201
|
Song Y, Wu X, Li Z, Ma QQ, Bao R. Molecular mechanism of siderophore regulation by the Pseudomonas aeruginosa BfmRS two-component system in response to osmotic stress. Commun Biol 2024; 7:295. [PMID: 38461208 PMCID: PMC10924945 DOI: 10.1038/s42003-024-05995-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: 11/13/2023] [Accepted: 02/29/2024] [Indexed: 03/11/2024] Open
Abstract
Pseudomonas aeruginosa, a common nosocomial pathogen, relies on siderophores to acquire iron, crucial for its survival in various environments and during host infections. However, understanding the molecular mechanisms of siderophore regulation remains incomplete. In this study, we found that the BfmRS two-component system, previously associated with biofilm formation and quorum sensing, is essential for siderophore regulation under high osmolality stress. Activated BfmR directly bound to the promoter regions of pvd, fpv, and femARI gene clusters, thereby activating their transcription and promoting siderophore production. Subsequent proteomic and phenotypic analyses confirmed that deletion of BfmRS reduces siderophore-related proteins and impairs bacterial survival in iron-deficient conditions. Furthermore, phylogenetic analysis demonstrated the high conservation of the BfmRS system across Pseudomonas species, functional evidences also indicated that BfmR homologues from Pseudomonas putida KT2440 and Pseudomonas sp. MRSN12121 could bind to the promoter regions of key siderophore genes and osmolality-mediated increases in siderophore production were observed. This work illuminates a novel signaling pathway for siderophore regulation and enhances our understanding of siderophore-mediated bacterial interactions and community establishment.
Collapse
Affiliation(s)
- Yingjie Song
- College of Life Science, Sichuan Normal University, Chengdu, 610101, China
| | - Xiyu Wu
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610213, China
| | - Ze Li
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qin Qin Ma
- College of Life Science, Sichuan Normal University, Chengdu, 610101, China
| | - Rui Bao
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
202
|
Zhao C, Wang C, Zhou Y, Hu T, Zhang Y, Lv X, Li J, Zhou Y. Discovery of Potential Anti-Microbial Molecules and Spectrum Correlation Effect of Ardisia crenata Sims via High-Performance Liquid Chromatography Fingerprints and Molecular Docking. Molecules 2024; 29:1178. [PMID: 38474690 DOI: 10.3390/molecules29051178] [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: 01/14/2024] [Revised: 02/22/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Ardisia crenata Sims, an important ethnic medicine, is recorded in the Chinese Pharmacopoeia for treating laryngeal diseases and upper respiratory tract infections. This study aimed to evaluate the antimicrobial effect of extracts and potential antimicrobial compounds of A. crenata Sims. It was found that the roots of A. crenata Sims have a potential inhibitory effect on Candida albicans and Aspergillus flavus, with MICs of 1.56 mg/mL and 0.39 mg/mL, and the leaves of A. crenata Sims have a potential inhibitory effect on Pseudomonas aeruginosa and Staphylococcus aureus, with MICs of 3.12 mg/mL and 6.77 mg/mL, respectively. Meanwhile, five compounds including one catechin and four bergenins were obtained from roots. These components were identified on the fingerprint spectrum, representing chromatographic peaks 16, 21, 22, 23, and 25, respectively. Among these, 11-β-d-glucopyranosyl-bergenin and (-)-gallocatechin showed potential inhibition for Staphylococcus aureus and Pseudomonas aeruginosa with MIC of 0.26 and 0.33 mg/mL, respectively. The roots, stems, and leaves of A. crenata Sims are very similar in chemical composition, with large differences in content. Principal component analysis (PCA) and Hierarchical cluster analysis (HCA) showed that 16 batches of A. crenata Sims could be divided into four main production areas: Guizhou, Jiangsu, Guangxi, and Jiangxi. Furthermore, molecular docking results showed that 11-β-d-glucopyranosyl-bergenin had a better affinity for Casein lytic proteinase P (ClpP), and (-)-gallocatechin possessed a strong affinity for LasA hydrolysis protease and LasB elastase. These findings suggest catechin and bergenins from A. crenata Sims can be used as antimicrobial activity molecules.
Collapse
Affiliation(s)
- Chunli Zhao
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Changbin Wang
- Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Yongqiang Zhou
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Tao Hu
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Yan Zhang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Xiang Lv
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Jiaxin Li
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Ying Zhou
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| |
Collapse
|
203
|
Hong Q, Chang RYK, Assafiri O, Morales S, Chan HK. Optimizing in vitro phage-ciprofloxacin combination formulation for respiratory therapy of multi-drug resistant Pseudomonas aeruginosa infections. Int J Pharm 2024; 652:123853. [PMID: 38280500 DOI: 10.1016/j.ijpharm.2024.123853] [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: 11/26/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 01/29/2024]
Abstract
Respiratory infection caused by multi-drug resistant (MDR) Pseudomonas aeruginosa is challenging to treat. In this study, we investigate the optimal dose of anti-pseudomonas phage PEV31 (103, 105, and 108 PFU/mL) combined with ciprofloxacin (ranging from 1/8× MIC to 8× MIC) to treat the MDR P. aeruginosa strain FADD1-PA001 using time-kill studies. We determined the impact of phage growth kinetics in the presence of ciprofloxacin through one-step growth analysis. Single treatments with either phage PEV31 or ciprofloxacin (except at 8× MIC) showed limited bactericidal efficiency, with bacterial regrowth observed at 48 h. The most effective treatments were PEV31 at multiplicity of infection (MOI) of 0.1 and 100 combined with ciprofloxacin at concentrations above 1× MIC, resulting in a >4 log10 reduction in bacterial counts. While the burst size of phage PEV31 was decreased with increasing ciprofloxacin concentration, robust antimicrobial effects were still maintained in the combination treatment. Aerosol samples collected from vibrating mesh nebulization of the combination formulation at phage MOI of 100 with 2× MIC effectively inhibited bacterial density. In summary, our combination treatments eradicated in vitro bacterial growth and sustained antimicrobial effects for 48 h. These results indicated the potential application of nebulization-based strategies for the combination treatment against MDR lung infections.
Collapse
Affiliation(s)
- Qixuan Hong
- Advanced Drug Delivery Group, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Rachel Yoon Kyung Chang
- Advanced Drug Delivery Group, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Omar Assafiri
- Advanced Drug Delivery Group, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | | | - Hak-Kim Chan
- Advanced Drug Delivery Group, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
204
|
Shi Q, Zeng S, Yu R, Li M, Shen C, Zhang X, Zhao C, Zeng J, Huang B, Pu J, Chen C. The small RNA PrrH aggravates Pseudomonas aeruginosa-induced acute lung injury by regulating the type III secretion system activator ExsA. Microbiol Spectr 2024; 12:e0062623. [PMID: 38289930 PMCID: PMC10913731 DOI: 10.1128/spectrum.00626-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 01/09/2024] [Indexed: 02/01/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that causes acute and chronic infections in immunocompromised individuals. Small regulatory RNAs (sRNAs) regulate multiple bacterial adaptations to environmental changes, especially virulence. Our previous study showed that sRNA PrrH negatively regulates the expression of a number of virulence factors, such as pyocyanin, rhamnolipid, biofilm, and elastase in the P. aeruginosa strain PAO1. However, previous studies have shown that the prrH-deficient mutant attenuates virulence in an acute murine lung infection model. All ΔprrH-infected mice survived the entire 28-day course of the experiment, whereas all mice inoculated with the wild-type or the complemented mutant succumbed to lung infection within 4 days of injection, but the specific mechanism is unclear. Herein, we explored how PrrH mediates severe lung injury by regulating the expression of virulence factors. In vivo mouse and in vitro cellular assays demonstrated that PrrH enhanced the pathogenicity of PAO1, causing severe lung injury. Mechanistically, PrrH binds to the coding sequence region of the mRNA of exsA, which encodes the type III secretion system master regulatory protein. We further demonstrated that PrrH mediates a severe inflammatory response and exacerbates the apoptosis of A549 cells. Overall, our results revealed that PrrH positively regulates ExsA, enhances the pathogenicity of P. aeruginosa, and causes severe lung injury. IMPORTANCE Pseudomonas aeruginosa is a Gram-negative bacterium and the leading cause of nosocomial pneumonia. The pathogenicity of P. aeruginosa is due to the secretion of many virulence factors. Small regulatory RNAs (sRNAs) regulate various bacterial adaptations, especially virulence. Therefore, understanding the mechanism by which sRNAs regulate virulence is necessary for understanding the pathogenicity of P. aeruginosa and the treatment of the related disease. In this study, we demonstrated that PrrH enhances the pathogenicity of P. aeruginosa by binding to the coding sequence regions of the ExsA, the master regulatory protein of type III secretion system, causing severe lung injury and exacerbating the inflammatory response and apoptosis. These findings revealed that PrrH is a crucial molecule that positively regulates ExsA. Type III-positive strains are often associated with a high mortality rate in P. aeruginosa infections in clinical practice. Therefore, this discovery may provide a new target for treating P. aeruginosa infections, especially type III-positive strains.
Collapse
Affiliation(s)
- Qixuan Shi
- Department of Laboratory Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shenghe Zeng
- Department of Laboratory Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruiqi Yu
- Department of Laboratory Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mo Li
- Department of Laboratory Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cong Shen
- Department of Laboratory Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuan Zhang
- Department of Laboratory Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chanjing Zhao
- Department of Laboratory Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianming Zeng
- Department of Laboratory Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bin Huang
- Department of Laboratory Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jieying Pu
- Department of Laboratory Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cha Chen
- Department of Laboratory Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
205
|
Bai Y, Gong YE, Shen F, Li H, Cheng Y, Guo J, Liu G, Ji AF. Molecular epidemiological characteristics of carbapenem-resistant Pseudomonas aeruginosa clinical isolates in southeast Shanxi, China. J Glob Antimicrob Resist 2024; 36:301-306. [PMID: 38272212 DOI: 10.1016/j.jgar.2023.12.029] [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: 10/14/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/27/2024] Open
Abstract
OBJECTIVES Infection by carbapenem-resistant Pseudomonas aeruginosa (CRPA) is a serious clinical problem worldwide. However, the molecular epidemiology of the clinical isolates varies depending on the region. This study was conducted to analyse the resistance phenotype and clarify the genetic and epidemiological properties of CRPA clinical isolates from southeast Shanxi, China. METHODS Fifty-seven isolates of CRPA were collected from a hospital in this region. These isolates were reidentified by MALDI-TOF and subjected to whole-genome sequencing by next-generation sequencing. Phylogenetic trees were constructed based on single nucleotide polymorphisms (SNPs), after which multilocus sequence typing (MLST) was performed and antimicrobial resistance genes were identified. RESULTS All the 57 CRPA isolates carried at least one kind of gene encoding carbapenemase, such as blaIMP-1, blaIMP-10, blaOXA-10, blaOXA-395, blaOXA-396, blaOXA-485, blaOXA-486, blaOXA-488, blaOXA-494, and blaOXA-50. The isolates harboured AIM-1, CMY-51, mecD, and NmcR genes and carried one kind of Pseudomonas-derived cephalosporinase (PDC) β-lactamase-encoding gene, such as blaPCD-1 to blaPCD-3, blaPCD-5, or blaPCD-7 to blaPCD-10. Two isolates were found to harbour the aminoglycoside-modifying enzyme genes aadA1 and aadA7; however, no isolates were found to harbour genes encoding 16S rRNA methylase or quinolone resistance-related genes. These CRPA isolates belonged to various sequence types (STs), two of which, namely, ST235 and ST277, were high-risk types. CONCLUSIONS Our findings indicate that CRPA isolates carrying resistance genes with unique regional characteristics are spreading in this region, with a high diversity of STs, especially in high-risk clones. These findings highlight the necessity for further measures to prevent CRPA spread in Shanxi.
Collapse
Affiliation(s)
- Yang Bai
- Heping Hospital, Changzhi Medical College, Changzhi, Shanxi, China; Department of Medical Laboratory, Changzhi Medical College, Changzhi, Shanxi, China
| | - Yan-E Gong
- Heping Hospital, Changzhi Medical College, Changzhi, Shanxi, China; Department of Medical Laboratory, Changzhi Medical College, Changzhi, Shanxi, China
| | - Fangfang Shen
- Heping Hospital, Changzhi Medical College, Changzhi, Shanxi, China; Department of Medical Laboratory, Changzhi Medical College, Changzhi, Shanxi, China
| | - Hui Li
- Heping Hospital, Changzhi Medical College, Changzhi, Shanxi, China; Department of Medical Laboratory, Changzhi Medical College, Changzhi, Shanxi, China
| | - Yan Cheng
- Heping Hospital, Changzhi Medical College, Changzhi, Shanxi, China; Department of Medical Laboratory, Changzhi Medical College, Changzhi, Shanxi, China
| | - Jinying Guo
- Heping Hospital, Changzhi Medical College, Changzhi, Shanxi, China; Department of Medical Laboratory, Changzhi Medical College, Changzhi, Shanxi, China
| | - Guangming Liu
- Heping Hospital, Changzhi Medical College, Changzhi, Shanxi, China; Department of Medical Laboratory, Changzhi Medical College, Changzhi, Shanxi, China
| | - Ai-Fang Ji
- Heping Hospital, Changzhi Medical College, Changzhi, Shanxi, China; Department of Medical Laboratory, Changzhi Medical College, Changzhi, Shanxi, China.
| |
Collapse
|
206
|
Shrestha PM, Kattel HP, Sharma S, Bista P, Basnet BK, Ghimire P, Rijal KR. Metallo-β-lactamase-producing Pseudomonas aeruginosa Isolates from Two Tertiary Care Centres in a District of Nepal: A Descriptive Cross-sectional Study. JNMA J Nepal Med Assoc 2024; 62:202-206. [PMID: 39356784 PMCID: PMC10924479 DOI: 10.31729/jnma.8498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Indexed: 10/04/2024] Open
Abstract
INTRODUCTION Pseudomonas aeruginosa isolates producing metallo-β-lactamase have caused nosocomial outbreaks, severe infections, and ineffective carbapenem therapy worldwide since 1991. Due to their prevalence, hospital infection control techniques are difficult. This study aimed to find out the prevalence of metallo-β-lactamase among P. aeruginosa isolates from two tertiary care hospitals in Kathmandu. METHODS A descriptive cross-sectional study was conducted at the Department of Microbiology and Department of Pathology of two tertiary care centres in Kathmandu from 7 December 2021 to 6 April 2023, after receiving ethical approval from the Ethical Review Board. Isolated strains were identified and tested for antibiotic susceptibility by modified Kirby-Bauer Methods. Metallo-β-lactamase presence was confirmed using an imipenem-imipenem/ ethylenediaminetetraacetic acid disc. A convenience sampling method was used. The point estimate was calculated at 95% Confidence Interval. RESULTS Among 255, Pseudomanas aeruginosa isolates, the distribution of metallo-β-lactamase-producing Pseudomanas aeruginosa was 103 (40.39%) (34.32-46.69 at 95% Confidence Interval). Multidrug resistance categories included multidrug resistance 74 (71.80%), extensively drug resistance 32 (31.10%), P. aeruginosa difficult-to-treat 16 (15.53%) and carbapenem-resistant P. aeruginosa was determined to be 82 (79.60%). CONCLUSIONS The study found a high prevalence of metallo-β-lactamase-producing Pseudomanas aeruginosa isolates, requiring early identification, infection control measures, and an all-inclusive antimicrobial therapy protocol to reduce their spread in medical settings.
