1
|
Jeznach A, Sidor-Dzitkowska K, Bandyszewska M, Grzanka M, Popławski P, Marszalik A, Domagała-Kulawik J, Stachowiak R, Hoser G, Skirecki T. Sepsis-induced inflammasome impairment facilitates development of secondary A. baumannii pneumonia. Emerg Microbes Infect 2025; 14:2492206. [PMID: 40202049 PMCID: PMC12016274 DOI: 10.1080/22221751.2025.2492206] [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: 09/04/2024] [Revised: 03/07/2025] [Accepted: 04/07/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Acinetobacter baumannii has become one of the most critical pathogens causing nosocomial pneumonia. Existing animal models of A. baumannii pneumonia are not relevant to the majority of critical care patients. We aimed to develop a novel model of secondary A. baumannii pneumonia in post-sepsis mice. METHODS A two-hit model of sepsis induced by cecal ligation and puncture followed by A. baumannii pneumonia on day 5 was established. In addition, the two-hit model was established in humanized mice. A period of 2 h of mechanical ventilation followed by observation was used in additional experiments. Lung histopathology, bacterial cultures, and cellular infiltration were analysed as well as markers of the inflammasome activity in vivo and ex vivo. RESULTS A. baumannii infection caused mortality and loss of body weight and temperature in post-sepsis mice. Increased lung bacterial burden and dissemination together with signs of enhanced inflammatory injury were observed in post-sepsis mice but not control mice that were challenged with A. baumannii. Post-sepsis mice were unable to mount inflammasome activation in response to secondary pneumonia to the level of control mice. Transfer of wild-type but not capsase-1 KO alveolar macrophages was able to restore the pulmonary protection against A. baumannii. Mechanical ventilation exacerbated the pathological response to pneumonia in post-sepsis mice but enhanced inflammasome signalling in non-sepsis mice with pneumonia. CONCLUSIONS We established a novel model of A. baumannii pneumonia that revealed sepsis-induced impairment of inflammasome activation in alveolar macrophages is critical for the control of secondary A. baumannii pneumonia.
Collapse
Affiliation(s)
- Aldona Jeznach
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Karolina Sidor-Dzitkowska
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Magdalena Bandyszewska
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Małgorzata Grzanka
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Piotr Popławski
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Anna Marszalik
- Department of Bacterial Physiology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | | | - Radosław Stachowiak
- Department of Bacterial Physiology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Grażyna Hoser
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
2
|
Klubdaeng A, Tovichien P. Diffuse panbronchiolitis in children misdiagnosed as asthma: A case report. World J Clin Cases 2025; 13:103501. [DOI: 10.12998/wjcc.v13.i14.103501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/19/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Diffuse panbronchiolitis (DPB) is a rare, chronic inflammatory lung disease marked by chronic cough, breathlessness, and preceding sinusitis. Symptoms often persist for years and can be misdiagnosed as asthma, particularly in children. This report describes a DPB case resolved with long-term azithromycin therapy, emphasizing the need for a timely and accurate diagnosis.
CASE SUMMARY A 12-year-old girl, diagnosed with asthma at age five and managed with inhaled corticosteroids and long-acting beta-2 agonists, developed a history of chronic productive cough and chronic sinusitis for a year. On examination, she exhibited wheezing and coarse crackles. Despite receiving treatment for an asthma exacerbation, her symptoms did not improve. A chest X-ray revealed reticulonodular infiltration in both lower lungs, prompting further evaluation with high-resolution computed tomography (HRCT). The HRCT confirmed centrilobular nodule opacities, a 'tree-in-bud' pattern, and non-tapering bronchi, suggesting DPB. Elevated cold hemagglutinin titers at 128 further supported the diagnosis. Her cough and sinusitis resolved within a month after starting azithromycin therapy, chosen for its anti-inflammatory and immunomodulatory effects. Follow-up HRCT scans after 1 year of continuous treatment showed complete normalization.
CONCLUSION This case highlights the importance of early diagnosis and prompt treatment in achieving favorable outcomes for DPB.
