. It is also widespread in various indoor and outdoor settings, including sewage, surface waters, soil, recreational waters (both treated and untreated), and industrial effluents. Surveillance efforts for P. aeruginosa are primarily focused on hospitals rather than built environments. However, evidence links multidrug-resistant P. aeruginosa of human origin with activity in built environments and hospital settings. Consequently, tracking this pathogen across all environments is crucial for understanding the mechanisms of reverse transmission from built environments to humans. This review explores public health hygiene by examining the prevalence of P. aeruginosa in various environments, its sequence types, the factors contributing to multidrug resistance, and the identification methods through global surveillance. Whole-genome sequencing with sequence typing and real-time quantitative PCR are widely used to identify and study antimicrobial-resistant strains worldwide. Additionally, advanced techniques such as functional metagenomics, next-generation sequencing, MALDI-TOF, and biosensors are being extensively employed to detect antimicrobial-resistant strains and mitigate the ongoing evolution of bacterial resistance to antibiotics. Our review strongly underscores the importance of environmental monitoring of P. aeruginosa in preventing human infections. Furthermore, strategic planning in built environments is essential for effective epidemiological surveillance of P. aeruginosa and the development of comprehensive risk assessment models.
Collapse
Affiliation(s)
- Tushar Koujalagi
- School of Bio Science and Technology, VIT University, Vellore, Tamil Nadu, 632014, India
| | - Rohit Ruhal
- School of Bio Science and Technology, VIT University, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
33
|
Li D, Zhang K, Xue X, Bai Z, Yang L, Qi J, Suolang S. An Epidemiological Study on Salmonella in Tibetan Yaks from the Qinghai-Tibet Plateau Area in China. Animals (Basel) 2024; 14:3697. [PMID: 39765601 PMCID: PMC11672581 DOI: 10.3390/ani14243697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Salmonella is an important foodborne pathogen that can cause a range of illnesses in humans; it has also been a key focus for monitoring in the field of public health, including gastroenteritis, sepsis, and arthritis, and can also cause a decline in egg production in poultry and diarrhea and abortion in livestock, leading to death in severe cases, resulting in huge economic losses. This study aimed to investigate the isolation rate, antimicrobial resistance, serotypes, and genetic diversity of Salmonella isolated from yak feces in various regions on the Qinghai-Tibet Plateau. A total of 1222 samples of yak dung were collected from major cities in the Qinghai-Tibet Plateau area, and the sensitivity of the isolated bacteria to 10 major classes of antibiotics was determined using the K-B paper disk diffusion method for drug susceptibility. Meanwhile, the serotypes of the isolated bacteria were analyzed using the plate agglutination test for serum antigens, and their carriage of drug resistance and virulence genes was determined using PCR and gel electrophoresis experiments. The isolated bacteria were also classified using MLST (Multi-Locus Sequence Typing). The overall isolation rate for Salmonella was 18.25% (223/1222), and the results of the antibiotic susceptibility tests showed that 98.65% (220/223) of the isolated bacteria were resistant to multiple antibiotics. In the 223 isolates of Salmonella, eight classes of 20 different resistance genes, 30 serotypes, and 15 different types of virulence genes were detected. The MLST analysis identified 45 distinct sequence types (STs), including five clonal complexes, of which ST34, ST11, and ST19 were the most common. These findings contribute valuable information about strain resources, genetic profiles, and typing data for Salmonella in the Qinghai-Tibet Plateau area, facilitating improved bacterial surveillance, identification, and control in yak populations. They also provide certain data supplements for animal Salmonella infections globally, filling research gaps.
Collapse
Affiliation(s)
- Dengyu Li
- College of Animal Science, Tibet Agricultural and Animal Husbandry University, Nyingchi 860000, China; (D.L.)
| | - Kaiqin Zhang
- College of Animal Science, Tibet Agricultural and Animal Husbandry University, Nyingchi 860000, China; (D.L.)
| | - Xiaofeng Xue
- College of Animal Science, Tibet Agricultural and Animal Husbandry University, Nyingchi 860000, China; (D.L.)
| | - Zhanchun Bai
- College of Animal Science, Tibet Agricultural and Animal Husbandry University, Nyingchi 860000, China; (D.L.)
| | - La Yang
- College of Animal Science, Tibet Agricultural and Animal Husbandry University, Nyingchi 860000, China; (D.L.)
| | - Jingjing Qi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences China, Shanghai 200241, China
| | - Sizhu Suolang
- College of Animal Science, Tibet Agricultural and Animal Husbandry University, Nyingchi 860000, China; (D.L.)
- “Fourteenth Five-Year Plan” China Agricultural Rural Ministry Key Laboratory (Jointly Built by the Ministry and Provincial Government), Nyingchi 860000, China
| |
Collapse
|
34
|
Krucon T, Uhrynowski W, Piatkowska K, Styczynski M, Stasiuk R, Dziewit L, Drewniak L. Application of xylene-degrading bacteria in the treatment of soil contaminated with petroleum hydrocarbons - A comprehensive laboratory to pilot-scale analysis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177501. [PMID: 39536865 DOI: 10.1016/j.scitotenv.2024.177501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/20/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Petroleum hydrocarbons, including both aliphatic (gasoline, mineral oil) and aromatic compounds (BTEX), are known for their harmful effects on ecosystems and human health. Despite many studies, large-scale treatment of contaminated soils continues to be challenging, especially at lower temperatures. The use of metabolically-versatile, psychrotolerant, cold-active microorganisms, seems a promising, cost-effective and eco-friendly solution to boost remediation rates. In this study, a suitable microbial consortium was prepared and tested both in lab- and pilot-scale. To achieve the best bioremediation results, bacterial strains were isolated from BTEX-contaminated soil and then tested for the desired traits over a wide range of conditions. Of 5 preselected strains, 3 Pseudomonas strains capable of denitrification and aerobic/anaerobic degradation of hydrocarbons (up to 41.53±7.39 %), further characterized by a broad temperature (4-37 °C), pH (3-4 to 11) and salinity (0-8 %) tolerance, as well as resistance to freezing, were selected. Physiological studies were supported by genetic analyses, which indicated the presence of both alkB and xylM genes, and excluded similarity of the strains to the known opportunistic pathogens. To further confirm the applicability of the consortium, lab-scale analyses were followed by comprehensive pilot-scale tests on ~5 m3 biopile/biocell, at different conditions. The results revealed increased efficacy of the consortium in bioremediation, when compared to biostimulated indigenous strains, for volatile hydrocarbons (93 % vs 88 %) and mineral oil (23 % vs 15 %), as well as 175 % and 136 % acceleration of remediation for the respective compounds in terms of time needed to complete the process. Moreover, the high survivability and metabolic activity of the consortium at different temperatures indicate the possibility of its year-round use for bioremediation of soil contaminated with petroleum hydrocarbons. The study proves the potential of specialized bacteria in the removal of pollutants, and emphasizes the role of bio-based strategies in addressing complex environmental challenges and remediation of contaminated sites.