Collapse
Affiliation(s)
- Pushpa Man Shrestha
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Hari Prasad Kattel
- Department of Microbiology, Tribhuvan University Teaching Hospital, Maharajgunj, Kathmandu, Nepal
| | - Sangita Sharma
- Department of Microbiology, Maharajgunj Medical Campus, Maharajgunj, Kathmandu, Nepal
| | - Pratibha Bista
- Department of Pathology, Bir Hospital, Mahabaudha, Kathmandu, Nepal
| | | | - Prakash Ghimire
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Komal Raj Rijal
- Central Department of Microbiology, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| |
Collapse
|
207
|
Navarro-López DE, Perfecto-Avalos Y, Zavala A, de Luna MA, Sanchez-Martinez A, Ceballos-Sanchez O, Tiwari N, López-Mena ER, Sanchez-Ante G. Unraveling the Complex Interactions: Machine Learning Approaches to Predict Bacterial Survival against ZnO and Lanthanum-Doped ZnO Nanoparticles. Antibiotics (Basel) 2024; 13:220. [PMID: 38534655 DOI: 10.3390/antibiotics13030220] [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: 01/23/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
The rise in antibiotic-resistant bacteria is a global health challenge. Due to their unique properties, metal oxide nanoparticles show promise in addressing this issue. However, optimizing these properties requires a deep understanding of complex interactions. This study incorporated data-driven machine learning to predict bacterial survival against lanthanum-doped ZnO nanoparticles. The effect of incorporation of lanthanum ions on ZnO was analyzed. Even with high lanthanum concentration, no significant variations in structural, morphological, and optical properties were observed. The antibacterial activity of La-doped ZnO nanoparticles against Gram-positive and Gram-negative bacteria was qualitatively and quantitatively evaluated. Nanoparticles induce 60%, 95%, and 55% bacterial death against Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus aureus, respectively. Algorithms such as Multilayer Perceptron, K-Nearest Neighbors, Gradient Boosting, and Extremely Random Trees were used to predict the bacterial survival percentage. Extremely Random Trees performed the best among these models with 95.08% accuracy. A feature relevance analysis extracted the most significant attributes to predict the bacterial survival percentage. Lanthanum content and particle size were irrelevant, despite what can be assumed. This approach offers a promising avenue for developing effective and tailored strategies to reduce the time and cost of developing antimicrobial nanoparticles.
Collapse
Affiliation(s)
- Diego E Navarro-López
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Gral Ramón Corona No. 2514, Colonia Nuevo México, Zapopan 45121, Jalisco, Mexico
| | - Yocanxóchitl Perfecto-Avalos
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Gral Ramón Corona No. 2514, Colonia Nuevo México, Zapopan 45121, Jalisco, Mexico
| | - Araceli Zavala
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Gral Ramón Corona No. 2514, Colonia Nuevo México, Zapopan 45121, Jalisco, Mexico
| | - Marco A de Luna
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Gral Ramón Corona No. 2514, Colonia Nuevo México, Zapopan 45121, Jalisco, Mexico
| | - Araceli Sanchez-Martinez
- Departamento de Ingenieria de Proyectos, Centro Universitario de Ciencias Exactas e Ingenierias (CUCEI), Universidad de Guadalajara, Av. José Guadalupe Zuno # 48, Industrial Los Belenes, Zapopan 45157, Jalisco, Mexico
| | - Oscar Ceballos-Sanchez
- Departamento de Ingenieria de Proyectos, Centro Universitario de Ciencias Exactas e Ingenierias (CUCEI), Universidad de Guadalajara, Av. José Guadalupe Zuno # 48, Industrial Los Belenes, Zapopan 45157, Jalisco, Mexico
| | - Naveen Tiwari
- Center for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela, Rúa Jenaro de La Fuente S/N, 15782 Santiago de Compostela, Spain
| | - Edgar R López-Mena
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Gral Ramón Corona No. 2514, Colonia Nuevo México, Zapopan 45121, Jalisco, Mexico
| | - Gildardo Sanchez-Ante
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Gral Ramón Corona No. 2514, Colonia Nuevo México, Zapopan 45121, Jalisco, Mexico
| |
Collapse
|
208
|
Crippen TL, Kim D, Poole TL, Swiger SL, Anderson RC. The bacterial and archaeal communities of flies, manure, lagoons, and troughs at a working dairy. Front Microbiol 2024; 14:1327841. [PMID: 38449879 PMCID: PMC10915237 DOI: 10.3389/fmicb.2023.1327841] [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: 10/25/2023] [Accepted: 12/19/2023] [Indexed: 03/08/2024] Open
Abstract
Background Fundamental investigations into the location, load, and persistence of microbes, whether beneficial or detrimental, are scarce. Many questions about the retention and survival of microbes on various surfaces, as well as the load necessary for spread, exist. To answer these questions, we must know more about where to find various microbes and in what concentrations, the composition of the microbial communities, and the extent of dissemination between various elements. This study investigated the diversity, composition, and relative abundance of the communities associated with manure, lagoons, troughs, house flies, and stable flies present at a dairy, implementing two different free-stall management systems: flow-through and cross-vent. Shotgun metagenomics at the community level was used to compare the microbiomes within the dairy, allowing confident interpretation at the species level. Results The results showed that there were significant difference in microbial composition between not only each of the dairy elements but also management styles. The primary exceptions were the microbiomes of the house fly and the stable fly. Their compositions heavily overlapped with one another, but interestingly, not with the other components sampled. Additionally, both species of flies carried more pathogens than the other elements of the dairy, indicating that they may not share these organisms with the other components, or that the environments offered by the other components are unsatisfactory for the survival of some pathogens.. Conclusion The lack of overlapping pathogen profiles suggests a lack of transfer from flies to other dairy elements. Dairy health data, showing a low incidence of disease, suggests minimal sharing of bacteria by the flies at a level required for infection, given the health program of this dairy. While flies did carry a multitude of pathogenic bacteria, the mere presence of the bacteria associated with the flies did not necessarily translate into high risk leading to morbidity and mortality at this dairy. Thus, using flies as the sole sentinel of dairy health may not be appropriate for all bacterial pathogens or dairies.
Collapse
Affiliation(s)
- Tawni L. Crippen
- Southern Plains Agricultural Research Center, Agricultural Research Service, US Department of Agriculture, College Station, TX, United States
| | - Dongmin Kim
- Department of Entomology, Texas A & M University, College Station, TX, United States
| | - Toni L. Poole
- Southern Plains Agricultural Research Center, Agricultural Research Service, US Department of Agriculture, College Station, TX, United States
| | - Sonja L. Swiger
- Entomology Extension, Texas AgriLife, Texas A & M University, College Station, TX, United States
| | - Robin C. Anderson
- Southern Plains Agricultural Research Center, Agricultural Research Service, US Department of Agriculture, College Station, TX, United States
| |
Collapse
|
209
|
Singothu S, Bhandari V. Computational assessment of marine natural products as LasR inhibitors for attenuating quorum sensing in Pseudomonas aeruginosa. J Biomol Struct Dyn 2024:1-15. [PMID: 38379380 DOI: 10.1080/07391102.2024.2319110] [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: 10/11/2023] [Accepted: 02/11/2024] [Indexed: 02/22/2024]
Abstract
The Quorum Sensing (QS) system in bacteria has become a focal point for researchers aiming to develop novel antimicrobials to combat multidrug-resistant bacteria. Pseudomonas aeruginosa, an opportunistic Gram-negative bacterium, has developed resistance against a variety of antimicrobial agents, making it a formidable pathogen responsible for nosocomial infections. QS system mainly controls the expression of genes responsible for biofilm formation and virulence of bacteria. Within the QS system of P. aeruginosa, the transcription activator LasR plays a pivotal role and is an appealing target for the development of antimicrobial agents. In this study, we employed molecular docking and molecular dynamics simulations to identify potential inhibitors of LasR by screening marine natural products (MNPs) from the CMNPD database. We identified ten MNPs with excellent docking scores (less than -11.7 kcal/mol) against LasR, surpassing the binding energy of the co-crystal 3-oxo-C12-HSL (-8.594 kcal/mol) and the reference compound cladodionen (-6.71 kcal/mol). Furthermore, we selected five of these MNPs with the highest MM/GBSA binding energies for extensive 100 ns molecular simulations to assess their stability. The molecular dynamics simulations indicated three MNPs, namely CMNPD10886, CMNPD20987, and CMNPD20960, maintained high stability throughout the 100 ns simulation period, as evidenced by their root mean square deviation, root mean square fluctuation, radius of gyration, and hydrogen bond interactions within the ligand-protein complex analysis. Furthermore, essential dynamics (PCA and DCCM) were performed to analyse the correlated motion of amino acids. These findings suggest that these compounds hold potential as inhibitors of LasR, offering promising prospects for the development of treatments against infections.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Siva Singothu
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Vasundhra Bhandari
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| |
Collapse
|
210
|
Yu JS, Kim M, Cho IH, Sim YM, Hwang YS. Evidence Supporting Oral Hygiene Management by Owners through a Genetic Analysis of Dental Plaque Bacteria in Dogs. Vet Sci 2024; 11:96. [PMID: 38393114 PMCID: PMC10893504 DOI: 10.3390/vetsci11020096] [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: 01/16/2024] [Revised: 02/07/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
With the increase in the number of households raising dogs and the reports of human-to-dog transmission of oral bacteria, concerns about dogs' oral health and the need for oral hygiene management are increasing. In this study, the owners' perceptions about their dogs' oral health and the frequency of oral hygiene were determined along with the analysis of dog dental plaque bacteria through metagenomic amplicon sequencing so as to support the need for oral hygiene management for dogs. Although the perception of 63.2% of the owners about their dogs' oral health was consistent with the veterinarian's diagnosis, the owners' oral hygiene practices regarding their dogs were very poor. The calculi index (CI) and gingiva index (GI) were lower in dogs who had their teeth brushed more than once a week (57.89%) than in dogs brushed less than once a month (42.10%); however, the difference was nonsignificant (CI: p = 0.479, GI: p = 0.840). Genomic DNA was extracted from dental plaque bacteria removed during dog teeth scaling, and metagenomic amplicons were sequenced. The 16S amplicons of 73 species were identified from among the plaque bacteria of the dogs. These amplicons were of oral disease-causing bacteria in humans and dogs. The 16S amplicon of Streptococcus mutans matched that of the human S. mutans, with type c identified as the main serotype. This result suggests that human oral bacteria can be transmitted to dogs. Therefore, considering the high frequency of contact between dogs and humans because of communal living and the current poor oral health of dogs, owners must improve the oral hygiene management of their dogs.
Collapse
Affiliation(s)
- Jeong suk Yu
- Yeah Dental Animal Clinic, Seolleung-ro 126-6, Gangnam-Gu, Seoul 06092, Republic of Korea
| | - Minhee Kim
- Department of Physical Therapy, College of Health Science, Eulji University, Seongnam 13135, Republic of Korea
| | - Il-Hoon Cho
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam 13135, Republic of Korea
| | - Yu-Min Sim
- Department of Dental Hygiene, College of Health Science, Eulji University, Seongnam 13135, Republic of Korea
| | - Young Sun Hwang
- Department of Dental Hygiene, College of Health Science, Eulji University, Seongnam 13135, Republic of Korea
| |
Collapse
|
211
|
Wen F, Wu Y, Yuan Y, Yang X, Ran Q, Gan X, Guo Y, Wang X, Chu Y, Zhao K. Discovery of psoralen as a quorum sensing inhibitor suppresses Pseudomonas aeruginosa virulence. Appl Microbiol Biotechnol 2024; 108:222. [PMID: 38372782 PMCID: PMC10876730 DOI: 10.1007/s00253-024-13067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/10/2024] [Accepted: 02/11/2024] [Indexed: 02/20/2024]
Abstract
Pseudomonas aeruginosa is a common opportunistic pathogen with growing resistance and presents heightened treatment challenges. Quorum sensing (QS) is a cell-to-cell communication system that contributes to the production of a variety of virulence factors and is also related to biofilm formation of P. aeruginosa. Compared to traditional antibiotics which kill bacteria directly, the anti-virulence strategy by targeting QS is a promising strategy for combating pseudomonal infections. In this study, the QS inhibition potential of the compounds derived from the Traditional Chinese Medicines was evaluated by using in silico, in vitro, and in vivo analyses. The results showed that psoralen, a natural furocoumarin compound derived from Psoralea corylifolia L., was capable of simultaneously inhibiting the three main QS regulators, LasR, RhlR, and PqsR of P. aeruginosa. Psoralen had no bactericidal activity but could widely inhibit the production of extracellular proteases, pyocyanin, and biofilm, and the cell motilities of the model and clinical P. aeruginosa strains. RNA-sequencing and quantitative PCR analyses further demonstrated that a majority of QS-activated genes in P. aeruginosa were suppressed by psoralen. The supplementation of psoralen could protect Caenorhabditis elegans from P. aeruginosa challenge, especially for the hypervirulent strain PA14. Moreover, psoralen showed synergistic antibacterial effects with polymyxin B, levofloxacin, and kanamycin. In conclusions, this study identifies the anti-QS and antibiofilm effects of psoralen against P. aeruginosa strains and sheds light on the discovery of anti-pseudomonal drugs among Traditional Chinese Medicines. KEY POINTS: • Psoralen derived from Psoralea corylifolia L. inhibits the virulence-related phenotypes of P. aeruginosa. • Psoralen simultaneously targets the three core regulators of P. aeruginosa QS system and inhibits the expression of a large part of downstream genes. • Psoralen protects C. elegans from P. aeruginosa challenge and enhances the susceptibility of P. aeruginosa to antibiotics.
Collapse
Affiliation(s)
- Fulong Wen
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China
| | - Yi Wu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China
| | - Yang Yuan
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China
| | - Xiting Yang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China
| | - Qiman Ran
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China
| | - Xiongyao Gan
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China
| | - Yidong Guo
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China
| | - Xinrong Wang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China
| | - Yiwen Chu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China.
| | - Kelei Zhao
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China.
| |
Collapse
|
212
|
Abdelhamid AG, Yousef AE. Untargeted metabolomics unveiled the role of butanoate metabolism in the development of Pseudomonas aeruginosa hypoxic biofilm. Front Cell Infect Microbiol 2024; 14:1346813. [PMID: 38435305 PMCID: PMC10904581 DOI: 10.3389/fcimb.2024.1346813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024] Open
Abstract
Pseudomonas aeruginosa is a versatile opportunistic pathogen which causes a variety of acute and chronic human infections, some of which are associated with the biofilm phenotype of the pathogen. We hypothesize that defining the intracellular metabolome of biofilm cells, compared to that of planktonic cells, will elucidate the metabolic pathways and biomarkers indicative of biofilm inception. Disc-shaped stainless-steel coupons (12.7 mm diameter) were employed as a surface for static biofilm establishment. Each disc was immersed in a well, of a 24-well microtiter plate, containing a 1-mL Lysogeny broth (LB) suspension of P. aeruginosa ATCC 9027, a strain known for its biofilm prolificacy. This setup underwent oxygen-depleted incubation at 37°C for 24 hours to yield hypoxic biofilms and the co-existing static planktonic cells. In parallel, another planktonic phenotype of ATCC 9027 was produced in LB under shaking (200 rpm) incubation at 37°C for 24 hours. Planktonic and biofilm cells were harvested, and the intracellular metabolites were subjected to global untargeted metabolomic analysis using LC-MS technology, where small metabolites (below 1.5 kDa) were selected. Data analysis showed the presence of 324 metabolites that differed (p < 0.05) in abundance between planktonic and biofilm cells, whereas 70 metabolites did not vary between these phenotypes (p > 0.05). Correlation, principal components, and partial least square discriminant analyses proved that the biofilm metabolome is distinctly clustered away from that of the two planktonic phenotypes. Based on the functional enrichment analysis, arginine and proline metabolism were enriched in planktonic cells, but butanoate metabolism was enriched in biofilm cells. Key differential metabolites within the butanoate pathway included acetoacetate, 2,3-butandiol, diacetyl, and acetoin, which were highly upregulated in the biofilm compared to the planktonic cells. Exogenous supplementation of acetoin (2 mM), a critical metabolite in butanoate metabolism, augmented biofilm mass, increased the structural integrity and thickness of the biofilm, and maintained the intracellular redox potential by balancing NADH/NAD+ ratio. In conclusion, P. aeruginosa hypoxic biofilm has a specialized metabolic landscape, and butanoate pathway is a metabolic preference and possibly required for promoting planktonic cells to the biofilm state. The butanoate pathway metabolites, particularly acetoin, could serve as markers for biofilm development.