Collapse
Affiliation(s)
- Anuvat Klubdaeng
- Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Prakarn Tovichien
- Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
3
|
Song J, Tong L, Xu J, Li Y, Zeng Y, Chen C, Wang J, Song Y. Edaravone alleviates Pseudomonas aeruginosa associated-acute lung injury by inhibiting inflammation and promoting anti-microbial peptide production. Int Immunopharmacol 2025; 154:114511. [PMID: 40158431 DOI: 10.1016/j.intimp.2025.114511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 03/16/2025] [Accepted: 03/16/2025] [Indexed: 04/02/2025]
Abstract
Acute respiratory distress syndrome (ARDS) is the most common respiratory emergency and one of the most severe clinical syndromes. Bacterial and viral infections are the frequent etiological factors. Pseudomonas aeruginosa (PA) is the most significant gram-negative pathogen associated with pneumonia and ARDS in critically ill patients with respiratory diseases. However, multi-drug resistance and biofilm formation pose significant challenges to the clinical treatment of PA-associated pulmonary infections. In this study, we focused on edaravone (EDA), a brain-protective agent and free-radical scavenger commonly used in neurology, and examined its role in PA-ALI. We found EDA significantly mitigated pulmonary pathological damage, inflammatory responses and Reactive Oxygen Species (ROS) generation induced by PA in vivo. The in-vitro assays revealed EDA inhibited the transcription and secretion of pro-inflammatory factors induced by PA in RAW264.7 cells by targeting the TLR4/MyD88/NF-κB signaling pathways. Additionally, EDA reduced the production of intracellular ROS and cell death. EDA treatment enhanced the transcription of antimicrobial peptides, including defensin beta 1 (Defb1), defensin beta 2 (Defb2), CC motif chemokine ligand 20 (Ccl20), secretory leukocyte peptidase inhibitor (Slpi), and lactotransferrin (Ltf), with a significant upregulation of Defb1 expression. We also explored the role of EDA in lung endogenous stem cells using Sftpc-DreER; Scgb1a1-CreER; R26-TLR mice. Our findings indicated that EDA promoted the regeneration of club cells in response to PA stimulation by promoting their proliferation. And also, EDA inhibited PA infection induced cell apoptosis in lung tissues. In conclusion, EDA acts as a protective agent in PA-ALI. It not only inhibits inflammatory responses induced by PA but also enhances the expression of antimicrobial peptides and promotes club cell regeneration. Therefore, EDA may serve as an adjunctive treatment for PA-ARDS.
Collapse
Affiliation(s)
- Juan Song
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Lin Tong
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jian Xu
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yufan Li
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Yingying Zeng
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Cuicui Chen
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jian Wang
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Yuanlin Song
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China.
| |
Collapse
|
4
|
Saleh NM, Sudan ESF, Mohamed SH, El-Shahed MM, Hamouda RE, El-Gendy AO, Farag AA. Pathogen Partnerships or Power Struggles? Pseudomonas aeruginosa, and Staphylococcus aureus Dynamics in Cystic Fibrosis. Curr Microbiol 2025; 82:236. [PMID: 40198369 DOI: 10.1007/s00284-025-04167-4] [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: 03/28/2024] [Accepted: 02/26/2025] [Indexed: 04/10/2025]
Abstract
Cystic fibrosis (CF) is a polymicrobial infection characterized by interactions among various bacterial species that affect one another's cohabitation. The investigation of interspecies interactions in dual infections is essential to understand their reaction in the environment better and assist in the development of treatment regimens and innovative disease control approaches. Our hypothesis posits that co-infection interactions promote the adaptation of Staphylococcus aureus and Pseudomonas aeruginosa, potentially leading to synergistic action. To explore this, we examined dual-species interactions in co-isolated pairs of these organisms from Egyptian CF patients using laboratory media and artificial sputum media (ASM). Based on demographic data, 82 collected bacterial isolates from single, dual, and triple cultures were identified from 50 enrolled patients. In the interaction of the pairs in mimic media, P. aeruginosa exo-products significantly enhanced the biofilm formation and growth of S. aureus. Conversely, S. aureus did not inhibit P. aeruginosa biofilm formation. Furthermore, the biofilm mode of dual-organism growth provides protection in the CF context, as bacterial biofilms can withstand much higher antimicrobial levels compared to planktonically grown bacteria. Additionally, key biofilm genes regulated by quorum sensing were differentially expressed in both species in an isolate-dependent manner, highlighting their significant role in coexistence dual-species biofilm coexistence. In conclusion, our study illuminates the competitive and cooperative interactions between these two pathogens, which impact their coexistence and encourage biofilm production. This, in turn, accelerates disease progression and compromises patient health.