Collapse
Affiliation(s)
- Tomasz Krucon
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| | | | - Katarzyna Piatkowska
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| | - Michal Styczynski
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| | - Robert Stasiuk
- Department of Geomicrobiology, Institute of Microbiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| | - Lukasz Dziewit
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| | - Lukasz Drewniak
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; Department of Microbiology, Institute of Biology, Jan Kochanowski University, 25-406 Kielce, Poland.
| |
Collapse
|
35
|
Ren Y, Zhu R, You X, Li D, Guo M, Fei B, Liu Y, Yang X, Liu X, Li Y. Quercetin: a promising virulence inhibitor of Pseudomonas aeruginosa LasB in vitro. Appl Microbiol Biotechnol 2024; 108:57. [PMID: 38180553 PMCID: PMC10770215 DOI: 10.1007/s00253-023-12890-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 11/15/2023] [Accepted: 11/19/2023] [Indexed: 01/06/2024]
Abstract
With the inappropriate use of antibiotics, antibiotic resistance has emerged as a major dilemma for patients infected with Pseudomonas aeruginosa. Elastase B (LasB), a crucial extracellular virulence factor secreted by P. aeruginosa, has been identified as a key target for antivirulence therapy. Quercetin, a natural flavonoid, exhibits promising potential as an antivirulence agent. We aim to evaluate the impact of quercetin on P. aeruginosa LasB and elucidate the underlying mechanism. Molecular docking and molecular dynamics simulation revealed a rather favorable intermolecular interaction between quercetin and LasB. At the sub-MICs of ≤256 μg/ml, quercetin was found to effectively inhibit the production and activity of LasB elastase, as well as downregulate the transcription level of the lasB gene in both PAO1 and clinical strains of P. aeruginosa. Through correlation analysis, significant positive correlations were shown between the virulence gene lasB and the QS system regulatory genes lasI, lasR, rhlI, and rhlR in clinical strains of P. aeruginosa. Then, we found the lasB gene expression and LasB activity were significantly deficient in PAO1 ΔlasI and ΔlasIΔrhlI mutants. In addition, quercetin significantly downregulated the expression levels of regulated genes lasI, lasR, rhlI, rhlR, pqsA, and pqsR as well as effectively attenuated the synthesis of signaling molecules 3-oxo-C12-HSL and C4-HSL in the QS system of PAO1. Quercetin was also able to compete with the natural ligands OdDHL, BHL, and PQS for binding to the receptor proteins LasR, RhlR, and PqsR, respectively, resulting in the formation of more stabilized complexes. Taken together, quercetin exhibits enormous potential in combating LasB production and activity by disrupting the QS system of P. aeruginosa in vitro, thereby offering an alternative approach for the antivirulence therapy of P. aeruginosa infections. KEY POINTS: • Quercetin diminished the content and activity of LasB elastase of P. aeruginosa. • Quercetin inhibited the QS system activity of P. aeruginosa. • Quercetin acted on LasB based on the QS system.
Collapse
Affiliation(s)
- Yanying Ren
- Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Rui Zhu
- Henan University of Chinese Medicine, Zhengzhou, 450046, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450002, China
| | - Xiaojuan You
- Henan University of Chinese Medicine, Zhengzhou, 450046, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450002, China
| | - Dengzhou Li
- Henan University of Chinese Medicine, Zhengzhou, 450046, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450002, China
| | - Mengyu Guo
- Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Bing Fei
- Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Ying Liu
- Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Ximing Yang
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Peking, 100700, China.
| | - Xinwei Liu
- Henan University of Chinese Medicine, Zhengzhou, 450046, China.
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450002, China.
| | - Yongwei Li
- Henan University of Chinese Medicine, Zhengzhou, 450046, China.
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450002, China.
- The Key Laboratory of Pathogenic Microbes & Antimicrobial Resistance Surveillance of Zhengzhou, Zhengzhou, 450002, China.
- Henan Engineering Research Center for Identification of Pathogenic Microbes, Zhengzhou, 450002, China.
- Henan Provincial Key Laboratory of Antibiotics-Resistant Bacterial Infection Prevention & Therapy with Traditional Chinese Medicine, Zhengzhou, 450002, China.