Collapse
Affiliation(s)
- Ahmed G. Abdelhamid
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, Egypt
| | - Ahmed E. Yousef
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
- Department of Microbiology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
213
|
Bonacorsi A, Trespidi G, Scoffone VC, Irudal S, Barbieri G, Riabova O, Monakhova N, Makarov V, Buroni S. Characterization of the dispirotripiperazine derivative PDSTP as antibiotic adjuvant and antivirulence compound against Pseudomonas aeruginosa. Front Microbiol 2024; 15:1357708. [PMID: 38435690 PMCID: PMC10904629 DOI: 10.3389/fmicb.2024.1357708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/06/2024] [Indexed: 03/05/2024] Open
Abstract
Pseudomonas aeruginosa is a major human pathogen, able to establish difficult-to-treat infections in immunocompromised and people with cystic fibrosis (CF). The high rate of antibiotic treatment failure is due to its notorious drug resistance, often mediated by the formation of persistent biofilms. Alternative strategies, capable of overcoming P. aeruginosa resistance, include antivirulence compounds which impair bacterial pathogenesis without exerting a strong selective pressure, and the use of antimicrobial adjuvants that can resensitize drug-resistant bacteria to specific antibiotics. In this work, the dispirotripiperazine derivative PDSTP, already studied as antiviral, was characterized for its activity against P. aeruginosa adhesion to epithelial cells, its antibiotic adjuvant ability and its biofilm inhibitory potential. PDSTP was effective in impairing the adhesion of P. aeruginosa to various immortalized cell lines. Moreover, the combination of clinically relevant antibiotics with the compound led to a remarkable enhancement of the antibiotic efficacy towards multidrug-resistant CF clinical strains. PDSTP-ceftazidime combination maintained its efficacy in vivo in a Galleria mellonella infection model. Finally, the compound showed a promising biofilm inhibitory activity at low concentrations when tested both in vitro and using an ex vivo pig lung model. Altogether, these results validate PDSTP as a promising compound, combining the ability to decrease P. aeruginosa virulence by impairing its adhesion and biofilm formation, with the capability to increase antibiotic efficacy against antibiotic resistant strains.
Collapse
Affiliation(s)
- Andrea Bonacorsi
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Gabriele Trespidi
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Viola C. Scoffone
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Samuele Irudal
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Giulia Barbieri
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Olga Riabova
- Research Center of Biotechnology RAS, Moscow, Russia
| | | | - Vadim Makarov
- Research Center of Biotechnology RAS, Moscow, Russia
| | - Silvia Buroni
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
214
|
Dhakal A, Salim C, Skelly M, Amichan Y, Lamm AT, Hundley HA. ADARs regulate cuticle collagen expression and promote survival to pathogen infection. BMC Biol 2024; 22:37. [PMID: 38360623 PMCID: PMC10870475 DOI: 10.1186/s12915-024-01840-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 02/02/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND In all organisms, the innate immune system defends against pathogens through basal expression of molecules that provide critical barriers to invasion and inducible expression of effectors that combat infection. The adenosine deaminase that act on RNA (ADAR) family of RNA-binding proteins has been reported to influence innate immunity in metazoans. However, studies on the susceptibility of ADAR mutant animals to infection are largely lacking. RESULTS Here, by analyzing adr-1 and adr-2 null mutants in well-established slow-killing assays, we find that both Caenorhabditis elegans ADARs are important for organismal survival to gram-negative and gram-positive bacteria, all of which are pathogenic to humans. Furthermore, our high-throughput sequencing and genetic analysis reveal that ADR-1 and ADR-2 function in the same pathway to regulate collagen expression. Consistent with this finding, our scanning electron microscopy studies indicate adr-1;adr-2 mutant animals also have altered cuticle morphology prior to pathogen exposure. CONCLUSIONS Our data uncover a critical role of the C. elegans ADAR family of RNA-binding proteins in promoting cuticular collagen expression, which represents a new post-transcriptional regulatory node that influences the extracellular matrix. In addition, we provide the first evidence that ADAR mutant animals have altered susceptibility to infection with several opportunistic human pathogens, suggesting a broader role of ADARs in altering physical barriers to infection to influence innate immunity.
Collapse
Affiliation(s)
- Alfa Dhakal
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine-Bloomington, Bloomington, IN, 47405, USA
| | - Chinnu Salim
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Mary Skelly
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Yarden Amichan
- Faculty of Biology, Technion Institute of Technology, Haifa, Israel
| | - Ayelet T Lamm
- Faculty of Biology, Technion Institute of Technology, Haifa, Israel
| | - Heather A Hundley
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
215
|
Nazari-Vanani R, Negahdary M. Recent advances in electrochemical aptasensors and genosensors for the detection of pathogens. ENVIRONMENTAL RESEARCH 2024; 243:117850. [PMID: 38081349 DOI: 10.1016/j.envres.2023.117850] [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: 10/03/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
In recent years, pathogenic microorganisms have caused significant mortality rates and antibiotic resistance and triggered exorbitant healthcare costs. These pathogens often have high transmission rates within human populations. Rapid diagnosis is crucial in controlling and reducing the spread of pathogenic infections. The diagnostic methods currently used against individuals infected with these pathogens include relying on outward symptoms, immunological-based and, some biomolecular ones, which mainly have limitations such as diagnostic errors, time-consuming processes, and high-cost platforms. Electrochemical aptasensors and genosensors have emerged as promising diagnostic tools for rapid, accurate, and cost-effective pathogen detection. These bio-electrochemical platforms have been optimized for diagnostic purposes by incorporating advanced materials (mainly nanomaterials), biomolecular technologies, and innovative designs. This review classifies electrochemical aptasensors and genosensors developed between 2021 and 2023 based on their use of different nanomaterials, such as gold-based, carbon-based, and others that employed other innovative assemblies without the use of nanomaterials. Inspecting the diagnostic features of various sensing platforms against pathogenic analytes can identify research gaps and open new avenues for exploration.
Collapse
Affiliation(s)
- Razieh Nazari-Vanani
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Negahdary
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes, 748, São Paulo, 05508-000, Brazil.
| |
Collapse
|
216
|
Tabassum N, Khan F, Jeong GJ, Oh D, Kim YM. Antibiofilm and antivirulence activities of laminarin-gold nanoparticles in standard and host-mimicking media. Appl Microbiol Biotechnol 2024; 108:203. [PMID: 38349556 PMCID: PMC10864539 DOI: 10.1007/s00253-024-13050-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/02/2024] [Accepted: 02/05/2024] [Indexed: 02/15/2024]
Abstract
The rapidly rising antimicrobial resistance (AMR) in pathogenic bacteria has become one of the most serious public health challenges, with a high death rate. Most pathogenic bacteria have been recognized as a source of AMR and a primary barrier to antimicrobial treatment failure due to the development of biofilms and the production of virulence factors. In this work, nanotechnology was employed as a substitute method to control the formation of biofilms and attenuate virulence features in Pseudomonas aeruginosa and Staphylococcus aureus. We synthesized biocompatible gold nanoparticles from marine-derived laminarin as potential biofilm and virulence treatments. Laminarin-gold nanoparticles (Lam-AuNPs) have been identified as spherical, 49.84 ± 7.32 nm in size and - 26.49 ± 1.29 mV zeta potential. The MIC value of Lam-AuNPs against several drug-resistant microbial pathogens varied from 2 to 1024 μg/mL in both standard and host-mimicking media. Sub-MIC values of Lam-AuNPs were reported to effectively reduce the production of P. aeruginosa and S. aureus biofilms in both standard and host-mimicking growth media. Furthermore, the sub-MIC of Lam-AuNPs strongly reduced hemolysis, pyocyanin, pyoverdine, protease, and several forms of flagellar and pili-mediated motility in P. aeruginosa. Lam-AuNPs also inhibited S. aureus hemolysis and the production of amyloid fibrils. The Lam-AuNPs strongly dispersed the preformed mature biofilm of these pathogens in a dose-dependent manner. The Lam-AuNPs would be considered an alternative antibiofilm and antivirulence agent to control P. aeruginosa and S. aureus infections. KEY POINTS: • Lam-AuNPs were biosynthesized to control biofilm and virulence. • Lam-AuNPs show effective biofilm inhibition in standard and host-mimicking media. • Lam-AuNPs suppress various virulence factors of P. aeruginosa and S. aureus.
Collapse
Affiliation(s)
- Nazia Tabassum
- 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
| | - Fazlurrahman Khan
- 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.
- Institute of Fisheries Sciences, Pukyong National University, Busan, 48513, Republic of Korea.
| | - Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Dokyung Oh
- 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
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Young-Mog Kim
- 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.
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
217
|
Kar A, Mukherjee SK, Barik S, Hossain ST. Antimicrobial Activity of Trigonelline Hydrochloride Against Pseudomonas aeruginosa and Its Quorum-Sensing Regulated Molecular Mechanisms on Biofilm Formation and Virulence. ACS Infect Dis 2024; 10:746-762. [PMID: 38232080 DOI: 10.1021/acsinfecdis.3c00617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Pseudomonas aeruginosa, a vivid biofilm-producing bacterium, is considered a dreadful opportunistic pathogen, and thus, management of biofilm-associated infections due to multidrug resistant strains by traditional drugs currently is of great concern. This study was aimed to assess the impact of trigonelline hydrochloride, a pyridine alkaloid, on P. aeruginosa PAO1, in search of an alternative therapeutant. The effect of trigonelline on colony morphology and motility was studied along with its role on biofilm and expression virulence factors. Trigonelline influenced the colony structure, motility, biofilm architecture, and the production of virulence factors in a dose-dependent manner. Alterations in quorum sending (QS)-regulated gene expression after treatment and molecular docking analysis for certain regulator proteins confirmed its effect on the QS-system network by affecting Las, Rhl, and Pqs signaling pathways and as possible molecular targets. Thus, trigonelline might be considered as a potential chemical lead to manage biofilm-associated pathogenesis or to develop other analogues with enhanced pharmacokinetic actions.
Collapse
Affiliation(s)
- Amiya Kar
- Department of Microbiology, University of Kalyani, Kalyani 741235, India
| | | | - Subhasis Barik
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, Kolkata, West Bengal 700026, India
| | | |
Collapse
|
218
|
Safwan SM, Kumar N, Mehta D, Singh M, Saini V, Pandey N, Khatol S, Batheja S, Singh J, Walia P, Bajaj A. Xanthone Derivatives Enhance the Therapeutic Potential of Neomycin against Polymicrobial Gram-Negative Bacterial Infections. ACS Infect Dis 2024; 10:527-540. [PMID: 38294409 DOI: 10.1021/acsinfecdis.3c00471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Gram-negative bacterial infections are difficult to manage as many antibiotics are ineffective owing to the presence of impermeable bacterial membranes. Polymicrobial infections pose a serious threat due to the inadequate efficacy of available antibiotics, thereby necessitating the administration of antibiotics at higher doses. Antibiotic adjuvants have emerged as a boon as they can augment the therapeutic potential of available antibiotics. However, the toxicity profile of antibiotic adjuvants is a major hurdle in clinical translation. Here, we report the design, synthesis, and biological activities of xanthone-derived molecules as potential antibiotic adjuvants. Our SAR studies witnessed that the p-dimethylamino pyridine-derivative of xanthone (X8) enhances the efficacy of neomycin (NEO) against Escherichia coli and Pseudomonas aeruginosa and causes a synergistic antimicrobial effect without any toxicity against mammalian cells. Biochemical studies suggest that the combination of X8 and NEO, apart from inhibiting protein synthesis, enhances the membrane permeability by binding to lipopolysaccharide. Notably, the combination of X8 and NEO can disrupt the monomicrobial and polymicrobial biofilms and show promising therapeutic potential against a murine wound infection model. Collectively, our results unveil the combination of X8 and NEO as a suitable adjuvant therapy for the inhibition of the Gram-negative bacterial infections.
Collapse
Affiliation(s)
- Sayed Mohamad Safwan
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Neeraj Kumar
- Lord Shiva College of Pharmacy, Near Civil Hospital, Sirsa 125055, Haryana, India
| | - Devashish Mehta
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Mohit Singh
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Varsha Saini
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Nishant Pandey
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Steffi Khatol
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| | - Shalini Batheja
- Lord Shiva College of Pharmacy, Near Civil Hospital, Sirsa 125055, Haryana, India
| | - Jitender Singh
- Lord Shiva College of Pharmacy, Near Civil Hospital, Sirsa 125055, Haryana, India
| | - Preeti Walia
- Lord Shiva College of Pharmacy, Near Civil Hospital, Sirsa 125055, Haryana, India
| | - Avinash Bajaj
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Third Milestone, Faridabad-Gurgaon Expressway, Faridabad 121001, Haryana, India
| |
Collapse
|
219
|
Luo M, Li S, Luo W. Comparative analysis of antibiotic susceptibility patterns and clinical features of mucoid and non-mucoid Pseudomonas aeruginosa infections: a retrospective study. Front Public Health 2024; 12:1333477. [PMID: 38389944 PMCID: PMC10881668 DOI: 10.3389/fpubh.2024.1333477] [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: 11/05/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Background Pseudomonas aeruginosa (PA) is a prevalent opportunistic pathogen that has close associations with both acute and chronic infections. However, there exists an insufficiency of accurate and comprehensive data pertaining to the antimicrobial susceptibility patterns and clinical characteristics of both mucoid and non-mucoid strains of PA (mPA and non-mPA, respectively). Methods From January 1, 2021 to December 31, 2022, a thorough retrospective study was carried out to examine and compare the antibiotic susceptibility test outcomes and clinical characteristics of hospitalized patients with mPA and non-mPA infections. Results This study investigated a cohort of 111 patients who were diagnosed with mPA infections, as well as 792 patients diagnosed with non-mPA infections. Significant demographic disparities, including gender (p < 0.001), age (p < 0.001), length of hospital stay (p < 0.001), diabetes (p = 0.043), and hypertension (p < 0.001), are evident between the mPA and non-mPA groups. The mPA group commonly necessitates hospitalization for respiratory system diseases, whereas the non-mPA group is associated with concomitant cardiovascular and cerebrovascular diseases. The mPA group demonstrates lower utilization rates of medical devices, such as Foley catheter (p < 0.001), nasogastric tube (p < 0.001), mechanical ventilation (p < 0.001), tracheostomy (p < 0.001), arterial and venous catheterization (p < 0.001), and exhibits superior organ function status, including lower incidences of hypoalbuminemia (p < 0.001), septic shock (p < 0.001), liver dysfunction (p < 0.001), renal failure (p < 0.001), and respiratory failure (p < 0.001). The non-mPA group is more vulnerable to infection with two or more bacterial pathogens compared to the mPA group, with the non-mPA group frequently resulting in Enterobacteriaceae infections and the mPA group being associated with fungal infections. Variations in antibiotic sensitivity are noted for Amikacin (p < 0.001), Ciprofloxacin (p < 0.001), Cefepime (p = 0.003), and Levofloxacin (p < 0.001) in antibiotic susceptibility testing, with resistance patterns closely tied to specific antibiotic usage. Conclusion There are significant demographic characteristics, clinical manifestations and antibiotic susceptibility between mPA and non-mPA infections. It is crucial to emphasize these characteristics due to their significant role in preventing and treating PA infections.