Collapse
Affiliation(s)
- Neveen M Saleh
- Department of Microbiology, Egyptian Drug Authority (Former National Organization for Drug Control and Research (NODCAR)), Giza, Egypt.
- College of Public Health, University of Nebraska Medical Center (UNMC), Omaha, United States.
| | - Esraa S F Sudan
- Department of Microbiology, Egyptian Drug Authority (Former National Organization for Drug Control and Research (NODCAR)), Giza, Egypt
| | - Sara H Mohamed
- Department of Microbiology, Egyptian Drug Authority (Former National Organization for Drug Control and Research (NODCAR)), Giza, Egypt
| | - Maram M El-Shahed
- Department of Microbiology, Egyptian Drug Authority (Former National Organization for Drug Control and Research (NODCAR)), Giza, Egypt
| | - Reda E Hamouda
- Department of Animal Production Systems Research, Animal Production Research Institute, Agricultural Research Center, Ministry of Agriculture and Land Reclamation, Giza, Egypt
| | - Ahmed Osama El-Gendy
- Microbiology and Immunology Department, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62514, Egypt
| | - Ahmed A Farag
- Microbiology and Immunology Department, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62514, Egypt
| |
Collapse
|
5
|
Huang Q, Yan K, Li G. Molecular characterization of virulent genes in Pseudomonas aeruginosa based on componential usage divergence. Sci Rep 2025; 15:11246. [PMID: 40175567 PMCID: PMC11965391 DOI: 10.1038/s41598-025-95579-6] [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: 11/23/2024] [Accepted: 03/21/2025] [Indexed: 04/04/2025] Open
Abstract
Genetic characteristics of virulent genes in Pseudomonas aeruginosa attracted significant attention for they could govern their drug-resistances. Studies on the componential usage divergences in the virulent genes are beneficial for further explicating their molecular characteristics. In present study, one thousand complete genomes of Pseudomonas aeruginosa were considered to study the molecular characteristics of 21 typical virulent genes. The important componential usage patterns (i.e., the base usage pattern, the codon usage pattern and their divergences) of 21 specific virulent genes were counted and calculated. The results show that (1) most virulent genes concerned in the present study are high GC sequences (overall GC ratio > 50%), especially from the codon usage perspective, the virulent genes are obviously GC3-abundant sequences (GC3 ratio > 70%); (2) the relative synonymous codon usage of all concerned virulent genes are uneven, especially in the anvM and the lptA, there is no codon for some certain amino acids, which could reveal their obvious codon usage bias; (3) some genes (i.e., the oprF and the fadD1) with lower divergence have steady effective number of codons. The findings of the present work would improve novel insights on the genetic characteristics of virulent genes in Pseudomonas aeruginosa.
Collapse
Affiliation(s)
- Qian Huang
- School of Computer Engineering, Shanxi Vocational University of Engineering Science and Technology, Jinzhong, Shanxi, China
| | - Keding Yan
- Department of Biomedical Engineering, School of Electronic Information Engineering, Xi'An Technological University, Xi'An, Shaanxi, China.
| | - Gun Li
- Department of Biomedical Engineering, School of Electronic Information Engineering, Xi'An Technological University, Xi'An, Shaanxi, China.