| |
Collapse
|
36
|
Do DT, Yang MR, Vo TNS, Le NQK, Wu YW. Unitig-centered pan-genome machine learning approach for predicting antibiotic resistance and discovering novel resistance genes in bacterial strains. Comput Struct Biotechnol J 2024; 23:1864-1876. [PMID: 38707536 PMCID: PMC11067008 DOI: 10.1016/j.csbj.2024.04.035] [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: 10/11/2023] [Revised: 04/13/2024] [Accepted: 04/13/2024] [Indexed: 05/07/2024] Open
Abstract
In current genomic research, the widely used methods for predicting antimicrobial resistance (AMR) often rely on prior knowledge of known AMR genes or reference genomes. However, these methods have limitations, potentially resulting in imprecise predictions owing to incomplete coverage of AMR mechanisms and genetic variations. To overcome these limitations, we propose a pan-genome-based machine learning approach to advance our understanding of AMR gene repertoires and uncover possible feature sets for precise AMR classification. By building compacted de Brujin graphs (cDBGs) from thousands of genomes and collecting the presence/absence patterns of unique sequences (unitigs) for Pseudomonas aeruginosa, we determined that using machine learning models on unitig-centered pan-genomes showed significant promise for accurately predicting the antibiotic resistance or susceptibility of microbial strains. Applying a feature-selection-based machine learning algorithm led to satisfactory predictive performance for the training dataset (with an area under the receiver operating characteristic curve (AUC) of > 0.929) and an independent validation dataset (AUC, approximately 0.77). Furthermore, the selected unitigs revealed previously unidentified resistance genes, allowing for the expansion of the resistance gene repertoire to those that have not previously been described in the literature on antibiotic resistance. These results demonstrate that our proposed unitig-based pan-genome feature set was effective in constructing machine learning predictors that could accurately identify AMR pathogens. Gene sets extracted using this approach may offer valuable insights into expanding known AMR genes and forming new hypotheses to uncover the underlying mechanisms of bacterial AMR.
Collapse
Affiliation(s)
- Duyen Thi Do
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ming-Ren Yang
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Department of Electrical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Tran Nam Son Vo
- Department of Business Administration, College of Management, Lunghwa University of Science and Technology, Taoyuan City, Taiwan
| | - Nguyen Quoc Khanh Le
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wei Wu
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
37
|
Carević T, Kolarević S, Kolarević MK, Nestorović N, Novović K, Nikolić B, Ivanov M. Citrus flavonoids diosmin, myricetin and neohesperidin as inhibitors of Pseudomonas aeruginosa: Evidence from antibiofilm, gene expression and in vivo analysis. Biomed Pharmacother 2024; 181:117642. [PMID: 39486364 DOI: 10.1016/j.biopha.2024.117642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Citrus flavonoids are group of bioactive polyphenols. Here, we investigated the potential of diosmin, myricetin and neohesperidin as possible inhibitors of Pseudomonas aeruginosa. This bacterium is a major clinical challenge due to its propensity to form resistant biofilm. The aims of this study were to examine flavonoids antibacterial activity using the microdilution method, assays intended to determine several antibiofilm mechanisms (crystal violet, congo red binding, extracellular DNA (eDNA) test and confocal laser scanning microscopy (CLSM) live/dead cell imaging), followed by virulence genes RT-qPCR analysis. Furthermore, we aimed to examine in vivo toxicity of the compounds as well as their efficacy in P. aeruginosa zebrafish embryo infection model. Minimal inhibitory concentrations of tested flavonoids towards P. aeruginosa were in range 0.05 - 0.4 mg/mL. A high potential of the compounds to disturb both the formation of the bacterial biofilm and its eradication was recorded, including significant reduction in biofilm biomass, exopolysaccharide and eDNA production. Biofilm treatment with diosmin resulted in the lowest percentage of live microbial cells as observed in the CLSM live/dead cell imaging. The lasI, pvdS, and rhlC genes were found to be downregulated in the presence of diosmin and myricetin. Only diosmin stood out as non-embryotoxic. Consequently, in vivo analysis using a zebrafish model of P. aeruginosa infection showed an antivirulence effect of diosmin. Our findings suggest that diosmin could be potential candidate for the development of new agent that target P. aeruginosa infections by reducing its virulence mechanisms.
Collapse
Affiliation(s)
- Tamara Carević
- Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, Belgrade 11108, Serbia
| | - Stoimir Kolarević
- Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, Belgrade 11108, Serbia
| | - Margareta Kračun Kolarević
- Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, Belgrade 11108, Serbia
| | - Nataša Nestorović
- Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, Belgrade 11108, Serbia
| | - Katarina Novović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, Belgrade 11042, Serbia
| | - Biljana Nikolić
- Faculty of Biology, University of Belgrade, Studentski trg 16, Belgrade 11000, Serbia
| | - Marija Ivanov
- Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, Belgrade 11108, Serbia.
| |
Collapse
|
38
|
Zanditenas E, Ankri S. Unraveling the interplay between unicellular parasites and bacterial biofilms: Implications for disease persistence and antibiotic resistance. Virulence 2024; 15:2289775. [PMID: 38058008 PMCID: PMC10761080 DOI: 10.1080/21505594.2023.2289775] [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: 07/26/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023] Open
Abstract
Bacterial biofilms have attracted significant attention due to their involvement in persistent infections, food and water contamination, and infrastructure corrosion. This review delves into the intricate interactions between bacterial biofilms and unicellular parasites, shedding light on their impact on biofilm formation, structure, and function. Unicellular parasites, including protozoa, influence bacterial biofilms through grazing activities, leading to adaptive changes in bacterial communities. Moreover, parasites like Leishmania and Giardia can shape biofilm composition in a grazing independent manner, potentially influencing disease outcomes. Biofilms, acting as reservoirs, enable the survival of protozoan parasites against environmental stressors and antimicrobial agents. Furthermore, these biofilms may influence parasite virulence and stress responses, posing challenges in disease treatment. Interactions between unicellular parasites and fungal-containing biofilms is also discussed, hinting at complex microbial relationships in various ecosystems. Understanding these interactions offers insights into disease mechanisms and antibiotic resistance dissemination, paving the way for innovative therapeutic strategies and ecosystem-level implications.