Collapse
Affiliation(s)
- Maoling Luo
- Medical Laboratory Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Si Li
- General Medicine, Clinical Medicine, Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Wenying Luo
- Medical Laboratory Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
220
|
Vaňková E, Julák J, Machková A, Obrová K, Klančnik A, Smole Možina S, Scholtz V. Overcoming antibiotic resistance: non-thermal plasma and antibiotics combination inhibits important pathogens. Pathog Dis 2024; 82:ftae007. [PMID: 38730561 PMCID: PMC11094553 DOI: 10.1093/femspd/ftae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/27/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024] Open
Abstract
Antibiotic resistance (ATBR) is increasing every year as the overuse of antibiotics (ATBs) and the lack of newly emerging antimicrobial agents lead to an efficient pathogen escape from ATBs action. This trend is alarming and the World Health Organization warned in 2021 that ATBR could become the leading cause of death worldwide by 2050. The development of novel ATBs is not fast enough considering the situation, and alternative strategies are therefore urgently required. One such alternative may be the use of non-thermal plasma (NTP), a well-established antimicrobial agent actively used in a growing number of medical fields. Despite its efficiency, NTP alone is not always sufficient to completely eliminate pathogens. However, NTP combined with ATBs is more potent and evidence has been emerging over the last few years proving this is a robust and highly effective strategy to fight resistant pathogens. This minireview summarizes experimental research addressing the potential of the NTP-ATBs combination, particularly for inhibiting planktonic and biofilm growth and treating infections in mouse models caused by methicillin-resistant Staphylococcus aureus or Pseudomonas aeruginosa. The published studies highlight this combination as a promising solution to emerging ATBR, and further research is therefore highly desirable.
Collapse
Affiliation(s)
- Eva Vaňková
- Department of Physics and Measurements, University of Chemistry and Technology in Prague, 160 00 Prague, Czech Republic
- Department of Biotechnology, University of Chemistry and Technology in Prague, 160 00 Prague, Czech Republic
| | - Jaroslav Julák
- Department of Physics and Measurements, University of Chemistry and Technology in Prague, 160 00 Prague, Czech Republic
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University in Prague, 160 00 Prague, Czech Republic
| | - Anna Machková
- Department of Physics and Measurements, University of Chemistry and Technology in Prague, 160 00 Prague, Czech Republic
| | - Klára Obrová
- Department of Physics and Measurements, University of Chemistry and Technology in Prague, 160 00 Prague, Czech Republic
| | - Anja Klančnik
- Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Sonja Smole Možina
- Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Vladimír Scholtz
- Department of Physics and Measurements, University of Chemistry and Technology in Prague, 160 00 Prague, Czech Republic
| |
Collapse
|
221
|
Papa-Ezdra R, Outeda M, Cordeiro NF, Araújo L, Gadea P, Garcia-Fulgueiras V, Seija V, Bado I, Vignoli R. Outbreak of Pseudomonas aeruginosa High-Risk Clone ST309 Serotype O11 Featuring blaPER-1 and qnrVC6. Antibiotics (Basel) 2024; 13:159. [PMID: 38391545 PMCID: PMC10885872 DOI: 10.3390/antibiotics13020159] [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: 11/30/2023] [Revised: 12/26/2023] [Accepted: 12/27/2023] [Indexed: 02/24/2024] Open
Abstract
Pseudomonas aeruginosa is a leading cause of hospital-acquired infections worldwide. Biofilm production, antibiotic resistance, and a wide range of virulence factors contribute to their persistence in nosocomial environments. We describe an outbreak caused by a multidrug-resistant P. aeruginosa strain in an ICU. Antibiotic susceptibility was determined and blaPER-1 and qnrVC were amplified via PCR. Clonality was determined using PFGE and biofilm formation was studied with a static model. A combination of antibiotics was assessed on both planktonic cells and biofilms. WGS was performed on five isolates. All isolates were clonally related, resistant to ceftazidime, cefepime, amikacin, and ceftolozane-tazobactam, and harbored blaPER-1; 11/19 possessed qnrVC. Meropenem and ciprofloxacin reduced the biofilm biomass; however, the response to antibiotic combinations with rifampicin was different between planktonic cells and biofilms. WGS revealed that the isolates belonged to ST309 and serotype O11. blaPER-1 and qnrVC6 were associated with a tandem of ISCR1 as part of a complex class one integron, with aac(6')-Il and ltrA as gene cassettes. The structure was associated upstream and downstream with Tn4662 and flanked by direct repeats, suggesting its horizontal mobilization capability as a composite transposon. ST309 is considered an emerging high-risk clone that should be monitored in the Americas.
Collapse
Affiliation(s)
- Romina Papa-Ezdra
- Departamento de Bacteriología y Virología, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Av. Alfredo Navarro 3051, CP 11600 Montevideo, Uruguay
| | - Matilde Outeda
- Departamento de Laboratorio Clínico, Área Microbiología, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Av. Italia s/n, CP 11600 Montevideo, Uruguay
| | - Nicolás F Cordeiro
- Departamento de Bacteriología y Virología, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Av. Alfredo Navarro 3051, CP 11600 Montevideo, Uruguay
| | - Lucía Araújo
- Departamento de Bacteriología y Virología, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Av. Alfredo Navarro 3051, CP 11600 Montevideo, Uruguay
| | - Pilar Gadea
- Departamento de Laboratorio Clínico, Área Microbiología, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Av. Italia s/n, CP 11600 Montevideo, Uruguay
| | - Virginia Garcia-Fulgueiras
- Departamento de Bacteriología y Virología, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Av. Alfredo Navarro 3051, CP 11600 Montevideo, Uruguay
| | - Verónica Seija
- Departamento de Laboratorio Clínico, Área Microbiología, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Av. Italia s/n, CP 11600 Montevideo, Uruguay
| | - Inés Bado
- Departamento de Bacteriología y Virología, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Av. Alfredo Navarro 3051, CP 11600 Montevideo, Uruguay
| | - Rafael Vignoli
- Departamento de Bacteriología y Virología, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Av. Alfredo Navarro 3051, CP 11600 Montevideo, Uruguay
| |
Collapse
|
222
|
Mancuso G, Trinchera M, Midiri A, Zummo S, Vitale G, Biondo C. Novel Antimicrobial Approaches to Combat Bacterial Biofilms Associated with Urinary Tract Infections. Antibiotics (Basel) 2024; 13:154. [PMID: 38391540 PMCID: PMC10886225 DOI: 10.3390/antibiotics13020154] [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: 01/16/2024] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Urinary tract infections (UTIs) are prevalent bacterial infections in both community and healthcare settings. They account for approximately 40% of all bacterial infections and require around 15% of all antibiotic prescriptions. Although antibiotics have traditionally been used to treat UTIs for several decades, the significant increase in antibiotic resistance in recent years has made many previously effective treatments ineffective. Biofilm on medical equipment in healthcare settings creates a reservoir of pathogens that can easily be transmitted to patients. Urinary catheter infections are frequently observed in hospitals and are caused by microbes that form a biofilm after a catheter is inserted into the bladder. Managing infections caused by biofilms is challenging due to the emergence of antibiotic resistance. Biofilms enable pathogens to evade the host's innate immune defences, resulting in long-term persistence. The incidence of sepsis caused by UTIs that have spread to the bloodstream is increasing, and drug-resistant infections may be even more prevalent. While the availability of upcoming tests to identify the bacterial cause of infection and its resistance spectrum is critical, it alone will not solve the problem; innovative treatment approaches are also needed. This review analyses the main characteristics of biofilm formation and drug resistance in recurrent uropathogen-induced UTIs. The importance of innovative and alternative therapies for combatting biofilm-caused UTI is emphasised.
Collapse
Affiliation(s)
- Giuseppe Mancuso
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Marilena Trinchera
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Angelina Midiri
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Sebastiana Zummo
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Giulia Vitale
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Carmelo Biondo
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| |
Collapse
|
223
|
Sikder S, Toha M, Anik AH, Sultan MB, Alam M, Parvin F, Tareq SM. A comprehensive review on the fate and impact of antibiotic residues in the environment and public health: A special focus on the developing countries. WATER ENVIRONMENT RESEARCH : A RESEARCH PUBLICATION OF THE WATER ENVIRONMENT FEDERATION 2024; 96:e10987. [PMID: 38342763 DOI: 10.1002/wer.10987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 02/13/2024]
Abstract
The widespread application of antibiotics in human and veterinary medicine has led to the pervasive presence of antibiotic residues in the environment, posing a potential hazard to public health. This comprehensive review aims to scrutinize the fate and impact of antibiotic residues, with a particular focus on the context of developing nations. The investigation delves into the diverse pathways facilitating the entry of antibiotics into the environment and meticulously examines their effects on human health. The review delineates the current state of antibiotic residues, evaluates their exposure in developing nations, and elucidates existing removal methodologies. Additionally, it probes into the factors contributing to the endurance and ecotoxicity of antibiotic residues, correlating these aspects with usage rates and associated mortalities in these nations. The study also investigates removal techniques for antibiotic residues, assessing their efficiency in environmental compartments. The concurrent emergence of antibiotic-resistant bacteria, engendered by antibiotic residues, and their adverse ecological threats underscore the necessity for enhanced regulations, vigilant surveillance programs, and the adoption of sustainable alternatives. The review underlines the pivotal role of public education and awareness campaigns in promoting responsible antibiotic use. The synthesis concludes with strategic recommendations, strengthening the imperative for further research encompassing comprehensive monitoring, ecotoxicological effects, alternative strategies, socio-economic considerations, and international collaborations, all aimed at mitigating the detrimental effects of antibiotic residues on human health and the environment. PRACTITIONER POINTS: Antibiotic residues are widely distributed in different environmental compartments. Developing countries use more antibiotics than developed countries. Human and veterinary wastes are one of the most responsible sources of antibiotic pollution. Antibiotics interact with biological systems and trigger pharmacological reactions at low doses. Antibiotics can be removed using modern biological, chemical, and physical-chemical techniques.
Collapse
Affiliation(s)
- Sadia Sikder
- Department of Environmental Science, Bangladesh University of Professionals (BUP), Bangladesh
- Department of Environmental Science and Disaster Management, Daffodil International University, Birulia, Savar, Dhaka, Bangladesh
| | - Mohammad Toha
- Department of Environmental Science, Bangladesh University of Professionals (BUP), Bangladesh
| | - Amit Hasan Anik
- Department of Environmental Science, Bangladesh University of Professionals (BUP), Bangladesh
| | - Maisha Binte Sultan
- Department of Environmental Science, Bangladesh University of Professionals (BUP), Bangladesh
| | - Mahbub Alam
- Department of Environmental Science, Bangladesh University of Professionals (BUP), Bangladesh
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Fahmida Parvin
- Hydrobiogeochemistry and Pollution Control Laboratory, Department of Environmental Sciences, Jahangirnagar University, Dhaka, Bangladesh
| | - Shafi M Tareq
- Department of Environmental Science, Bangladesh University of Professionals (BUP), Bangladesh
- Hydrobiogeochemistry and Pollution Control Laboratory, Department of Environmental Sciences, Jahangirnagar University, Dhaka, Bangladesh
| |
Collapse
|
224
|
Deery J, Carmody M, Flavin R, Tomanek M, O'Keeffe M, McGlacken GP, Reen FJ. Comparative genomics reveals distinct diversification patterns among LysR-type transcriptional regulators in the ESKAPE pathogen Pseudomonas aeruginosa. Microb Genom 2024; 10:001205. [PMID: 38421269 PMCID: PMC10926688 DOI: 10.1099/mgen.0.001205] [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: 10/05/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Pseudomonas aeruginosa, a harmful nosocomial pathogen associated with cystic fibrosis and burn wounds, encodes for a large number of LysR-type transcriptional regulator proteins. To understand how and why LTTR proteins evolved with such frequency and to establish whether any relationships exist within the distribution we set out to identify the patterns underpinning LTTR distribution in P. aeruginosa and to uncover cluster-based relationships within the pangenome. Comparative genomic studies revealed that in the JGI IMG database alone ~86 000 LTTRs are present across the sequenced genomes (n=699). They are widely distributed across the species, with core LTTRs present in >93 % of the genomes and accessory LTTRs present in <7 %. Analysis showed that subsets of core LTTRs can be classified as either variable (typically specific to P. aeruginosa) or conserved (and found to be distributed in other Pseudomonas species). Extending the analysis to the more extensive Pseudomonas database, PA14 rooted analysis confirmed the diversification patterns and revealed PqsR, the receptor for the Pseudomonas quinolone signal (PQS) and 2-heptyl-4-quinolone (HHQ) quorum-sensing signals, to be amongst the most variable in the dataset. Successful complementation of the PAO1 pqsR - mutant using representative variant pqsR sequences suggests a degree of structural promiscuity within the most variable of LTTRs, several of which play a prominent role in signalling and communication. These findings provide a new insight into the diversification of LTTR proteins within the P. aeruginosa species and suggests a functional significance to the cluster, conservation and distribution patterns identified.
Collapse
Affiliation(s)
- Jamie Deery
- School of Microbiology, University College Cork, Cork, Ireland
| | - Muireann Carmody
- School of Microbiology, University College Cork, Cork, Ireland
- School of Chemistry, University College Cork, Cork, Ireland
| | - Rhiannon Flavin
- School of Microbiology, University College Cork, Cork, Ireland
| | - Malwina Tomanek
- School of Microbiology, University College Cork, Cork, Ireland
| | - Maria O'Keeffe
- School of Microbiology, University College Cork, Cork, Ireland
| | - Gerard P. McGlacken
- School of Chemistry, University College Cork, Cork, Ireland
- Synthesis and Solid State Pharmaceutical Centre, University College Cork, Cork, Ireland
| | - F. Jerry Reen
- School of Microbiology, University College Cork, Cork, Ireland
- Synthesis and Solid State Pharmaceutical Centre, University College Cork, Cork, Ireland
| |
Collapse
|
225
|
Nayak SPRR, Basty C, Boopathi S, Dhivya LS, Alarjani KM, Gawwad MRA, Hager R, Kathiravan MK, Arockiaraj J. Furan-based Chalcone Annihilates the Multi-Drug-Resistant Pseudomonas aeruginosa and Protects Zebra Fish Against its Infection. J Microbiol 2024; 62:75-89. [PMID: 38383881 DOI: 10.1007/s12275-024-00103-6] [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: 09/19/2023] [Revised: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 02/23/2024]
Abstract
The emergence of carbapenem-resistant Pseudomonas aeruginosa, a multi-drug-resistant bacteria, is becoming a serious public health concern. This bacterium infects immunocompromised patients and has a high fatality rate. Both naturally and synthetically produced chalcones are known to have a wide array of biological activities. The antibacterial properties of synthetically produced chalcone were studied against P. aeruginosa. In vitro, study of the compound (chalcone derivative named DKO1), also known as (2E)-1-(5-methylfuran-2-yl)-3-(4-nitrophenyl) prop-2-en-1-one, had substantial antibacterial and biofilm disruptive action. DKO1 effectively shielded against P. aeruginosa-induced inflammation, oxidative stress, lipid peroxidation, and apoptosis in zebrafish larvae. In adult zebrafish, the treatment enhanced the chances of survivability and reduced the sickness-like behaviors. Gene expression, biochemical analysis, and histopathology studies found that proinflammatory cytokines (TNF-α, IL-1β, IL-6, iNOS) were down regulated; antioxidant enzymes such as superoxide dismutase (SOD) and catalase (CAT) levels increased, and histoarchitecture was restored in zebrafish. The data indicate that DKO1 is an effective antibacterial agent against P. aeruginosa demonstrated both in vitro and in vivo.