| |
Collapse
|
6
|
Qiu H, Dandekar AA, Dai W. Co-regulation of cooperative and private traits by PsdR in Pseudomonas aeruginosa. Evol Lett 2025; 9:273-281. [PMID: 40191406 PMCID: PMC11968183 DOI: 10.1093/evlett/qrae067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/07/2024] [Accepted: 12/06/2024] [Indexed: 04/09/2025] Open
Abstract
Social interactions profoundly shape the dynamics and functionality of microbial populations. However, mechanisms governing the regulation of cooperative or individual traits have remained elusive. Here, we investigated the regulatory mechanisms of social behaviors by characterizing the fitness of transcriptional regulator PsdR mutants in cooperating Pseudomonas aeruginosa populations. In a canonical model described previously, PsdR was shown to solely have a nonsocial role in adaptation of these populations by controlling the intracellular uptake and processing of dipeptides. In addition to these known private traits, we found that PsdR mutants also enhanced cooperation by increasing the production of quorum sensing (QS)-regulated public goods. Although private dipeptide utilization promotes individual absolute fitness, it only partially accounts for the growth advantage of PsdR mutants. The absence of the QS master regulator LasR delayed the appearance of PsdR variants in an evolution experiment. We also demonstrated that the growth fitness of PsdR mutants is determined by a combination of the QS-mediated cooperative trait and the dipeptide metabolism-related private trait. This dual trait is co-regulated by PsdR, leading to the rapid spread of PsdR variants throughout the population. In conclusion, we identified a new social model of co-regulating cooperative and private traits in PsdR variants, uncovering the social and nonsocial roles of this transcriptional regulator in cooperating bacterial populations. Our findings advance the fundamental understanding of bacterial social interactions and provide insights into population evolution, pathogen infection control and synthetic biotechnology.
Collapse
Affiliation(s)
- Huifang Qiu
- Integrative Microbiology Research Center, College of Plant Protection, South China of Agricultural University, Guangzhou, China
| | - Ajai A Dandekar
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Weijun Dai
- Integrative Microbiology Research Center, College of Plant Protection, South China of Agricultural University, Guangzhou, China
| |
Collapse
|
7
|
Hou X, Wang M, Hu T, Wu Z, Liang H, Zhong Y, Ma Z, Zhang H, Xiao L, Zhang W, Zou Y. Evaluation of the safety and probiotic properties of Limosilactobacillus fermentum BGI-AF16, a uric acid-lowering probiotic strain. Microb Pathog 2025; 201:107382. [PMID: 39961375 DOI: 10.1016/j.micpath.2025.107382] [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: 05/06/2024] [Revised: 01/08/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025]
Abstract
Some beneficial microorganisms in the intestine have the potential to degrade uric acid, offering a novel strategy for the prevention of hyperuricemia. In this study, the safety and probiotic potentials of Limosilactobacillus fermentum BGI-AF16 were evaluated by whole genome sequence analysis and in vitro experiments. Based on the gene analysis of antibiotic resistance and virulence factors, L. fermentum BGI-AF16 has been shown to be safe. We identified probiotic-related genes by genome annotation tools and conducted in vitro experiments to evaluate the ability of L. fermentum BGI-AF16 to inhibit pathogenic bacteria, tolerate a simulated gastrointestinal environment, and degrade uric acid. The results from in vitro experiments showed that L. fermentum BGI-AF16 had inhibitory effects on four clinically relevant pathogens and was highly tolerant to the gastrointestinal environment. In addition, L. fermentum BGI-AF16 was able to rapidly degrade uric acid within the first hour, and the strain could degrade 56.36 ± 2.32 % of uric acid by the third hour. The genome of the strain contains genes encoding flavin adenine dinucleotide (FAD)-dependent urate hydroxylase (EC.1.14.13.113), an enzyme that directly metabolizes uric acid. And the strain has a complete uric acid metabolic pathway. These results suggest that L. fermentum BGI-AF16 is a probiotic candidate with significant potential for reducing uric acid level.
Collapse
Affiliation(s)
- Xiaoxue Hou
- BGI Research, Shenzhen, 518083, China; College of Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Mengmeng Wang
- BGI Research, Shenzhen, 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | - Zhinan Wu
- BGI Research, Shenzhen, 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | - Yiyi Zhong
- BGI Precision Nutrition, Shenzhen, 518083, China
| | - Zhihui Ma
- BGI Precision Nutrition, Shenzhen, 518083, China
| | | | - Liang Xiao
- BGI Research, Shenzhen, 518083, China; Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, 518083, China
| | - Wenjin Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China; BGI Genomics, Shenzhen, 518083, China
| | - Yuanqiang Zou
- BGI Research, Shenzhen, 518083, China; Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, 518083, China.