Collapse
Affiliation(s)
- Eva Zanditenas
- Department of Molecular Microbiology, Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Serge Ankri
- Department of Molecular Microbiology, Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
39
|
Khadraoui N, Essid R, Damergi B, Fares N, Gharbi D, Forero AM, Rodríguez J, Abid G, Kerekes EB, Limam F, Jiménez C, Tabbene O. Myrtus communis leaf compounds as novel inhibitors of quorum sensing-regulated virulence factors and biofilm formation: In vitro and in silico investigations. Biofilm 2024; 8:100205. [PMID: 38988475 PMCID: PMC11231753 DOI: 10.1016/j.bioflm.2024.100205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/18/2024] [Accepted: 06/05/2024] [Indexed: 07/12/2024] Open
Abstract
Antibiotic resistance of the Gram-negative bacterium Pseudomonas aeruginosa and its ability to form biofilm through the Quorum Sensing (QS) mechanism are important challenges in the control of infections caused by this pathogen. The extract of Myrtus communis (myrtle) showed strong anti-QS effect on C hromobacterium . violaceum 6267 by inhibiting 80 % of the production of violacein pigment at a sub-MIC concentration of 1/8 (31.25 μg/mL). In addition, the extract exhibited an inhibitory effect on virulence factors of P. aeruginosa PAO1 at half MIC (125 μg/mL), significantly reducing the formation of biofilms (72.02 %), the swarming activity (75 %), and the production of protease (61.83 %) and pyocyanin (97 %). The active fraction also downregulated the expression of selected regulatory genes involved in the biofilm formation and QS in the P. aeruginosa PAO1 strain. These genes included the autoinducer synthase genes (lasI and rhlI), the genes involved in the expression of their corresponding receptors (lasR and rhlR), and the pqsA genes. The analysis of the active fraction by HPLC/UV/MS and NMR allowed the identification of three phenolic compounds, 3,5-di-O-galloylquinic acid, myricetin 3-O-α-l-rhamnopyranoside (myricitrin), and myricetin 3-O-(2″-O-galloyl)-ß-d-galactopyranoside. In silico studies showed that 3,5-di-O-galloylquinic acid, with an affinity score of -9.20 kcal/mol, had the highest affinity to the active site of the CviR protein (3QP8), a QS receptor from C. violaceum. Additionally, myricetin 3-O-α-l-rhamnopyranoside (myricitrin) and myricetin 3-O-(2″-O-galloyl)-ß-d-galactopyranoside interact to a lesser extent with 3QP8. In conclusion, this study contributed significantly to the discovery of new QS inhibitors from M. communis leaves against resistant Gram-negative pathogens.
Collapse
Affiliation(s)
- Nadine Khadraoui
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
| | - Rym Essid
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
| | - Bilel Damergi
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
| | - Nadia Fares
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
| | - Dorra Gharbi
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
| | - Abel Mateo Forero
- CICA-Centro Interdisciplinar de Química e Bioloxía e Departamento de Química, Facultade de Ciencias. Universidade da Coruña, 15071, A Coruña, Spain
| | - Jaime Rodríguez
- CICA-Centro Interdisciplinar de Química e Bioloxía e Departamento de Química, Facultade de Ciencias. Universidade da Coruña, 15071, A Coruña, Spain
| | - Ghassen Abid
- Laboratory of Legumes and Sustainable Agrosystems, Centre de Biotechnology de Borj Cedria, BP-901, 2050, Hammam-Lif, Tunisia
| | - Erika-Beáta Kerekes
- Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Közép fasor 52, Hungary
| | - Ferid Limam
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
| | - Carlos Jiménez
- CICA-Centro Interdisciplinar de Química e Bioloxía e Departamento de Química, Facultade de Ciencias. Universidade da Coruña, 15071, A Coruña, Spain
| | - Olfa Tabbene
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP-901, Hammam-Lif 2050, Tunisia
| |
Collapse
|
40
|
Kumar N, Mattoo SS, Sanghvi S, Ellendula MP, Mahajan S, Planner C, Bednash JS, Khan M, Ganesan LP, Singh H, Lafuse WP, Wozniak DJ, Rajaram MVS. Pseudomonas aeruginosa- mediated cardiac dysfunction is driven by extracellular vesicles released during infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624948. [PMID: 39651123 PMCID: PMC11623511 DOI: 10.1101/2024.11.22.624948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Pseudomonas aeruginosa (P.a.) is a gram-negative, opportunistic bacterium abundantly present in the environment. Often P.a. infections cause severe pneumonia, if left untreated. Surprisingly, up to 30% of patients admitted to the hospital for community- acquired pneumonia develop adverse cardiovascular complications such as myocardial infarction, arrhythmia, left ventricular dysfunction, and heart failure. However, the underlying mechanism of infection-mediated cardiac dysfunction is not yet known. Recently, we demonstrated that P.a. infection of the lungs led to severe cardiac electrical abnormalities and left ventricular dysfunction with limited P.a. dissemination to the heart tissue. To understand the mechanism of cardiac dysfunction during P.a. infection, we utilized both in vitro and in vivo models. Our results revealed that inflammatory cytokines contribute but are not solely responsible for severe contractile dysfunction in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Instead, exposure of hiPSC-CMs with conditioned media from P.a. infected human monocyte-derived macrophages (hMDMs) was sufficient to cause severe contractile dysfunction and arrhythmia in hiPSC-CMs. Specifically, exosomes released from infected hMDMs and bacterial outer membrane vesicles (OMVs) are the major drivers of cardiomyocyte contractile dysfunction. By using LC-MS/MS, we identified bacterial proteins, including toxins that are packaged in the exosomes and OMVs, which are responsible for contractile dysfunction. Furthermore, we demonstrated that systemic delivery of bacterial OMVs to mice caused severe cardiac dysfunction, mimicking the natural bacterial infection. In summary, we conclude that OMVs released during infection enter circulation and drive cardiac dysfunction.