Collapse
Affiliation(s)
- Santosh Pushpa Ramya Ranjan Nayak
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India
| | - Catharine Basty
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India
| | - Seenivasan Boopathi
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India
| | - Loganathan Sumathi Dhivya
- Dr. APJ Abdul Kalam Research Lab, Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India
| | - Khaloud Mohammed Alarjani
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mohamed Ragab Abdel Gawwad
- Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, 71210, Bosnia and Herzegovina
| | - Raghda Hager
- Department of Medical Microbiology and Immunology, King Salman International University, South Sinai, Egypt
| | - Muthu Kumaradoss Kathiravan
- Dr. APJ Abdul Kalam Research Lab, Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India.
| |
Collapse
|
226
|
Xi W, Zhang X, Zhu X, Wang J, Xue H, Pan H. Distribution patterns and influential factors of pathogenic bacteria in freshwater aquaculture sediments. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:16028-16047. [PMID: 38308166 DOI: 10.1007/s11356-024-31897-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/03/2024] [Indexed: 02/04/2024]
Abstract
Pathogenic bacteria, the major causative agents of aquaculture diseases, are a serious impediment to the aquaculture industry. However, the bioinformatics of pathogenic bacteria and virulence factors (VFs) in sediments, an important component of freshwater aquaculture ecosystems, are not well characterized. In this study, 20 sediment samples were collected from fish pond sediments (FPS), shrimp field sediments (SFS), fish pond sediment control (FPSC), and shrimp field sediment control (SFSC). Molecular biological information was obtained on a total of 173 pathogenic bacteria, 1093 virulence factors (VFs), and 8475 mobile genetic elements (MGEs) from these samples. The results indicated that (1) aquaculture patterns and sediment characteristics can affect the distribution of pathogenic bacteria. According to the results of the Kruskal-Wallis H test, except for Mycobacterium gilvum, there were significant differences (P < 0.05) among the four sediment types in the average abundance of major pathogenic bacteria (top 30 in abundance), and the average abundance of major pathogenic bacteria in the four sediment types followed the following pattern: FPS > SFS > FPSC > SFSC. (2) Pathogenic bacteria are able to implement a variety of complex pathogenic mechanisms such as adhesion, invasion, immune evasion, and metabolic regulation in the host because they carry a variety of VFs such as type IV pili, HSI-I, Alginate, Colibactin, and Capsule. According to the primary classification of the Virulence Factor Database (VFDB), the abundance of VFs in all four types of sediments showed the following pattern: offensive VFs > non-specific VFs > defensive VFs > regulation of virulence-related genes. (3) Total organic carbon (TOC), total phosphorus (TP), available phosphorus (AP), nitrite, and nitrate were mostly only weakly positively correlated with the major pathogenic bacteria and could promote the growth of pathogenic bacteria to some extent, whereas ammonia was significantly positively correlated with most of the major pathogenic bacteria and could play an important role in promoting the growth and reproduction of pathogenic bacteria. (4) Meanwhile, there was also a significant positive correlation between CAZyme genes and major pathogenic bacteria (0.62 ≤ R ≤ 0.89, P < 0.05). This suggests that these pathogenic bacteria could be the main carriers of CAZyme genes and, to some extent, gained a higher level of metabolic activity by degrading organic matter in the sediments to maintain their competitive advantage. (5) Worryingly, the results of correlation analyses indicated that MGEs in aquaculture sediments could play an important role in the spread of VFs (R = 0.82, P < 0.01), and in particular, plasmids (R = 0.75, P < 0.01) and integrative and conjugative elements (ICEs, R = 0.65, P < 0.05) could be these major vectors of VFs. The results of this study contribute to a comprehensive understanding of the health of freshwater aquaculture sediments and provide a scientific basis for aquaculture management and conservation.
Collapse
Affiliation(s)
- Wenxiang Xi
- Hubei Key Laboratory of Petroleum Geochemistry and Environment, Yangtze University, Wuhan, 430100, Hubei, China
- College of Resources and Environment, Yangtze University, Wuhan, 430100, Hubei, China
| | - Xun Zhang
- China Coal Mine Construction Group Co., LTD, Hefei, 230071, Anhui, China
| | - Xianbin Zhu
- Hubei Key Laboratory of Petroleum Geochemistry and Environment, Yangtze University, Wuhan, 430100, Hubei, China
- College of Resources and Environment, Yangtze University, Wuhan, 430100, Hubei, China
| | - Jiaming Wang
- Hubei Key Laboratory of Petroleum Geochemistry and Environment, Yangtze University, Wuhan, 430100, Hubei, China
- College of Resources and Environment, Yangtze University, Wuhan, 430100, Hubei, China
| | - Han Xue
- Hubei Key Laboratory of Petroleum Geochemistry and Environment, Yangtze University, Wuhan, 430100, Hubei, China
- College of Resources and Environment, Yangtze University, Wuhan, 430100, Hubei, China
| | - Hongzhong Pan
- Hubei Key Laboratory of Petroleum Geochemistry and Environment, Yangtze University, Wuhan, 430100, Hubei, China.
- College of Resources and Environment, Yangtze University, Wuhan, 430100, Hubei, China.
| |
Collapse
|
227
|
Abdelmalek S, Hajar M, Salah L, Abdel-Halim H. In Silico Screening and Experimental Validation of Novel MexAB-OprM Efflux Pump Inhibitors of Pseudomonas aeruginosa. Microb Drug Resist 2024; 30:73-81. [PMID: 38150012 DOI: 10.1089/mdr.2023.0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023] Open
Abstract
The emergence of multidrug-resistant Pseudomonas aeruginosa possesses a significant public health concern. Constitutively expressed MexAB-OprM efflux pumps in P. aeruginosa significantly contribute to its resistance to a variety of antibiotics. The development of efflux pump inhibitors (EPIs) has emerged as an attractive strategy in reversing antibiotic resistance. In this study, structure-based virtual screening techniques were used for the identification of new MexAB-OprM efflux inhibitors. The predicted poses were thoroughly filtered by induced fit docking procedures followed by in vitro microbiological assays for the validation of in silico results. Two compounds, NSC-147850 and NSC-112703, were able to restore tetracycline susceptibility in MexAB-OprM overexpressing Pseudomonas aeruginosa ATCC® 27853™ strain. This correlation observed between in silico screening and positive efflux inhibitory activity in vitro suggests that NSC-147850 and NSC-112703 have potential as EPIs and may be effective in combination therapy against drug-resistant strains of P. aeruginosa.
Collapse
Affiliation(s)
- Suzanne Abdelmalek
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| | - Malak Hajar
- Health Data Science Lab, Department of Genetics and Genomics, College of Medical and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Luma Salah
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| | - Heba Abdel-Halim
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| |
Collapse
|
228
|
Wang C, Xiao R, Yang Q, Pan J, Cui P, Zhou S, Qiu L, Zhang Y, Wang J. Green synthesis of epigallocatechin gallate-ferric complex nanoparticles for photothermal enhanced antibacterial and wound healing. Biomed Pharmacother 2024; 171:116175. [PMID: 38266620 DOI: 10.1016/j.biopha.2024.116175] [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: 10/12/2023] [Revised: 01/11/2024] [Accepted: 01/13/2024] [Indexed: 01/26/2024] Open
Abstract
Bacterial infections are a significant global health concern, particularly in the context of skin infections and chronic wounds, which was further exacerbated by the emerging of antibiotic resistance. Therefore, there are urgent needs to develop alternative antibacterial strategies without inducing significant resistance. Photothermal therapy (PTT) is a promising alternative approach but usually faces limitations such as the need for stable and environmental-friendly PTT agents and ensuring biocompatibility with living tissues, necessitating ongoing research for its clinical advancement. Herein, in this study, with the aim to develop a green synthesized PTT agent for photothermal enhanced antibacterial and wound healing, we proposed a facile one-pot method to prepare epigallocatechin gallate-ferric (EGCG-Fe) complex nanoparticles. The obtained nanoparticles showed improved good size distribution and stability with high reproducibility. More importantly, EGCG-Fe complex nanoparticles have additional photothermal conversion ability which can give photothermal enhanced antibacterial effect on various pathogens, including Gram-positive Staphylococcus aureus (S. aureus) and Gram-negative Escherichia coli (E. coli) strains. EGCG-Fe complex nanoparticles also showed powerful biofilm prevention and destruction effects with promoted antibacterial and wound healing on mice model. In conclusion, EGCG-Fe complex nanoparticles can be a robust green material with effective and novel light controllable antibacterial properties for photothermal enhanced antibacterial and wound healing applications.
Collapse
Affiliation(s)
- Cheng Wang
- School of Pharmacy Changzhou University, Changzhou, Jiangsu 213164, PR China.
| | - Ru Xiao
- School of Pharmacy Changzhou University, Changzhou, Jiangsu 213164, PR China
| | - Qingbo Yang
- School of Pharmacy Changzhou University, Changzhou, Jiangsu 213164, PR China
| | - Jiaoyang Pan
- School of Pharmacy Changzhou University, Changzhou, Jiangsu 213164, PR China
| | - Pengfei Cui
- School of Pharmacy Changzhou University, Changzhou, Jiangsu 213164, PR China
| | - Shuwen Zhou
- School of Pharmacy Changzhou University, Changzhou, Jiangsu 213164, PR China
| | - Lin Qiu
- School of Pharmacy Changzhou University, Changzhou, Jiangsu 213164, PR China
| | - Yajing Zhang
- College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, PR China; Hebei Higher Education Applied Technology Research Center of TCM Development and Industrialization, Hebei University of Chinese Medicine, Shijiazhuang 050200, PR China.
| | - Jianhao Wang
- School of Pharmacy Changzhou University, Changzhou, Jiangsu 213164, PR China.
| |
Collapse
|
229
|
Mahdi LH, Hasoon BA, Sulaiman GM, Mohammed HA, Jawad KH, Al-Dulimi AG, Essa RH, Albukhaty S, Khan R. Anti-microbial efficacy of L-glutaminase (EC 3.5.1.2) against multidrug-resistant Pseudomonas aeruginosa infection. J Antibiot (Tokyo) 2024; 77:111-119. [PMID: 38017084 DOI: 10.1038/s41429-023-00678-z] [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: 04/09/2023] [Revised: 09/12/2023] [Accepted: 10/28/2023] [Indexed: 11/30/2023]
Abstract
The aims of this study were isolation-purification and characterization of L-glutaminase from L. gasseri BRLHM clinical isolates and investigation of its efficiency as an antimicrobial agent against multidrug-resistant P. aeruginosa. The MICs of L-glutaminase and gentamicin reference were evaluated by the well-diffusion method. The biofilm on the IUD contraceptive was visualized using atomic force microscopy (AFM) image analyses. The purified L-glutaminase possessed significant antimicrobial activity against P. aeruginosa isolates (p < 0.05), and the antibiofilm formation activity of the purified L-glutaminase was stronger than the antibiofilm activity of the referral standard drug, gentamicin (P < 0.05), which were checked by the inhibition of the biofilm formation on the IUD contraceptive device. Investigations indicated that L-glutaminase may have a crucial role in future clinical applications.
Collapse
Affiliation(s)
- Likaa H Mahdi
- Department of Biology, College of Science, Mustansiriyah University, Baghdad, Iraq
| | - Buthenia A Hasoon
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Ghassan M Sulaiman
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, Iraq.
| | - Hamdoon A Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim, 51452, Saudi Arabia.
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt.
| | - Kareem H Jawad
- Department of LASER and Optoelectronic Engineering, University of Technology, Baghdad, Iraq
| | - Ali G Al-Dulimi
- Department of Dentistry, Bilad Alrafidain University College, Diyala, 32001, Iraq
| | - Rajwa H Essa
- Department of Biology, College of Science, Mustansiriyah University, Baghdad, Iraq
| | - Salim Albukhaty
- Department of Chemistry, College of Science, University of Misan, Maysan, 62001, Iraq
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
| | - Riaz Khan
- Manav Rachna International Institute of Research and Study (MRIIRS), Faridabad, HR, 121 001, India.
| |
Collapse
|
230
|
Vinayavekhin N, Wattanophas T, Murphy MF, Vangnai AS, Hobbs G. Metabolomics responses and tolerance of Pseudomonas aeruginosa under acoustic vibration stress. PLoS One 2024; 19:e0297030. [PMID: 38285708 PMCID: PMC10824448 DOI: 10.1371/journal.pone.0297030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/25/2023] [Indexed: 01/31/2024] Open
Abstract
Sound has been shown to impact microbial behaviors. However, our understanding of the chemical and molecular mechanisms underlying these microbial responses to acoustic vibration is limited. In this study, we used untargeted metabolomics analysis to investigate the effects of 100-Hz acoustic vibration on the intra- and extracellular hydrophobic metabolites of P. aeruginosa PAO1. Our findings revealed increased levels of fatty acids and their derivatives, quinolones, and N-acylethanolamines upon sound exposure, while rhamnolipids (RLs) showed decreased levels. Further quantitative real-time polymerase chain reaction experiments showed slight downregulation of the rhlA gene (1.3-fold) and upregulation of fabY (1.5-fold), fadE (1.7-fold), and pqsA (1.4-fold) genes, which are associated with RL, fatty acid, and quinolone biosynthesis. However, no alterations in the genes related to the rpoS regulators or quorum-sensing networks were observed. Supplementing sodium oleate to P. aeruginosa cultures to simulate the effects of sound resulted in increased tolerance of P. aeruginosa in the presence of sound at 48 h, suggesting a potential novel response-tolerance correlation. In contrast, adding RL, which went against the response direction, did not affect its growth. Overall, these findings provide potential implications for the control and manipulation of virulence and bacterial characteristics for medical and industrial applications.
Collapse
Affiliation(s)
- Nawaporn Vinayavekhin
- Center of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Thanyaporn Wattanophas
- Center of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Mark Francis Murphy
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Alisa S. Vangnai
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Glyn Hobbs
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
231
|
Yin C, Alam MZ, Fallon JT, Huang W. Advances in Development of Novel Therapeutic Strategies against Multi-Drug Resistant Pseudomonas aeruginosa. Antibiotics (Basel) 2024; 13:119. [PMID: 38391505 PMCID: PMC10885988 DOI: 10.3390/antibiotics13020119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) with multi-drug resistance (MDR) is a major cause of serious healthcare-associated infections, leading to high morbidity and mortality. This opportunistic pathogen is responsible for various infectious diseases, such as those seen in cystic fibrosis, ventilator-associated pneumonia, urinary tract infection, otitis externa, and burn and wound injuries. Due to its relatively large genome, P. aeruginosa has great diversity and can use various molecular mechanisms for antimicrobial resistance. For example, outer membrane permeability can contribute to antimicrobial resistance and is determined by lipopolysaccharide (LPS) and porin proteins. Recent findings on the regulatory interaction between peptidoglycan and LPS synthesis provide additional clues against pathogenic P. aeruginosa. This review focuses on recent advances in antimicrobial agents and inhibitors targeting LPS and porin proteins. In addition, we explore current and emerging treatment strategies for MDR P. aeruginosa, including phages, vaccines, nanoparticles, and their combinatorial therapies. Novel strategies and their corresponding therapeutic agents are urgently needed for combating MDR pathogens.