| |
Collapse
|
8
|
Qu J, Yin L, Qin S, Sun X, Gong X, Li S, Pan X, Jin Y, Cheng Z, Jin S, Wu W. Identification of the Pseudomonas aeruginosa AgtR-CspC-RsaL pathway that controls Las quorum sensing in response to metabolic perturbation and Staphylococcus aureus. PLoS Pathog 2025; 21:e1013054. [PMID: 40198682 DOI: 10.1371/journal.ppat.1013054] [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: 10/17/2024] [Revised: 04/21/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025] Open
Abstract
Environmental metabolites and metabolic pathways significantly influence bacterial pathogenesis and interspecies competition. We previously discovered that a mutation in the triosephosphate isomerase gene, tpiA, in Pseudomonas aeruginosa led to defective type III secretion and increased susceptibility to aminoglycoside antibiotics. In this study, we found that the tpiA mutation enhances the Las quorum sensing system due to reduced translation of the negative regulator RsaL. Further investigations demonstrated an upregulation of CspC, a CspA family protein that represses rsaL translation. DNA pull-down assay, along with genetic studies, revealed the role of AgtR in regulating cspC transcription. AgtR is known to regulate pyocyanin production in response to N-acetylglucosamine (GlcNAc), contributing to competition against Staphylococcus aureus. We demonstrated that CspC activates the Las quorum sensing system and subsequent pyocyanin production in response to GlcNAc and S. aureus. Overall, our results elucidate the AgtR-CspC-RsaL-LasI pathway that regulates bacterial virulence factors and its role in competition against S. aureus.
Collapse
Affiliation(s)
- Junze Qu
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Liwen Yin
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Shanhua Qin
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaomeng Sun
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Xuetao Gong
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Shouyi Li
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, National Key laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Xiaolei Pan
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yongxin Jin
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhihui Cheng
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Shouguang Jin
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Weihui Wu
- Department of Microbiology, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
9
|
Gu M, Pang Z. Luteolin inhibits inflammation and M1 macrophage polarization in the treatment of Pseudomonas aeruginosa-induced acute pneumonia through suppressing EGFR/PI3K/AKT/NF-κB and EGFR/ERK/AP-1 signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156663. [PMID: 40133026 DOI: 10.1016/j.phymed.2025.156663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/07/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND The opportunistic pathogen Pseudomonas aeruginosa primarily causes infections in immunocompromised individuals. Luteolin, a natural flavonoid, is widely present in plants, which exerts various pharmacological activities, including anti-inflammatory and antimicrobial activities. PURPOSE This study aimed to explore the therapeutic efficacy of luteolin and the underlying molecular mechanisms in treating the P. aeruginosa-induced acute pneumonia. METHODS Network pharmacology was utilized to identify the core targets of luteolin for treating acute P. aeruginosa pneumonia. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed to dissect the potential effects of luteolin and the involved signaling pathways. Surface plasmon resonance (SPR) assay and molecular docking were employed for studying the binding affinities of luteolin with the key targets. Furthermore, we applied a mouse model of bacterial pneumonia for assessing the therapeutic effects of luteolin in vivo, and an in vitro infection model for specifically investigating the effects of luteolin on macrophages as well as the underlying mechanisms upon P. aeruginosa infection. RESULTS Network pharmacology identified TNF, IL-6, EGFR and AKT1 as the key targets of luteolin for treating acute P. aeruginosa pneumonia. Moreover, as revealed by GO and KEGG enrichment analysis, EGFR, MAPK and PI3K/AKT pathways were the potential pathways regulated the P. aeruginosa-induced inflammatory response. According to the in vivo results, luteolin effectively mitigated the P. aeruginosa-induced acute lung injury through reducing the pulmonary permeability, neutrophil infiltration, proinflammatory cytokine production (IL-1β, IL-6, TNF and MIP-2) and bacterial burden in lung tissues, which led to increased survival rate of mice. Furthermore, the luteolin-treated mice had diminished EGFR, PI3K, AKT, IκBα, NF-κB p65, ERK, c-Jun and c-Fos phosphorylation, down-regulated M1 macrophage marker levels (iNOS, CD86 and IL-1β) but up-regulated M2 macrophage marker levels (Ym1, CD206 and Arg1) in lung tissues. Consistently, the luteolin-pretreated macrophages exhibited reduced phosphorylation of these regulatory proteins, diminished proinflammatory cytokine production, and down-regulated expression of M1 macrophage markers, but up-regulated expression of IL-10 and M2 macrophage markers. CONCLUSION luteolin effectively suppressed the inflammatory responses and M1 macrophage polarization through inhibiting EGFR/PI3K/AKT/NF-κB and EGFR/ERK/AP-1 signaling pathways in the treatment of acute P. aeruginosa pneumonia. This study suggests that luteolin could be a promising candidate for development as a therapeutic agent for acute bacterial pneumonia.