Collapse
|
41
|
Joshi S, Barman P, Maan M, Goyal H, Sharma S, Kumar R, Verma G, Saini A. Development of a two-dimensional peptide functionalized-reduced graphene oxide biomaterial for wound care applications. NANOSCALE 2024; 16:20986-21001. [PMID: 39463433 DOI: 10.1039/d4nr02233e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Increased incidences of antibiotic resistance have necessitated the development of novel wound disinfection strategies with minimal risk of resistance development. This study aimed at developing a biocompatible wound dressing biomaterial with the potential to treat acute and chronic wounds infected with multidrug-resistant Pseudomonas aeruginosa. A multifunctional antibacterial nanoconjugate was synthesized by covalently coupling a synthetically designed peptide (DP1, i.e., RFGRFLRKILRFLKK) with reduced graphene oxide (rGO). The conjugate displayed antibacterial and antibiofilm activities against multidrug-resistant Pseudomonas aeruginosa. In vitro studies demonstrated 94% hemocompatibility of the nanoconjugate even at concentrations as high as 512 μg mL-1. Cytotoxicity studies on 3T3-L1 cells showed 95% cell viability, signifying biocompatibility. Owing to these properties, the biomedical applicability of the nanoconjugate was assessed as an antibacterial wound dressing agent. rGO-DP1-loaded wound dressing exhibited enhanced reduction in bacterial bioburden (6 log 10 CFU) with potential for wound re-epithelization (77.3%) compared to the uncoated bandage. Moreover, an improvement in the material properties of the bandage was observed in terms of enhanced tensile strength and decreased elongation at break (%). Collectively, these findings suggest that rGO-DP1 is an effective biomaterial that, when loaded on wound dressings, has the potential to be used as a facile, sustainable and progressive agent for bacterial wound disinfection as well as healing.
Collapse
Affiliation(s)
- Shubhi Joshi
- Department of Biophysics, Panjab University, Chandigarh, U.T., 160014, India.
- Energy Research Centre, Panjab University, Chandigarh, U.T., 160014, India
| | - Panchali Barman
- Institute of Forensic Science & Criminology, Panjab University, Chandigarh, U.T., 160014, India
| | - Mayank Maan
- Department of Biophysics, Panjab University, Chandigarh, U.T., 160014, India.
| | - Hemant Goyal
- Department of Biophysics, Panjab University, Chandigarh, U.T., 160014, India.
| | - Sheetal Sharma
- Department of Biophysics, Panjab University, Chandigarh, U.T., 160014, India.
| | - Rajesh Kumar
- Department of Physics, Panjab University, Chandigarh, U.T., 160014, India
| | - Gaurav Verma
- Dr Shanti Swarup Bhatnagar University Institute of Chemical Engineering & Technology, Panjab University, Chandigarh, U.T., 160014, India
- Centre for Nanoscience & Nanotechnology (U.I.E.A.S.T), Panjab University, Chandigarh, U.T., 160014, India
| | - Avneet Saini
- Department of Biophysics, Panjab University, Chandigarh, U.T., 160014, India.
| |
Collapse
|
42
|
Hou J, Wu Q, Xiong R, Malakar PK, Zhu Y, Zhao Y, Zhang Z. A Standardized Mouse Model for Wound Infection with Pseudomonas aeruginosa. Int J Mol Sci 2024; 25:11773. [PMID: 39519323 PMCID: PMC11546588 DOI: 10.3390/ijms252111773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Pseudomonas aeruginosa is a highly drug-resistant pathogen known to impair wound healing and provoke inflammatory responses, potentially leading to immune dysregulation. This study aimed to systematically investigate the immune response mechanisms mediated by cytokines following P. aeruginosa infection through the development of a standardized wound model. Kunming mice were selected as experimental subjects and given 8 mm diameter lesions on their backs and inoculated with standard strains PAO1 and PA14. The key parameters assessed included changes in body weight, wound redness and swelling, bacterial dynamics, protein content in wound tissues, immune responses, and pathological alterations. The results demonstrated that pathogen invasion significantly inhibited wound healing, with healing rates in the infected groups (87.5 ± 6.3% and 77.1 ± 3.6%) being notably lower than those in the uninfected control group. P. aeruginosa persisted in the wounds for up to 12 days, with bacterial loads decreasing from 8 log to 2 log. Additionally, there was a marked reduction in the protein content of the wound tissue and an increase in the expression levels of inflammatory factors such as IL-1β and TNF-α. The thickness of granulation tissue and the number of neovessels were significantly lower compared to the uninfected control group. This study establishes a standardized paradigm for creating a mouse model of P. aeruginosa infection in wounds, emphasizing the importance of appropriate mouse strains, uniform wound preparation methods, and moderate inoculation doses for reliable and accurate experimental results. These elements will facilitate the assessment of changes across six key indicators post-infection, providing a foundational data set and technical support for future mechanistic investigations of P. aeruginosa infection and the development of targeted antimicrobial strategies.
Collapse
Affiliation(s)
- Jie Hou
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (J.H.); (Q.W.); (R.X.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Qian Wu
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (J.H.); (Q.W.); (R.X.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Rongrong Xiong
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (J.H.); (Q.W.); (R.X.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Pradeep K. Malakar
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (J.H.); (Q.W.); (R.X.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Yongheng Zhu
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (J.H.); (Q.W.); (R.X.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Yong Zhao
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (J.H.); (Q.W.); (R.X.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs, 999# Hu Cheng Huan Road, Shanghai 201306, China
- Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, 999# Hu Cheng Huan Road, Shanghai 201306, China
| | - Zhaohuan Zhang
- College of Food Science and Technology, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China; (J.H.); (Q.W.); (R.X.); (P.K.M.); (Y.Z.)