Collapse
Affiliation(s)
- Changhong Yin
- Department of Pathology and Laboratory Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Md Zahidul Alam
- Department of Pathology and Laboratory Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - John T Fallon
- Department of Pathology and Laboratory Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Weihua Huang
- Department of Pathology and Laboratory Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
232
|
Mendoza AG, Guercio D, Smiley MK, Sharma GK, Withorn JM, Hudson-Smith NV, Ndukwe C, Dietrich LEP, Boon EM. The histidine kinase NahK regulates pyocyanin production through the PQS system. J Bacteriol 2024; 206:e0027623. [PMID: 38169296 PMCID: PMC10809955 DOI: 10.1128/jb.00276-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
Many bacterial histidine kinases work in two-component systems that combine into larger multi-kinase networks. NahK is one of the kinases in the GacS Multi-Kinase Network (MKN), which is the MKN that controls biofilm regulation in the opportunistic pathogen Pseudomonas aeruginosa. This network has also been associated with regulating many virulence factors P. aeruginosa secretes to cause disease. However, the individual role of each kinase is unknown. In this study, we identify NahK as a novel regulator of the phenazine pyocyanin (PYO). Deletion of nahK leads to a fourfold increase in PYO production, almost exclusively through upregulation of phenazine operon two (phz2). We determined that this upregulation is due to mis-regulation of all P. aeruginosa quorum-sensing (QS) systems, with a large upregulation of the Pseudomonas quinolone signal system and a decrease in production of the acyl-homoserine lactone-producing system, las. In addition, we see differences in expression of quorum-sensing inhibitor proteins that align with these changes. Together, these data contribute to understanding how the GacS MKN modulates QS and virulence and suggest a mechanism for cell density-independent regulation of quorum sensing. IMPORTANCE Pseudomonas aeruginosa is a Gram-negative bacterium that establishes biofilms as part of its pathogenicity. P. aeruginosa infections are associated with nosocomial infections. As the prevalence of multi-drug-resistant P. aeruginosa increases, it is essential to understand underlying virulence molecular mechanisms. Histidine kinase NahK is one of several kinases in P. aeruginosa implicated in biofilm formation and dispersal. Previous work has shown that the nitric oxide sensor, NosP, triggers biofilm dispersal by inhibiting NahK. The data presented here demonstrate that NahK plays additional important roles in the P. aeruginosa lifestyle, including regulating bacterial communication mechanisms such as quorum sensing. These effects have larger implications in infection as they affect toxin production and virulence.
Collapse
Affiliation(s)
- Alicia G. Mendoza
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Danielle Guercio
- Department of Molecular and Cellular Biology, Stony Brook University, Stony Brook, New York, USA
| | - Marina K. Smiley
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Gaurav K. Sharma
- Department of Chemistry, Stony Brook University, Stony Brook, New York, USA
| | - Jason M. Withorn
- Department of Chemistry, Stony Brook University, Stony Brook, New York, USA
| | | | - Chika Ndukwe
- Department of Chemistry, Stony Brook University, Stony Brook, New York, USA
| | - Lars E. P. Dietrich
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Elizabeth M. Boon
- Department of Chemistry, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
233
|
Rafiei S, Bouzari M. Genomic analysis of vB_PaS-HSN4 bacteriophage and its antibacterial activity (in vivo and in vitro) against Pseudomonas aeruginosa isolated from burn. Sci Rep 2024; 14:2007. [PMID: 38263187 PMCID: PMC10805781 DOI: 10.1038/s41598-023-50916-5] [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: 06/07/2023] [Accepted: 12/28/2023] [Indexed: 01/25/2024] Open
Abstract
The most frequent infections caused by Pseudomonas aeruginosa are local infections in soft tissues, including burns. Today, phage use is considered a suitable alternative to cure infections caused by multi-drug-resistant (MDR) and extensively drug-resistant (XDR) bacteria. We investigated the potential of a novel phage (vB_PaS-HSN4) belonging to Caudoviricetes class, against XDR and MDR P. aeruginosa strains in vivo and in vitro. Its biological and genetic characteristics were investigated. The phage burst size and latent were 119 and 20 min, respectively. It could tolerate a broad range of salt concentrations, pH values, and temperatures. The combination with ciprofloxacin significantly enhanced biofilm removal after 24 h. The genome was dsDNA with a size of 44,534 bp and encoded 61 ORFs with 3 tRNA and 5 promoters. No virulence factor was observed in the phage genome. In the in vivo infection model, treatment with vB_PaS-HSN4 increased Galleria mellonella larvae survival (80%, 66%, and 60%) (MOI 100) and (60%, 40%, and 26%) (MOI 1) in the pre-treatment, co-treatment, and post-treatment experiments, respectively. Based on these characteristics, it can be considered for the cure of infections of burns caused by P. aeruginosa.
Collapse
Affiliation(s)
- Solmaz Rafiei
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar-Jereeb Street, Isfahan, 81746-73441, Iran
| | - Majid Bouzari
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar-Jereeb Street, Isfahan, 81746-73441, Iran.
| |
Collapse
|
234
|
Mirpour M, Zahmatkesh H. Ketoprofen attenuates Las/Rhl quorum-sensing (QS) systems of Pseudomonas aeruginosa: molecular and docking studies. Mol Biol Rep 2024; 51:133. [PMID: 38236445 DOI: 10.1007/s11033-023-09071-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/02/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Quorum sensing (QS) is the leading cause of persistent infections and recalcitrance to antibiotic treatment of Pseudomonas aeruginosa. Hence, QS inhibitors are promising agents for the potential treatment of P. aeruginosa infections. METHODS AND RESULTS Herein, the reducing effect of ketoprofen on virulence factors production including protease, hemolysin, pyocyanin, hydrogen cyanide, biofilm, and motility of P. aeruginosa strains was investigated. Furthermore, the quorum quenching activity of ketoprofen at the molecular level was examined by real-time PCR assessment. Our results showed that ketoprofen significantly attenuates virulence factors and biofilm formation in P. aeruginosa strains. Moreover, ketoprofen down-regulated the expression of lasI, lasR, rhlI, and rhlR genes, by 35-47, 22-48, 34-67, and 43-56%, respectively. As well, molecular docking simulation showed a high binding affinity of ketoprofen with QS regulatory proteins. CONCLUSIONS Consequently, this study confirmed the quorum quenching activity of ketoprofen, which could be employed as a useful agent for the treatment of P. aeruginosa infections.
Collapse
Affiliation(s)
- Mirsasan Mirpour
- Department of Microbiology, Lahijan Branch, Islamic Azad University, Lahijan, Iran.
| | - Hossein Zahmatkesh
- Department of Microbiology, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| |
Collapse
|
235
|
Dey D, Kumar A. Unveiling the potential of recently FDA-approved drugs as quorum sensing inhibitors against P. Aeruginosa using high-performance computational techniques. J Biomol Struct Dyn 2024:1-18. [PMID: 38230441 DOI: 10.1080/07391102.2024.2304682] [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: 11/01/2023] [Accepted: 01/07/2024] [Indexed: 01/18/2024]
Abstract
Through cell-to-cell communication, activation of efflux pumps, formation of biofilms, and other mechanisms, pseudomonas aeruginosa's quorum sensing systems (QSS), notably the lasl/las-r system, contribute a vital role in the development of anti-microbial resistance (AMR). Identifying potential drugs against these targets could have significant implications for combating pseudomonal infections. The current study aims to identify promising recently FDA-approved drugs against lasl/las-r proteins. The ligands were selected from the FDA-approved drug lists of the last 5 years. Out of 202, 78 drugs were checked for interaction with lasl/las-r protein and 4 drugs revealed top binding conformations characterized by favorable energetic profiles within the active site of the las-r protein which were further assigned for 250-ns molecular dynamics (MD) simulation. The MD analysis confirmed the dynamical stability of brexanolone and oteseconazole with las-r protein. The root mean square deviation (RMSD), radius of gyration (Rg) and solvent-accessible surface area (SASA) analysis have indicated less deviation, more compactness of protein and less exposure of protein ligand complex to its surroundings as compared to the reference ligand-protein complex. The hydroxyl group in the oteseconazole whereas hydroxyl and ketone group in the brexanolone were responsible for hydrogen bonds with the active site residue of las r ptotein as indicated by ligand-protein contacts diagram. The binding energies per residue analysis revealed TYR-47 as the most contributing amino acid residue for interaction with oteseconazole and brexanolone. The identified drugs may be potential repurposing candidates against pseudomonal infections through inhibition of las-r protein.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Debanjan Dey
- Department of Pharmacology, Delhi Pharmaceutical Science, Research University (DPSRU), New Delhi, India
| | - Anoop Kumar
- Department of Pharmacology, Delhi Pharmaceutical Science, Research University (DPSRU), New Delhi, India
| |
Collapse
|
236
|
Gutiérrez-Santana JC, Coria-Jiménez VR. Diagnosis and Therapeutic Strategies Based on Nucleic Acid Aptamers Selected against Pseudomonas aeruginosa: The Challenge of Cystic Fibrosis. ChemMedChem 2024; 19:e202300544. [PMID: 38016927 DOI: 10.1002/cmdc.202300544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 11/30/2023]
Abstract
Antimicrobial resistance (AMR) is a rapidly spreading global health problem, and approximately five million deaths associated with AMR pathogens were identified prior to the COVID-19 pandemic. Pseudomonas aeruginosa has developed increasing AMR, and in patients with cystic fibrosis (CF) colonized by this bacterium, rare phenotypes have emerged that complicate the diagnosis and treatment of the hosts, in addition to multiple associated "epidemic strains" with high morbidities and mortalities. The conjugation of aptamers with fluorochromes or nanostructures has allowed the design of new identification strategies for Pseudomonas aeruginosa with detection limits of up to 1 cell ⋅ mL-1 , and the synergy of aptamers with antibiotics, antimicrobial peptides and nanostructures has exhibited promising therapeutic qualities. Some selected aptamers against this bacterium have shown intrinsic antimicrobial activity. However, these aptamers have been poorly evaluated in clinical isolates and have shown decreased interactions for CF isolates, demonstrating, in these cases, uncommon phenotypes resulting from the selective qualities of this disease as well as the great adaptive capacity of the pathogen. Therefore, finding an aptamer or set of aptamers that have the ability to recognize strange phenotypes of this bacillus is crucial in the battle against AMR.
Collapse
Affiliation(s)
- Juan Carlos Gutiérrez-Santana
- Laboratorio de Bacteriología Experimental, Instituto Nacional de Pediatría, Insurgentes sur 3700-C, Col. Insurgentes Cuicuilco Coyoacán, 04530, Ciudad de México, México
| | - Victor Rafael Coria-Jiménez
- Laboratorio de Bacteriología Experimental, Instituto Nacional de Pediatría, Insurgentes sur 3700-C, Col. Insurgentes Cuicuilco Coyoacán, 04530, Ciudad de México, México
| |
Collapse
|
237
|
Wu T, Zhang Z, Li T, Dong X, Wu D, Zhu L, Xu K, Zhang Y. The type III secretion system facilitates systemic infections of Pseudomonas aeruginosa in the clinic. Microbiol Spectr 2024; 12:e0222423. [PMID: 38088541 PMCID: PMC10783026 DOI: 10.1128/spectrum.02224-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 11/14/2023] [Indexed: 01/13/2024] Open
Abstract
IMPORTANCE The identification of decisive virulence-associated genes in highly pathogenic P. aeruginosa isolates in the clinic is essential for diagnosis and the start of appropriate treatment. Over the past decades, P. aeruginosa ST463 has spread rapidly in East China and is highly resistant to β-lactams. Given the poor clinical outcome caused by this phenotype, detailed information regarding its decisive virulence genes and factors affecting virulence expression needs to be deciphered. Here, we demonstrate that the T3SS effector ExoU has toxic effects on mammalian cells and is required for virulence in the murine bloodstream infection model. Moreover, a functional downstream SpcU is required for ExoU secretion and cytotoxicity. This work highlights the potential role of ExoU in the pathogenesis of disease and provides a new perspective for further research on the development of new antimicrobials with antivirulence ability.
Collapse
Affiliation(s)
- Tiantian Wu
- Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenchuan Zhang
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, China
| | - Tong Li
- Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xu Dong
- Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Wu
- Research and Service Center, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- College of Food and Health, Zhejiang A&F University, Lin'an, Hangzhou, China
| | - Lixia Zhu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kaijin Xu
- Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Zhang
- Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| |
Collapse
|
238
|
Pool-Yam L, Ramón-Sierra J, Oliva AI, Zamora-Bustillos R, Ortiz-Vázquez E. Effect of conA-unbound proteins from Melipona beecheii honey on the formation of Pseudomonas aeruginosa ATCC 27853 biofilm. Arch Microbiol 2024; 206:54. [PMID: 38180520 DOI: 10.1007/s00203-023-03783-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/20/2023] [Accepted: 12/01/2023] [Indexed: 01/06/2024]
Abstract
Pseudomonas aeruginosa is an opportunistic bacterium that can form a biofilm with the ability to colonize different surfaces and for increasing resistance to antibiotics. An alternative to solve this problem may be the use of non-glucose/mannose glycosylated proteins from Melipona beecheii honey, which are capable of inhibiting the growth of this pathogen. In this work, the antibiofilm activity of the conA-unbound protein fraction (F1) from M. beecheii was evaluated. The crude protein extract (CPE) and the F1 fraction inhibited the P. aeruginosa biofilm growth above 80% at 4 and 1.3 µg/mL, respectively. These proteins affected the structure of the biofilm, as well as fleQ and fleR gene expressions involved in the formation and regulation of the P. aeruginosa biofilm. The results demonstrated that the F1 fraction proteins of M. beecheii honey inhibit and affect the formation of the P. aeruginosa biofilm.