Collapse
Affiliation(s)
- Mengdi Gu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zheng Pang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
10
|
Boujnane M, Zommiti M, Lesouhaitier O, Ferchichi M, Tahrioui A, Boukerb AM, Connil N. Pediococcus pentosaceus MZF16 Probiotic Strain Prevents In Vitro Cytotoxic Effects of Pseudomonas aeruginosa H103 and Prolongs the Lifespan of Caenorhabditis elegans. Pathogens 2025; 14:244. [PMID: 40137729 PMCID: PMC11945076 DOI: 10.3390/pathogens14030244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogenic bacterium, responsible for several life-threatening infections due to its multiple virulence factors and problematic multi-drug resistance, hence the necessity to find alternatives such as competitive probiotics. Pediococcus pentosaceus MZF16 is an LAB strain, isolated from traditional dried meat "Ossban", with high probiotic potential. Our study investigated the capacity of P. pentosaceus MZF16 to counteract P. aeruginosa H103 using several tests on intestinal cells (analysis of cytotoxicity, inflammation, adhesion/invasion) and on the in vivo Caenorhabditis elegans model. The effect of MZF16 on the quorum sensing of the pathogen was also examined. We found that P. pentosaceus MZF16 was able to reduce H103 cytotoxicity and inflammatory activity and prevented pathogen colonization and translocation across Caco-2/TC7 cells. MZF16 also exerted an anti-virulence effect by attenuating quorum-sensing (QS) molecules and pyoverdine production and extended C. elegans lifespan. The obtained results highlight the potential of P. pentosaceus MZF16 probiotic strain as an anti-Pseudomonas aeruginosa alternative and establish a basis for elucidating the mechanisms of P. pentosaceus MZF16 involved in countering P. aeruginosa virulence.
Collapse
Affiliation(s)
- Meryem Boujnane
- CBSA UR 4312, Université de Rouen Normandie, Université de Caen Normandie, Normandie Université, F-76000 Rouen, France; (M.B.); (M.Z.); (O.L.); (A.T.); (A.M.B.)
| | - Mohamed Zommiti
- CBSA UR 4312, Université de Rouen Normandie, Université de Caen Normandie, Normandie Université, F-76000 Rouen, France; (M.B.); (M.Z.); (O.L.); (A.T.); (A.M.B.)
| | - Olivier Lesouhaitier
- CBSA UR 4312, Université de Rouen Normandie, Université de Caen Normandie, Normandie Université, F-76000 Rouen, France; (M.B.); (M.Z.); (O.L.); (A.T.); (A.M.B.)
| | - Mounir Ferchichi
- Unité de Protéomique Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université Tunis El-Manar, Tunis 1006, Tunisia;
| | - Ali Tahrioui
- CBSA UR 4312, Université de Rouen Normandie, Université de Caen Normandie, Normandie Université, F-76000 Rouen, France; (M.B.); (M.Z.); (O.L.); (A.T.); (A.M.B.)
| | - Amine M. Boukerb
- CBSA UR 4312, Université de Rouen Normandie, Université de Caen Normandie, Normandie Université, F-76000 Rouen, France; (M.B.); (M.Z.); (O.L.); (A.T.); (A.M.B.)
| | - Nathalie Connil
- CBSA UR 4312, Université de Rouen Normandie, Université de Caen Normandie, Normandie Université, F-76000 Rouen, France; (M.B.); (M.Z.); (O.L.); (A.T.); (A.M.B.)