- International Research Center for Food and Health, Shanghai Ocean University, 999# Hu Cheng Huan Road, Shanghai 201306, China
- Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs, 999# Hu Cheng Huan Road, Shanghai 201306, China
- Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, 999# Hu Cheng Huan Road, Shanghai 201306, China
| |
Collapse
|
43
|
Hibbert TM, Whiteley M, Renshaw SA, Neill DR, Fothergill JL. Emerging strategies to target virulence in Pseudomonas aeruginosa respiratory infections. Crit Rev Microbiol 2024; 50:1037-1052. [PMID: 37999716 DOI: 10.1080/1040841x.2023.2285995] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that is responsible for infections in people living with chronic respiratory conditions, such as cystic fibrosis (CF) and non-CF bronchiectasis (NCFB). Traditionally, in people with chronic respiratory disorders, P. aeruginosa infection has been managed with a combination of inhaled and intravenous antibiotic therapies. However, due in part to the prolonged use of antibiotics in these people, the emergence of multi-drug resistant P. aeruginosa strains is a growing concern. The development of anti-virulence therapeutics may provide a new means of treating P. aeruginosa lung infections whilst also combatting the AMR crisis, as these agents are presumed to exert reduced pressure for the emergence of drug resistance as compared to antibiotics. However, the pipeline for developing anti-virulence therapeutics is poorly defined, and it is currently unclear as to whether in vivo and in vitro models effectively replicate the complex pulmonary environment sufficiently to enable development and testing of such therapies for future clinical use. Here, we discuss potential targets for P. aeruginosa anti-virulence therapeutics and the effectiveness of the current models used to study them. Focus is given to the difficulty of replicating the virulence gene expression patterns of P. aeruginosa in the CF and NCFB lung under laboratory conditions and to the challenges this poses for anti-virulence therapeutic development.
Collapse
Affiliation(s)
- Tegan M Hibbert
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
| | - Marvin Whiteley
- School of Biological Sciences, Georgia Institute of Technology, Centre for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Stephen A Renshaw
- The Bateson Centre and Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Daniel R Neill
- Division of Molecular Microbiology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Joanne L Fothergill
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
| |
Collapse
|
44
|
Ndikubwimana I, Gahamanyi N, Bwanakweli T, Uwayo HD, Habimana G, Rogo T. Case Report: Pan-Drug Resistant Pseudomonas aeruginosa from a Child with an Infected Burn Wound at the University Teaching Hospital of Kigali, Rwanda. Infect Drug Resist 2024; 17:4637-4642. [PMID: 39469095 PMCID: PMC11516630 DOI: 10.2147/idr.s486519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024] Open
Abstract
Background Pseudomonas aeruginosa is a significant cause of morbidity and mortality in intensive care units, and is prevalent in nosocomial infections and cystic fibrosis. The increasing rates of antimicrobial resistance (AMR) complicate the treatment of P. aeruginosa infections, especially because of the multidrug resistance (MDR), extensively drug-resistant (XDR), and pan-drug resistant (PDR) strains. Case Presentation We report the case of a 4-year-old male with severe burns covering 45% of his body surface who developed nosocomial PDR P. aeruginosa infection at the University Teaching Hospital of Kigali (CHUK) in Rwanda. A wound culture yielded a PDR P. aeruginosa isolate that was resistant to all the tested antimicrobials, with intermediate resistance to colistin. However, the patient improved with a combination of ceftazidime and amikacin following cessation of fever and successful skin grafting. The patient was discharged on day 95. Conclusion P. aeruginosa is a common hospital-acquired pathogen that is particularly challenging to treat, owing to its antimicrobial resistance profile and biofilm production. Antibiotic-resistant strains are a significant public health threat, especially in pediatric burn units. This case underscores the critical need to strengthen infection prevention and control measures together with robust antimicrobial stewardship programs. Molecular characterization of this PDR strain will yield further details regarding its virulence and genotyping.
Collapse
Affiliation(s)
- Innocent Ndikubwimana
- Pediatric Department, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
| | - Noel Gahamanyi
- National Reference Laboratory, Rwanda Biomedical Centre, Kigali, Rwanda
- Biology Department, College of Science and Technology, University of Rwanda, Kigali, Rwanda
| | | | | | - Gaspard Habimana
- Pediatric Department, Kigali University Teaching Hospital, Kigali, Rwanda
| | - Tanya Rogo
- Pediatric Infectious Diseases, Brown University Alpert Medical School, Providence, RI, USA
| |
Collapse
|
45
|
Rodrigues YC, Silva MJA, dos Reis HS, dos Santos PAS, Sardinha DM, Gouveia MIM, dos Santos CS, Marcon DJ, Aires CAM, Souza CDO, Quaresma AJPG, Lima LNGC, Brasiliense DM, Lima KVB. Molecular Epidemiology of Pseudomonas aeruginosa in Brazil: A Systematic Review and Meta-Analysis. Antibiotics (Basel) 2024; 13:983. [PMID: 39452249 PMCID: PMC11504043 DOI: 10.3390/antibiotics13100983] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Globally, Pseudomonas aeruginosa is a high-priority opportunistic pathogen which displays several intrinsic and acquired antimicrobial resistance (AMR) mechanisms, leading to challenging treatments and mortality of patients. Moreover, its wide virulence arsenal, particularly the type III secretion system (T3SS) exoU+ virulotype, plays a crucial role in pathogenicity and poor outcome of infections. In depth insights into the molecular epidemiology of P. aeruginosa, especially the prevalence of high-risk clones (HRCs), are crucial for the comprehension of virulence and AMR features and their dissemination among distinct strains. This study aims to evaluate the prevalence and distribution of HRCs and non-HRCs among Brazilian isolates of P. aeruginosa. METHODS A systematic review and meta-analysis were conducted on studies published between 2011 and 2023, focusing on the prevalence of P. aeruginosa clones determined by multilocus sequence typing (MLST) in Brazil. Data were extracted from retrospective cross-sectional and case-control studies, encompassing clinical and non-clinical samples. The analysis included calculating the prevalence rates of various sequence types (STs) and assessing the regional variability in the distribution of HRCs and non-HRCs. RESULTS A total of 872 samples were analyzed within all studies, of which 298 (34.17%) were MLST typed, identifying 78 unique STs. HRCs accounted for 48.90% of the MLST-typed isolates, with ST277 being the most prevalent (100/298-33.55%), followed by ST244 (29/298-9.73%), ST235 (13/298-4.36%), ST111 (2/298-0.67%), and ST357 (2/298-0.67%). Significant regional variability was observed, with the Southeast region showing a high prevalence of ST277, while the North region shows a high prevalence of MLST-typed samples and HRCs. CONCLUSIONS Finally, this systematic review and meta-analysis highlight the role of P. aeruginosa clones in critical issue of AMR in P. aeruginosa in Brazil and the need of integration of comprehensive data from individual studies.