Collapse
Affiliation(s)
- Luis Pool-Yam
- División de Estudios de Posgrado E Investigación, Instituto Tecnológico de Conkal, Avenida Tecnológico S/N Conkal, C.P. 97345, Conkal, Yucatán, México
| | - Jesús Ramón-Sierra
- División de Estudios de Posgrado E Investigación, Instituto Tecnológico de Mérida, Av. Tecnológico Km. 4.5 S/N, C.P. 97118, Mérida, Yucatán, México
| | - A I Oliva
- Departamento de Física Aplicada, CINVESTAV-IPN, Unidad Mérida, Carretera Antigua a Progreso Km. 6, Cordemex, C.P. 97310, Mérida, Yucatán, México
| | - Roberto Zamora-Bustillos
- División de Estudios de Posgrado E Investigación, Instituto Tecnológico de Conkal, Avenida Tecnológico S/N Conkal, C.P. 97345, Conkal, Yucatán, México.
| | - Elizabeth Ortiz-Vázquez
- División de Estudios de Posgrado E Investigación, Instituto Tecnológico de Mérida, Av. Tecnológico Km. 4.5 S/N, C.P. 97118, Mérida, Yucatán, México.
| |
Collapse
|
239
|
Sadik A, Viswaswar JP, Rajamoney A, Rekha A, Raj DM, Prakashan D, Vasudevan M, Visakh JS, Renuka D, Hely S, Shaji SK, Chandran PR, Kumar G, Vijayan Nair S, Haripriyan J. Mitigation of quorum sensing mediated virulence factors of Pseudomonas aeruginosa: the role of Meldrum's acid activated furan. Front Microbiol 2024; 14:1272240. [PMID: 38235424 PMCID: PMC10791761 DOI: 10.3389/fmicb.2023.1272240] [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: 08/03/2023] [Accepted: 10/05/2023] [Indexed: 01/19/2024] Open
Abstract
The rapid emergence of drug resistant pathogens is a major threat which has warranted the development of alternative strategies to combat infectious diseases. In this work, we have tested the anti-virulent activity of Meldrum's acid activated furan (MAF) and 1,3-dimethyl barbituric acid activated furan (BAF) against Chromobacterium violaceum and Pseudomonas aeruginosa. It was found that MAF significantly reduced the violacein production and biofilm formation of C. violaceum at sub-inhibitory concentrations. The quorum sensing (QS) regulated virulence factors of P. aeruginosa including biofilm formation, motility, pigment production, and elastase activity were also found to be reduced considerably at sub-inhibitory concentrations of MAF. Additionally, MAF downregulated the expression of genes in the QS circuitry of P. aeruginosa, demonstrating the potential of MAF in lowering the pathogenicity of P. aeruginosa. In silico studies demonstrated the potential of MAF to compete with the signaling molecules of C. violaceum and P. aeruginosa for the QS receptor interaction. In vivo studies using Caenorhabditis elegans demonstrated the anti-pathogenicity of MAF by enhancing the survival of P. aeruginosa-infected C. elegans. These results suggest that activated furan compounds could be potential inhibitors of QS-mediated virulence factors in C. violaceum and P. aeruginosa, encouraging their use in combating multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Ajmal Sadik
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Jithin P. Viswaswar
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Ambili Rajamoney
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Anjali Rekha
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Darsana M. Raj
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Deepthi Prakashan
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Mydhili Vasudevan
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - J. S. Visakh
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Dhannya Renuka
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Sreetha Hely
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | | | - Prakash R. Chandran
- Department of Chemistry, Mannam Memorial N.S.S. College, Kottiyam, Kerala, India
| | - Geetha Kumar
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Sobha Vijayan Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | | |
Collapse
|
240
|
KavianFar A, Taherkhani H, Ahmadi A, Salimi M, Lanjanian H, Masoudi-Nejad A. Restoring the epigenetic landscape of lung microbiome: potential therapeutic approach for chronic respiratory diseases. BMC Pulm Med 2024; 24:2. [PMID: 38166878 PMCID: PMC10759706 DOI: 10.1186/s12890-023-02789-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Chronic respiratory diseases, such as chronic obstructive pulmonary disease (COPD) and bronchiectasis, present significant threats to global health. Recent studies have revealed the crucial role of the lung microbiome in the development of these diseases. Pathogens have evolved complex strategies to evade the immune response, with the manipulation of host cellular epigenetic mechanisms playing a pivotal role. There is existing evidence regarding the effects of Pseudomonas on epigenetic modifications and their association with pulmonary diseases. Therefore, this study aims to directly assess the connection between Pseudomonas abundance and chronic respiratory diseases. We hope that our findings will shed light on the molecular mechanisms behind lung pathogen infections. METHODS We analyzed data from 366 participants, including individuals with COPD, acute exacerbations of COPD (AECOPD), bronchiectasis, and healthy individuals. Previous studies have given limited attention to the impact of Pseudomonas on these groups and their comparison with healthy individuals. Two independent datasets from different ethnic backgrounds were used for external validation. Each dataset separately analyzed bacteria at the genus level. RESULTS The study reveals that Pseudomonas, a bacterium, was consistently found in high concentrations in all chronic lung disease datasets but it was present in very low abundance in the healthy datasets. This suggests that Pseudomonas may influence cellular mechanisms through epigenetics, contributing to the development and progression of chronic respiratory diseases. CONCLUSIONS This study emphasizes the importance of understanding the relationship between the lung microbiome, epigenetics, and the onset of chronic pulmonary disease. Enhanced recognition of molecular mechanisms and the impact of the microbiome on cellular functions, along with a better understanding of these concepts, can lead to improved diagnosis and treatment.
Collapse
Affiliation(s)
- Azadeh KavianFar
- Laboratory of Systems Biology and Bioinformatics (LBB), Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran
| | - Hamidreza Taherkhani
- Laboratory of Systems Biology and Bioinformatics (LBB), Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran
| | - Ali Ahmadi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Tehran, Iran.
| | - Mahdieh Salimi
- Department of Medical Genetics, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Hossein Lanjanian
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran.
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
241
|
Wells M, Mikesh M, Gordon V. Structure-preserving fixation allows scanning electron microscopy to reveal biofilm microstructure and interactions with immune cells. J Microsc 2024; 293:59-68. [PMID: 38098170 PMCID: PMC10764082 DOI: 10.1111/jmi.13252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
Pseudomonas aeruginosa is a pathogen that forms robust biofilms which are commonly associated with chronic infections and cannot be successfully cleared by the immune system. Neutrophils, the most common white blood cells, target infections with pathogen-killing mechanisms that are rendered largely ineffective by the protective physicochemical structure of a biofilm. Visualisation of the complex interactions between immune cells and biofilms will advance understanding of how biofilms evade the immune system and could aid in developing treatment methods that promote immune clearance with minimal harm to the host. Scanning electron microscopy (SEM) distinguishes itself as a powerful, high-resolution tool for obtaining strikingly clear and detailed topographical images. However, taking full advantage of SEM's potential for high-resolution imaging requires that the fixation process simultaneously preserve both intricate biofilm architecture and the morphologies and structural signatures characterising neutrophils responses at an infection site. Standard aldehyde-based fixation techniques result in significant loss of biofilm matrix material and morphologies of responding immune cells, thereby obscuring the details of immune interactions with the biofilm matrix. Here we show an improved fixation technique using the cationic dye alcian blue to preserve and visualise neutrophil interactions with the three-dimensional architecture of P. aeruginosa biofilms. We also demonstrate that this technique better preserves structures of biofilms grown from two other bacterial species, Klebsiella pneumoniae and Burkholderia thailandensis.
Collapse
Affiliation(s)
- Marilyn Wells
- Department of Physics, The University of Texas at Austin, Austin, Texas, USA
- Center for Nonlinear Dynamics, The University of Texas at Austin, Austin, Texas, USA
| | - Michelle Mikesh
- Center for Biomedical Research Support, The University of Texas at Austin, Austin, Texas, USA
| | - Vernita Gordon
- Department of Physics, The University of Texas at Austin, Austin, Texas, USA
- Center for Nonlinear Dynamics, The University of Texas at Austin, Austin, Texas, USA
- Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, Texas, USA
- LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
242
|
Alasiri A, Soltane R, Taha MN, Abd El-Aleam RH, Alshehri F, Sayed AM. Bakuchiol inhibits Pseudomonas aeruginosa's quorum sensing-dependent biofilm formation by selectively inhibiting its transcriptional activator protein LasR. Int J Biol Macromol 2024; 255:128025. [PMID: 37979739 DOI: 10.1016/j.ijbiomac.2023.128025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/26/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023]
Abstract
In the present study, we characterized Bakuchiol (Bak) as a new potent quorum sensing (QS) inhibitor against Pseudomonas aeruginosa biofilm formation. Upon extensive in vitro investigations, Bak was found to suppress the P. aeruginosa biofilm formation (75.5 % inhibition) and its associated virulence factor e.g., pyocyanin and rhamnolipids (% of inhibition = 71.5 % and 66.9 %, respectively). Upon LuxR-type receptors assay, Bak was found to selectively inhibit P. aeruginosa's LasR in a dose-dependent manner. Further in-depth molecular investigations (e.g., sedimentation velocity and thermal shift assays) revealed that Bak destabilized LasR upon binding and disrupted its functioning quaternary structure (i.e., the functioning dimeric form). The subsequent modeling and molecular dynamics (MD) simulations explained in more molecular detail how Bak interacts with LasR and how it can induce its dimeric form disruption. In conclusion, our study identified Bak as a potent and specific LasR antagonist that should be widely used as a chemical probe of QS in P. aeruginosa, offering new insights into LasR antagonism processes. The new findings shed light on the cryptic world of LuxR-type QS in this important opportunistic pathogen.
Collapse
Affiliation(s)
- Ahlam Alasiri
- Department of Biology, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia.
| | - Raya Soltane
- Department of Biology, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia.
| | - Mostafa N Taha
- Microbiology and Immunology Department, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt.
| | - Rehab H Abd El-Aleam
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information MTI, Cairo 11571, Egypt.
| | - Fatma Alshehri
- Department of Biology, College of Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Ahmed M Sayed
- Pharmacognosy Department, Faculty of Pharmacy, Nahda University, Beni Suef, Egypt; Department of Pharmacognosy, College of Pharmacy, Almaaqal University, 61014 Basra, Iraq.
| |
Collapse
|
243
|
Costa DCS, Froes TQ, Mendes MS, da S M Forezi L, Ferreira VF, Castilho MS, de C da Silva F. 1 H-1,2,3-triazol-1,4-naphthoquinone Derivatives: Novel Inhibitors Targeting Pyocyanin Biosynthesis for P. Aeruginosa Infection Treatment Advances. Curr Top Med Chem 2024; 24:2161-2171. [PMID: 39136508 DOI: 10.2174/0115680266327024240726111230] [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: 05/22/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND This study investigates the potential of eleven 1H-1,2,3-triazol-1,4-naphthoquinone conjugates as virulence factor inhibitors (like Pyocyanin) and their affinity for PhzM, a crucial enzyme for Pyocyanin biosynthesis in Pseudomonas aeruginosa infections. METHODS A straightforward synthetic pathway enabled the production of these compounds, which were characterized and structurally confirmed through spectroscopic analyses. Evaluation of their impact on PhzM thermal stability identified promising candidates for PhzM binders. RESULTS Concentration-response behavior elucidated their binding affinity, revealing them as the first reported micromolar affinity ligands for PhzM. Structure-activity relationship analysis emphasized the role of specific molecular moieties in binding affinity modulation, paving the way for future advanced inhibitors' development. CONCLUSION These findings highlight the potential of naphthoquinone-triazole derivatives as leads for novel therapeutics against P. aeruginosa infections.
Collapse
Affiliation(s)
- Dora C S Costa
- Departamento de Química Orgânica, Universidade Federal Fluminense, Instituto de Química, Niterói, RJ, 24020, 150, Brazil
| | - Thamires Q Froes
- Programa de pós-graduação em Biotecnologia da Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil
- Faculdade de Farmácia da Universidade Federal da Bahia, Bahia, Salvador, BA, Brazil
| | - Marina S Mendes
- Faculdade de Farmácia da Universidade Federal da Bahia, Bahia, Salvador, BA, Brazil
| | - Luana da S M Forezi
- Departamento de Química Orgânica, Universidade Federal Fluminense, Instituto de Química, Niterói, RJ, 24020, 150, Brazil
| | - Vitor F Ferreira
- Universidade Federal Fluminense, Departamento de Tecnologia Farmacêutica, Faculdade de Farmácia, Niterói, RJ, 24241, 002, Brazil
| | - Marcelo S Castilho
- Programa de pós-graduação em Biotecnologia da Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil
- Faculdade de Farmácia da Universidade Federal da Bahia, Bahia, Salvador, BA, Brazil
| | - Fernando de C da Silva
- Departamento de Química Orgânica, Universidade Federal Fluminense, Instituto de Química, Niterói, RJ, 24020, 150, Brazil
| |
Collapse
|
244
|
Chadha J, Khullar L, Gulati P, Chhibber S, Harjai K. Repurposing albendazole as a potent inhibitor of quorum sensing-regulated virulence factors in Pseudomonas aeruginosa: Novel prospects of a classical drug. Microb Pathog 2024; 186:106468. [PMID: 38036112 DOI: 10.1016/j.micpath.2023.106468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/02/2023]
Abstract
Pseudomonas aeruginosa has emerged as a critical superbug that poses a serious threat to public health. Owing to its virulence and multidrug resistance profiles, the pathogen demands immediate attention for devising alternate intervention strategies. In an attempt to repurpose drugs against P. aeruginosa, this preclinical study was aimed at investigating the antivirulence prospects of albendazole (AbZ), an FDA-approved anti-helminthic drug, recently predicted to disrupt quorum sensing (QS) in Chromobacterium violaceum. AbZ was scrutinized for its quorum quenching (QQ) prospects, effect on bacterial virulence, different motility phenotypes, and biofilm formation in vitro. Additionally, in silico analysis was employed to predict the molecular interactions between AbZ and QS receptors. At sub-inhibitory levels, AbZ demonstrated anti-QS activity and significantly abrogated AHL biosynthesis in P. aeruginosa. Moreover, AbZ significantly downregulated the transcript levels of QS- (lasI/lasR, rhlI/rhlR, and pqsA/pqsR) and QS-dependent virulence (aprA, lasA, lasB, plcH, and toxA) genes in P. aeruginosa. This coincided with reduced hemolysin, alginate, pyocyanin, rhamnolipids, total protease, and elastase production, thereby lowering phenotypic virulence. Molecular docking with AbZ further revealed strong associations and high binding energies with LasR (-8.8 kcal/mol), RhlR (-6.5 kcal/mol), and PqsR (-6.3 kcal/mol) receptors. AbZ also impeded bacterial motility and abolished EPS production, severely compromising pseudomonal biofilm formation. For the first time, AbZ was shown to interfere with QS circuitry and consequently disarming pseudomonal virulence. Hence, AbZ can be exploited for its antivirulence properties against P. aeruginosa.
Collapse
Affiliation(s)
- Jatin Chadha
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Lavanya Khullar
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Pallavi Gulati
- RLA College, University of Delhi (South Campus), New Delhi, India
| | - Sanjay Chhibber
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Kusum Harjai
- Department of Microbiology, Panjab University, Chandigarh, India.
| |
Collapse
|
245
|
Maha Swetha BR, Saravanan M, Piruthivraj P. Emerging trends in the inhibition of bacterial molecular communication: An overview. Microb Pathog 2024; 186:106495. [PMID: 38070626 DOI: 10.1016/j.micpath.2023.106495] [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: 08/04/2023] [Revised: 12/03/2023] [Accepted: 12/04/2023] [Indexed: 01/02/2024]
Abstract
Quorum sensing (QS) is a molecular cell-cell communication utilized by several bacteria and some fungi. It involves cell density dependent gene expression that includes extra polymeric substance production, sporulation, antibiotic production, motility, competence, symbiosis and conjugation. These expressions were carried out by different signaling molecules like acyl homo-serine lactone (AHL) and auto-inducing peptides (AIPs) which was effluxed by gram negative and gram positive bacteria. Pathogenic bacteria and biofilms often exhibit high resistance to antibiotics, attributed to the presence of antibiotic efflux pumps, reduced membrane permeability, and enzymes that deactivate quorum sensing (QS) inhibitors. To counteract virulence and multi-drug resistance (MDR), novel strategies such as employing quorum sensing (QS) inhibitors and quorum quenchers are employed. It targets signaling molecules with synthesis and prevents the signal from binding to receptors. In this present review, the mechanisms of QS along with inhibitors from different sources are described. These strategies potentially interfere with QS and it can be applied in different fields, mainly in hospitals and marine environments where the pathogenic infections and biofilm formation are highly involved.