| |
Collapse
|
11
|
Targońska-Karasek M, Polkowska I, Krukowski H. Is Pseudomonas aeruginosa a possible aetiological agent of periodontitis in dogs? J Vet Res 2025; 69:35-40. [PMID: 40144062 PMCID: PMC11936081 DOI: 10.2478/jvetres-2025-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 02/07/2025] [Indexed: 03/28/2025] Open
Abstract
Introduction Periodontal diseases are the most frequently diagnosed problem in small animal veterinary medicine. Although their exact cause is not fully understood, bacteria play an important role in their development. Pseudomonas aeruginosa is a Gram-negative, rod-shaped, non-spore-forming bacterium. The living environment of this bacterium may be soil and water; however, it can also be found in humans and animals. Antibiotic treatment of periodontitis may be complicated by the carbapenem resistance of some P. aeruginosa strains, if these bacteria are found to be an aetiological agent. The aim of the study was to identify all bacterial strains isolated from dog with periodontitis. Material and Methods After a clinical examination of a Schnauzer dog in the Department and Clinic of Animal Surgery in the University of Life Sciences in Lublin Faculty of Veterinary Medicine, periodontitis was diagnosed. A swab was taken from the diseased tissue and submitted for microbiological tests. Microorganisms were initially identified by colony morphology, haemolytic pattern and Gram staining, and subsequently by sensitivity tests, VITEK 2 and matrix-assisted laser desorption/ionisation-time-of-flight. Results Pseudomonas aeruginosa was isolated and identified as a probable aetiological factor of periodontitis in dogs. Conclusion In our opinion, attention should be paid to Pseudomonas aeruginosa as a possible aetiological factor of periodontal diseases in dogs.
Collapse
Affiliation(s)
- Małgorzata Targońska-Karasek
- Department of Animal Hygiene and Environmental Hazards, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, 20-950Lublin, Poland
| | - Izabela Polkowska
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, 20-612Lublin, Poland
| | - Henryk Krukowski
- Department of Animal Hygiene and Environmental Hazards, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, 20-950Lublin, Poland
| |
Collapse
|
12
|
Artono A, Purnami N, Handoko E, Widodo ADW, Juniastuti J. Pseudomonas aeruginosa in Chronic Suppurative Otitis Media. Infect Chemother 2025; 57:63-71. [PMID: 40183654 PMCID: PMC11972902 DOI: 10.3947/ic.2024.0062] [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: 06/10/2024] [Accepted: 11/25/2024] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Chronic suppurative otitis media (CSOM) has caused many hearing disorder cases in developing countries. Inappropriate antibiotic use resulted in a shift of bacterial resistance. The biofilm-forming bacteria, like Pseudomonas aeruginosa, was a common germ detected in CSOM that contributed to a poor prognosis. This study aimed to investigate the bacterial pattern from samples taken from CSOM patients regarding its antibiotic susceptibility and the antibiofilm activity of acetic acid against P. aeruginosa. MATERIALS AND METHODS Sterile swabs of forty-five patients with CSOM were collected, followed by isolation of bacterial pathogens, identification, and evaluation of antibiotic sensitivity using modified Kirby Bauer disc diffusion protocol. In vitro testing was done by adding acetic acid to P. aeruginosa culture to gauge the minimum concentration of biofilm inhibition and eradication. They were conducted using the microtiter plate assay method and quantified with an ELISA reader. The data were analyzed statistically using One-Way ANOVA and Tukey Honestly Significant Difference post hoc test. RESULTS The samples obtained from 31 of 45 CSOM patients showed positive microbial growth; 26 (57.78%) had a monomicrobial pattern, and 5 (11.11%) had a polymicrobial pattern. The researcher ascertained that 24 isolates, representing 66.67%, were gram-negative bacteria, with P.-aeruginosa identified as the predominant species. P. aeruginosa isolates were sensitive to several antibiotics, including meropenem, amikacin, piperacillin-tazobactam, ceftazidime, and cefoperazone-sulbactam with a rate of 93.33%. The minimum concentration of acetic acid required to qualify as the minimum biofilm inhibitory concentration (MBIC) was determined to be 0.16%, yielding an inhibition rate of 26.79%. A concentration of 0.31% was identified as the minimum biofilm eradication concentration (MBEC), achieving an eradication rate of 77.27%. CONCLUSION P. aeruginosa, the most common bacteria found in CSOM samples, was sensitive to imipenem, amikacin, piperacillin-tazobactam, ceftazidime, and cefoperazone-sulbactam. Acetic acid suppresses P. aeruginosa bacterial biofilm formation at MBIC of 0.16% and MBEC of 0.31%.