Collapse
Affiliation(s)
- Yan Corrêa Rodrigues
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | - Marcos Jessé Abrahão Silva
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Herald Souza dos Reis
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
| | - Pabllo Antonny Silva dos Santos
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Daniele Melo Sardinha
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
| | - Maria Isabel Montoril Gouveia
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | - Carolynne Silva dos Santos
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Davi Josué Marcon
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Caio Augusto Martins Aires
- Department of Health Sciences (DCS), Federal Rural University of the Semi-Arid Region (UFERSA), Av. Francisco Mota, 572-Bairro Costa e Silva, Mossoró 59625-900, RN, Brazil;
| | - Cintya de Oliveira Souza
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
| | - Ana Judith Pires Garcia Quaresma
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Luana Nepomuceno Gondim Costa Lima
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Danielle Murici Brasiliense
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Karla Valéria Batista Lima
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| |
Collapse
|
46
|
Ren Y, You X, Zhu R, Li D, Wang C, He Z, Hu Y, Li Y, Liu X, Li Y. Mutation of Pseudomonas aeruginosa lasI/rhlI diminishes its cytotoxicity, oxidative stress, inflammation, and apoptosis on THP-1 macrophages. Microbiol Spectr 2024; 12:e0414623. [PMID: 39162513 PMCID: PMC11448257 DOI: 10.1128/spectrum.04146-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/27/2024] [Indexed: 08/21/2024] Open
Abstract
The management of Pseudomonas aeruginosa (P. aeruginosa) infections presents a substantial challenge to clinics and public health, emphasizing the urgent need for innovative strategies to address this issue. Quorum sensing (QS) is an intercellular communication mechanism that coordinates bacterial activities involved in various virulence mechanisms, such as acquiring host nutrients, facilitating biofilm formation, enhancing motility, secreting virulence factors, and evading host immune responses, all of which play a crucial role in the colonization and infection of P. aeruginosa. The LasI/R and RhlI/R sub-systems dominate in the QS system of P. aeruginosa. Macrophages play a pivotal role in the host's innate immune response to P. aeruginosa invasion, particularly through phagocytosis as the initial host defense mechanism. This study investigated the effects of P. aeruginosa's QS system on THP-1 macrophages. Mutants of PAO1 with lasI/rhlI deletion, as well as their corresponding complemented strains, were obtained, and significant downregulation of QS-related genes was observed in the mutants. Furthermore, the ΔlasI and ΔlasIΔrhlI mutants exhibited significantly attenuated virulence in terms of biofilm formation, extracellular polymeric substances synthesis, bacterial adhesion, motility, and virulence factors production. When infected with ΔlasI and ΔlasIΔrhlI mutants, THP-1 macrophages exhibited enhanced scavenging ability against the mutants and demonstrated resistance to cytotoxicity, oxidative stress, inflammatory response, and apoptosis induced by the culture supernatants of these mutant strains. These findings offer novel insights into the mechanisms underlying how the lasI/rhlI mutation attenuates cytotoxicity, oxidative stress, inflammation, and apoptosis in macrophages induced by P. aeruginosa.IMPORTANCEP. aeruginosa is classified as one of the ESKAPE pathogens and poses a global public health concern. The QS system of this versatile pathogen contributes to a broad spectrum of virulence, thereby constraining therapeutic options for serious infections. This study illustrated that the lasI/rhlI mutation of the QS system plays a prominent role in attenuating the virulence of P. aeruginosa by affecting bacterial adhesion, biofilm formation, extracellular polymeric substances synthesis, bacterial motility, and virulence factors' production. Notably, THP-1 macrophages infected with mutant strains exhibited increased phagocytic activity in eliminating intracellular bacteria and enhanced resistance to cytotoxicity, oxidative stress, inflammation, and apoptosis. These findings suggest that targeted intervention toward the QS system is anticipated to diminish the pathogenicity of P. aeruginosa to THP-1 macrophages.
Collapse
Affiliation(s)
- Yanying Ren
- Dazhou integrated Traditional Chinese Medicine & Western Medicine Hospital, Dazhou Second People's Hospital, Dazhou, China
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaojuan You
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Rui Zhu
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Dengzhou Li
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Chunxia Wang
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Zhiqiang He
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Yue Hu
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Yifan Li
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Xinwei Liu
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
| | - Yongwei Li
- Henan University of Chinese Medicine, Zhengzhou, China
- Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhenghzhou, China
- The Key Laboratory of Pathogenic Microbes &Antimicrobial Resistance Surveillance of Zhengzhou, Zhengzhou, China
- Henan Engineering Research Center for Identification of Pathogenic Microbes, Zhengzhou, China
- Henan Provincial Key Laboratory of Antibiotics-Resistant Bacterial Infection Prevention & Therapy with Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
47
|
Mazloumi Jourkouyeh E, Taslimi Eshkalak M, Faezi Ghasemi M, Zahmatkesh H, Rasti B, Zamani H. Diclofenac Sodium and Gentamicin Co-Encapsulated PLGA Nanoparticles: Targeting Extracellular Matrix Components to Combat Biofilm Formation in Pseudomonas aeruginosa PAO1. J CLUST SCI 2024; 35:2475-2488. [DOI: 10.1007/s10876-024-02675-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/23/2024] [Indexed: 01/05/2025]
|
48
|
Khan F. Multifaceted strategies for alleviating Pseudomonas aeruginosa infection by targeting protease activity: Natural and synthetic molecules. Int J Biol Macromol 2024; 278:134533. [PMID: 39116989 DOI: 10.1016/j.ijbiomac.2024.134533] [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: 06/10/2024] [Revised: 07/29/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024]
Abstract