Collapse
Affiliation(s)
- B R Maha Swetha
- Department of Biotechnoloy, Srimad Andavan Arts and Science College (Autonomous), Tiruchirappalli, Tamil Nadu, India
| | - M Saravanan
- Department of Physics, University College of Engineering, Bharathidasan Institute of Technology Campus, Anna University, Tiruchirapalli, 620 024, Tamil Nadu, India
| | - Prakash Piruthivraj
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha Univerisy, Chennai, 600 077, Tamil Nadu, India; Department of Biotechnoloy, Srimad Andavan Arts and Science College (Autonomous), Tiruchirappalli, Tamil Nadu, India.
| |
Collapse
|
246
|
Vadakkan K, Ngangbam AK, Sathishkumar K, Rumjit NP, Cheruvathur MK. A review of chemical signaling pathways in the quorum sensing circuit of Pseudomonas aeruginosa. Int J Biol Macromol 2024; 254:127861. [PMID: 37939761 DOI: 10.1016/j.ijbiomac.2023.127861] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/10/2023]
Abstract
Pseudomonas aeruginosa, an increasingly common competitive and biofilm organism in healthcare infection with sophisticated, interlinked and hierarchic quorum systems (Las, Rhl, PQS, and IQS), creates the greatest threats to the medical industry and has rendered prevailing chemotherapy medications ineffective. The rise of multidrug resistance has evolved into a concerning and potentially fatal occurrence for human life. P. aeruginosa biofilm development is assisted by exopolysaccharides, extracellular DNA, proteins, macromolecules, cellular signaling and interaction. Quorum sensing is a communication process between cells that involves autonomous inducers and regulators. Quorum-induced infectious agent biofilms and the synthesis of virulence factors have increased disease transmission, medication resistance, infection episodes, hospitalizations and mortality. Hence, quorum sensing may be a potential therapeutical target for bacterial illness, and developing quorum inhibitors as an anti-virulent tool could be a promising treatment strategy for existing antibiotics. Quorum quenching is a prevalent technique for treating infections caused by microbes because it diminishes microbial pathogenesis and increases microbe biofilm sensitivity to antibiotics, making it a potential candidate for drug development. This paper examines P. aeruginosa quorum sensing, the hierarchy of quorum sensing mechanism, quorum sensing inhibition and quorum sensing inhibitory agents as a drug development strategy to supplement traditional antibiotic strategies.
Collapse
Affiliation(s)
- Kayeen Vadakkan
- Department of Biology, St. Mary's College, Thrissur, Kerala 680020, India; Manipur International University, Imphal, Manipur 795140, India.
| | | | - Kuppusamy Sathishkumar
- Rhizosphere Biology Laboratory, Department of Microbiology, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India; Department of Computational Biology, Institute of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai 602 105, Tamil Nadu, India
| | | | | |
Collapse
|
247
|
Alhajj N, Yahya MFZR, O'Reilly NJ, Cathcart H. Development and characterization of a spray-dried inhalable ternary combination for the treatment of Pseudomonas aeruginosa biofilm infection in cystic fibrosis. Eur J Pharm Sci 2024; 192:106654. [PMID: 38013123 DOI: 10.1016/j.ejps.2023.106654] [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: 06/04/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023]
Abstract
Cystic fibrosis (CF) is an inherited lung disease characterised by the accumulation of thick layers of dried mucus in the lungs which serve as a nidus for chronic infection. Pseudomonas aeruginosa is the predominant cause of chronic lung infection in cystic fibrosis. The dense mucus coupled with biofilm formation hinder antibiotic penetration and prevent them from reaching their target. Mucoactive agents are recommended in the treatment of CF in combination with antibiotics. In spite of the extensive research in developing novel drug combinations for the treatment of lung infection in CF, to our knowledge, there is no study that combines antibiotic, antibiofilm and mucoactive agent in a single inhaled dry powder formulation. In the present study, we investigate the possibility of adding a mucoactive agent to our previously developed ciprofloxacinquercetin (antibiotic-antibiofilm) dry powder for inhalation. Three mucoactive agents, namely mannitol (MAN), N-acetyl-L-cysteine (NAC) and ambroxol hydrochloride (AMB), were investigated for this purpose. The ternary combinations were prepared via spray drying without the addition of excipients. All ternary combinations conserved or improved the antibacterial and biofilm inhibition activities of ciprofloxacin against P. aeruginosa (ATCC 10145). The addition of AMB resulted in an amorphous ternary combination (SD-CQA) with superior physical stability as indicated by DSC and nonambient XRPD. Furthermore, SD-CQA displayed better in vitro aerosolization performance (ED ∼ 71 %; FPF ∼ 49 %) compared to formulations containing MAN and NAC (ED ∼ 64 % and 44 %; FPF ∼ 44 % and 29 %, respectively). In conclusion, a ternary drug combination powder with suitable aerosolization, physical stability and antibacterial/antibiofilm properties was prepared by a single spray drying step.
Collapse
Affiliation(s)
- Nasser Alhajj
- Pharmaceutical and Molecular Biotechnology Research Centre (PMBRC), South East Technological University (SETU), Main Campus, Cork Road, Waterford X91 K0EK, Ireland.
| | | | - Niall J O'Reilly
- Pharmaceutical and Molecular Biotechnology Research Centre (PMBRC), South East Technological University (SETU), Main Campus, Cork Road, Waterford X91 K0EK, Ireland; SSPC - The Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland
| | - Helen Cathcart
- Pharmaceutical and Molecular Biotechnology Research Centre (PMBRC), South East Technological University (SETU), Main Campus, Cork Road, Waterford X91 K0EK, Ireland
| |
Collapse
|
248
|
Choi SY, Romero-Calle DX, Cho HG, Bae HW, Cho YH. Use of Cas9 Targeting and Red Recombination for Designer Phage Engineering. J Microbiol 2024; 62:1-10. [PMID: 38300409 DOI: 10.1007/s12275-024-00107-2] [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: 11/21/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
Bacteriophages (phages) are natural antibiotics and biological nanoparticles, whose application is significantly boosted by recent advances of synthetic biology tools. Designer phages are synthetic phages created by genome engineering in a way to increase the benefits or decrease the drawbacks of natural phages. Here we report the development of a straightforward genome engineering method to efficiently obtain engineered phages in a model bacterial pathogen, Pseudomonas aeruginosa. This was achieved by eliminating the wild type phages based on the Streptococcus pyogenes Cas9 (SpCas9) and facilitating the recombinant generation based on the Red recombination system of the coliphage λ (λRed). The producer (PD) cells of P. aeruginosa strain PAO1 was created by miniTn7-based chromosomal integration of the genes for SpCas9 and λRed under an inducible promoter. To validate the efficiency of the recombinant generation, we created the fluorescent phages from a temperate phage MP29. A plasmid bearing the single guide RNA (sgRNA) gene for selectively targeting the wild type gp35 gene and the editing template for tagging the Gp35 with superfolder green fluorescent protein (sfGFP) was introduced into the PD cells by electroporation. We found that the targeting efficiency was affected by the position and number of sgRNA. The fluorescent phage particles were efficiently recovered from the culture of the PD cells expressing dual sgRNA molecules. This protocol can be used to create designer phages in P. aeruginosa for both application and research purposes.
Collapse
Affiliation(s)
- Shin-Yae Choi
- Program of Biopharmaceutical Science and Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Gyeonggi, 13488, Republic of Korea
| | - Danitza Xiomara Romero-Calle
- Program of Biopharmaceutical Science and Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Gyeonggi, 13488, Republic of Korea
| | - Han-Gyu Cho
- Program of Biopharmaceutical Science and Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Gyeonggi, 13488, Republic of Korea
| | - Hee-Won Bae
- Program of Biopharmaceutical Science and Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Gyeonggi, 13488, Republic of Korea
| | - You-Hee Cho
- Program of Biopharmaceutical Science and Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Gyeonggi, 13488, Republic of Korea.
| |
Collapse
|
249
|
Song Y, Zhang S, Zhao N, Nong C, He Y, Bao R. Pseudomonas aeruginosa two-component system CprRS regulates HigBA expression and bacterial cytotoxicity in response to LL-37 stress. PLoS Pathog 2024; 20:e1011946. [PMID: 38198506 PMCID: PMC10805311 DOI: 10.1371/journal.ppat.1011946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 01/23/2024] [Accepted: 01/04/2024] [Indexed: 01/12/2024] Open
Abstract
Pseudomonas aeruginosa is a highly pathogenic bacterium known for its ability to sense and coordinate the production of virulence factors in response to host immune responses. However, the regulatory mechanisms underlying this process have remained largely elusive. In this study, we investigate the two-component system CprRS in P. aeruginosa and unveil the crucial role of the sensor protein CprS in sensing the human host defense peptide LL-37, thereby modulating bacterial virulence. We demonstrate that CprS acts as a phosphatase in the presence of LL-37, leading to the phosphorylation and activation of the response regulator CprR. The results prove that CprR directly recognizes a specific sequence within the promoter region of the HigBA toxin-antitoxin system, resulting in enhanced expression of the toxin HigB. Importantly, LL-37-induced HigB expression promotes the production of type III secretion system effectors, leading to reduced expression of proinflammatory cytokines and increased cytotoxicity towards macrophages. Moreover, mutations in cprS or cprR significantly impair bacterial survival in both macrophage and insect infection models. This study uncovers the regulatory mechanism of the CprRS system, enabling P. aeruginosa to detect and respond to human innate immune responses while maintaining a balanced virulence gene expression profile. Additionally, this study provides new evidence and insights into the complex regulatory system of T3SS in P. aeruginosa within the host environment, contributing to a better understanding of host-microbe communication and the development of novel strategies to combat bacterial infections.
Collapse
Affiliation(s)
- Yingjie Song
- College of Life Science, Sichuan Normal University, Chengdu, China
| | - Siping Zhang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Ninglin Zhao
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Nong
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yongxing He
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Rui Bao
- Center of Infectious Diseases, Division of Infectious Diseases in State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
250
|
Alexeeva OV, Olkhov AA, Konstantinova ML, Podmasterev VV, Petrova TV, Martirosyan LY, Karyagina OK, Kozlov SS, Lomakin SM, Tretyakov IV, Siracusa V, Iordanskii AL. A Novel Approach for Glycero-(9,10-trioxolane)-Trialeate Incorporation into Poly(lactic acid)/Poly(ɛ-caprolactone) Blends for Biomedicine and Packaging. Polymers (Basel) 2023; 16:128. [PMID: 38201793 PMCID: PMC10780447 DOI: 10.3390/polym16010128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
The product of ozonolysis, glycero-(9,10-trioxolane)-trioleate (ozonide of oleic acid triglyceride, [OTOA]), was incorporated into polylactic acid/polycaprolactone (PLA/PCL) blend films in the amount of 1, 5, 10, 20, 30 and 40% w/w. The morphological, mechanical, thermal and antibacterial properties of the biodegradable PLA/PCL films after the OTOA addition were studied. According to DSC and XRD data, the degree of crystallinity of the PLA/PCL + OTOA films showed a general decreasing trend with an increase in OTOA content. Thus, a significant decrease from 34.0% for the reference PLA/PCL film to 15.7% for the PLA/PCL + 40% OTOA film was established using DSC. Observed results could be explained by the plasticizing effect of OTOA. On the other hand, the PLA/PCL film with 20% OTOA does not follow this trend, showing an increase in crystallinity both via DSC (20.3%) and XRD (34.6%). OTOA molecules, acting as a plasticizer, reduce the entropic barrier for nuclei formation, leading to large number of PLA spherulites in the plasticized PLA/PCL matrix. In addition, OTOA molecules could decrease the local melt viscosity at the vicinity of the growing lamellae, leading to faster crystal growth. Morphological analysis showed that the structure of the films with an OTOA concentration above 20% drastically changed. Specifically, an interface between the PLA/PCL matrix and OTOA was formed, thereby forming a capsule with the embedded antibacterial agent. The moisture permeability of the resulting PLA/PCL + OTOA films decreased due to the formation of uniformly distributed hydrophobic amorphous zones that prevented water penetration. This architecture affects the tensile characteristics of the films: strength decreases to 5.6 MPa, elastic modulus E by 40%. The behavior of film elasticity is associated with the redistribution of amorphous regions in the matrix. Additionally, PLA/PCL + OTOA films with 20, 30 and 40% of OTOA showed good antibacterial properties on Pseudomonas aeruginosa, Raoultella terrigena (Klebsiella terrigena) and Agrobacterium tumefaciens, making the developed films potentially promising materials for wound-dressing applications.
Collapse
Affiliation(s)
- Olga V. Alexeeva
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (A.A.O.); (M.L.K.); (V.V.P.); (L.Y.M.); (O.K.K.); (S.S.K.); (S.M.L.)
| | - Anatoliy A. Olkhov
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (A.A.O.); (M.L.K.); (V.V.P.); (L.Y.M.); (O.K.K.); (S.S.K.); (S.M.L.)
- N.N. Semenov Federal Research Center for Chemical Physics Russian Academy of Sciences, 119991 Moscow, Russia; (T.V.P.); (I.V.T.); (A.L.I.)
- Academic Department of Innovational Materials and Technologies Chemistry, Plekhanov Russian University of Economics, 117997 Moscow, Russia
| | - Marina L. Konstantinova
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (A.A.O.); (M.L.K.); (V.V.P.); (L.Y.M.); (O.K.K.); (S.S.K.); (S.M.L.)
| | - Vyacheslav V. Podmasterev
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (A.A.O.); (M.L.K.); (V.V.P.); (L.Y.M.); (O.K.K.); (S.S.K.); (S.M.L.)
| | - Tuyara V. Petrova
- N.N. Semenov Federal Research Center for Chemical Physics Russian Academy of Sciences, 119991 Moscow, Russia; (T.V.P.); (I.V.T.); (A.L.I.)
| | - Levon Yu. Martirosyan
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (A.A.O.); (M.L.K.); (V.V.P.); (L.Y.M.); (O.K.K.); (S.S.K.); (S.M.L.)
| | - Olga K. Karyagina
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (A.A.O.); (M.L.K.); (V.V.P.); (L.Y.M.); (O.K.K.); (S.S.K.); (S.M.L.)
| | - Sergey S. Kozlov
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (A.A.O.); (M.L.K.); (V.V.P.); (L.Y.M.); (O.K.K.); (S.S.K.); (S.M.L.)
| | - Sergey M. Lomakin
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119334 Moscow, Russia; (A.A.O.); (M.L.K.); (V.V.P.); (L.Y.M.); (O.K.K.); (S.S.K.); (S.M.L.)
- N.N. Semenov Federal Research Center for Chemical Physics Russian Academy of Sciences, 119991 Moscow, Russia; (T.V.P.); (I.V.T.); (A.L.I.)
| | - Ilya V. Tretyakov
- N.N. Semenov Federal Research Center for Chemical Physics Russian Academy of Sciences, 119991 Moscow, Russia; (T.V.P.); (I.V.T.); (A.L.I.)
| | - Valentina Siracusa
- Department of Chemical Science (DSC), University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Alexey L. Iordanskii
- N.N. Semenov Federal Research Center for Chemical Physics Russian Academy of Sciences, 119991 Moscow, Russia; (T.V.P.); (I.V.T.); (A.L.I.)
| |
Collapse
|