Pseudomonas aeruginosa has become a top-priority pathogen in the health sector because it is ubiquitous, has high metabolic/genetic versatility, and is identified as an opportunistic pathogen. The production of numerous virulence factors by P. aeruginosa was reported to act individually or cooperatively to make them robots invasion, adherences, persistence, proliferation, and protection against host immune systems. P. aeruginosa produces various kinds of extracellular proteases such as alkaline protease, protease IV, elastase A, elastase B, large protease A, Pseudomonas small protease, P. aeruginosa aminopeptidase, and MucD. These proteases effectively allow the cells to invade and destroy host cells. Thus, inhibiting these protease activities has been recognized as a promising approach to controlling the infection caused by P. aeruginosa. The present review discussed in detail the characteristics of these proteases and their role in infection to the host system. The second part of the review discussed the recent updates on the multiple strategies for attenuating or inhibiting protease activity. These strategies include the application of natural and synthetic molecules, as well as metallic/polymeric nanomaterials. It has also been reported that a propeptide present in the middle domain of protease IV also attenuates the virulence properties and infection ability of P. aeruginosa.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
49
|
Vadakkan K, Sathishkumar K, Mapranathukaran VO, Ngangbam AK, Nongmaithem BD, Hemapriya J, Nair JB. Critical review on plant-derived quorum sensing signaling inhibitors in pseudomonas aeruginosa. Bioorg Chem 2024; 151:107649. [PMID: 39029321 DOI: 10.1016/j.bioorg.2024.107649] [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: 05/05/2024] [Revised: 06/21/2024] [Accepted: 07/14/2024] [Indexed: 07/21/2024]
Abstract
Pseudomonas aeruginosa, a biofilm-forming organism with complex quorum mechanisms (Las, Rhl, PQS, and IQS), poses an imminent danger to the healthcare sector and renders current treatment options for chemotherapy ineffectual. The pathogen's diverse pathogenicity, antibiotic resistance, and biofilms make it difficult to eradicate it effectively. Quorum sensing, a complex system reliant on cell density, controls P. aeruginosa's pathogenesis. Quorum-sensing genes are key components of P. aeruginosa's pathogenic arsenal, and their expression determines how severe the spread of infection becomes. Over the past ten years, there has been a noticeable increase in the quest for and development of new antimicrobial medications. Quorum sensing may be an effective treatment for infections triggered by bacteria. Introducing quorum-sensing inhibitors as an anti-virulent strategy might be an intriguing therapeutic method that can be effectively employed along with current medications. Amongst the several speculated processes, a unique anti-virulence strategy using anti-quorum sensing and antibiofilm medications for targeting pseudomonal infestations seems to be at the forefront. Due to their noteworthy quorum quenching capabilities, biologically active phytochemicals have become more well-known in the realm of science in this context. Recent research showed how different phytochemical quorum quenching actions affect P. aeruginosa's QS-dependent pathogenicity. This review focuses on the most current data supporting the implementation of plant bio-actives to treat P.aeruginosa-associated diseases, as well as the benefits and future recommendationsof employing them in anti-virulence therapies as a supplementary drug development approach towards conventional antibiotic approaches.
Collapse
Affiliation(s)
- Kayeen Vadakkan
- Department of Biotechnology, St. Mary's College (Autonomous), Thrissur, Kerala 680020, India; Manipur International University, Imphal, Manipur 795140, India.
| | - Kuppusamy Sathishkumar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Thandalam, Chennai, Tamil Nadu 602105, India
| | | | | | | | - Janarthanam Hemapriya
- Department of Microbiology, DKM College for Women, Vellore, Tamil Nadu 632001, India
| | - Jyotsna B Nair
- Department of Biotechnology, JDT Islam College of Arts and Science, Vellimadukunnu, Kozhikode, Kerala 673012, India
| |
Collapse
|
50
|
Kumari K, Dey J, Mahapatra SR, Ma Y, Sharma PK, Misra N, Singh RP. Protein profiling and immunoinformatic analysis of the secretome of a metal-resistant environmental isolate Pseudomonas aeruginosa S-8. Folia Microbiol (Praha) 2024; 69:1095-1122. [PMID: 38457114 DOI: 10.1007/s12223-024-01152-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 02/03/2024] [Indexed: 03/09/2024]
Abstract
The bacterial secretome represents a comprehensive catalog of proteins released extracellularly that have multiple important roles in virulence and intercellular communication. This study aimed to characterize the secretome of an environmental isolate Pseudomonas aeruginosa S-8 by analyzing trypsin-digested culture supernatant proteins using nano-LC-MS/MS tool. Using a combined approach of bioinformatics and mass spectrometry, 1088 proteins in the secretome were analyzed by PREDLIPO, SecretomeP 2.0, SignalP 4.1, and PSORTb tool for their subcellular localization and further categorization of secretome proteins according to signal peptides. Using the gene ontology tool, secretome proteins were categorized into different functional categories. KEGG pathway analysis identified the secreted proteins into different metabolic functional pathways. Moreover, our LC-MS/MS data revealed the secretion of various CAZymes into the extracellular milieu, which suggests its strong biotechnological applications to breakdown complex carbohydrate polymers. The identified immunodominant epitopes from the secretome of P. aeruginosa showed the characteristic of being non-allergenic, highly antigenic, nontoxic, and having a low risk of triggering autoimmune responses, which highlights their potential as successful vaccine targets. Overall, the identification of secreted proteins of P. aeruginosa could be important for both diagnostic purposes and the development of an effective candidate vaccine.
Collapse
Affiliation(s)
- Kiran Kumari
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Ranchi, 835215, India
| | - Jyotirmayee Dey
- School of Biotechnology, Deemed to Be University, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, 751024, India
| | - Soumya Ranjan Mahapatra
- School of Biotechnology, Deemed to Be University, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, 751024, India
| | - Ying Ma
- College of Resources and Environment, Southwest University, Chongqing, China
| | - Parva Kumar Sharma
- Department of Plant Sciences and Landscape Architecture, University of Maryland, College Park, MD, 20742, USA
| | - Namrata Misra
- School of Biotechnology, Deemed to Be University, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, 751024, India
| | - Rajnish Prakash Singh
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, 201309, India.
| |
Collapse
|