Collapse
Affiliation(s)
- Artono Artono
- Doctoral Program, Faculty of Medicine, Airlangga University, Surabaya, Indonesia
| | - Nyilo Purnami
- Department of Otorhinolaryngology Head and Neck Surgery, Faculty of Medicine, Airlangga University/Dr. Soetomo General Hospital, Surabaya, Indonesia.
| | - Edi Handoko
- Department of Otorhinolaryngology Head and Neck Surgery, Faculty of Medicine, Brawijaya University, Malang, Indonesia
| | - Agung Dwi Wahyu Widodo
- Department of Clinical Microbiology, Faculty of Medicine, Airlangga University/Dr. Soetomo General Hospital, Surabaya, Indonesia
| | - Juniastuti Juniastuti
- Department of Clinical Microbiology, Faculty of Medicine, Airlangga University/Dr. Soetomo General Hospital, Surabaya, Indonesia
| |
Collapse
|
13
|
Alyahyawy OY, Munshi RM, Badr-Eldin SM, Aldawsari HM, Abualsunun W, Abbas HA, Salem IM, Hegazy WAH, Nazeih SI. Reprofiling lamivudine as an antibiofilm and anti-pathogenic agent against Pseudomonas aeruginosa. AMB Express 2025; 15:33. [PMID: 39985628 PMCID: PMC11846793 DOI: 10.1186/s13568-025-01835-3] [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: 10/20/2024] [Accepted: 02/02/2025] [Indexed: 02/24/2025] Open
Abstract
Resistance to antibiotics is a critical growing public health problem that needs urgent action to combat. To avoid the stress on bacterial growth that evokes the development of resistance, anti-virulence agents can be an attractive strategy as they do not target bacterial growth. There are FDA approved drugs have been screened for their anti-virulence activities. Lamivudine (LAM) is a synthetic nucleoside analogue used as an antiretroviral in treatment of HIV and can be used in treatment of HBV. The present study aimed to assess the anti-virulence activities of LAM against a clinically important pathogen Pseudomonas aeruginosa. The LAM's antibiofilm and anti-virulence activities were evaluated. The impact of LAM on the quorum sensing (QS) systems which control the production of these virulence factors was assessed virtually and by quantification of the expression of QS-encoding genes. Furthermore, in vivo mice protection assay was conducted to attest the LAM's anti-pathogenic activity. The current findings elaborated the promising anti-pathogenic and anti-QS activities of LAM. LAM interfered with biofilm formation in P. aeruginosa PAO1 strain. Moreover, swarming motility and production of pyocyanin and protease were significantly diminished. At the molecular level, LAM downregulated the QS-encoding genes LasI, LasR, RhlR, PqsA and PqsR. Additionally, the detailed in silico docking and molecular simulation studies showed the considered high LAM's ability to bind and hinder the QS receptors in the P. aeruginosa. In an agreement with in vitro and in silico, the in vivo results showed the LAM full protection of mice against P. aeruginosa. In conclusion, LAM could be repurposed to be employed as adjunct therapy with traditional antibiotics for treating serious pseudomonal infections.
Collapse
Affiliation(s)
- Othman Yahya Alyahyawy
- Department of Medical Laboratory Technology (MLT), Faculty of Applied Medical Sciences, King Abdulaziz University, 80200, Rabigh, Saudi Arabia
| | - Raafat M Munshi
- Department of Medical Laboratory Technology (MLT), Faculty of Applied Medical Sciences, King Abdulaziz University, 80200, Rabigh, Saudi Arabia
| | - Shaimaa M Badr-Eldin
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Hibah Mubarak Aldawsari
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Walaa Abualsunun
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Hisham A Abbas
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Ibrahim M Salem
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sphinx University, Assiut, Egypt
| | - Wael A H Hegazy
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
- Department of Pharmaceutical Sciences, Pharmacy Program, Oman College of Health Sciences, 113, Muscat, Oman.
| | - Shaimaa I Nazeih
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
14
|
Cabrera R, Rovira-Ribalta N, Motos A, Bueno-Freire L, Vázquez N, Soler-Comas A, Alcaraz-Serrano V, López-Aladid R, Muñoz L, Vila J, Torres A, Fernández-Barat L. Virulence factors of Pseudomonas aeruginosa and immune response during exacerbations and stable phase in bronchiectasis. Sci Rep 2025; 15:6520. [PMID: 39987197 PMCID: PMC11846832 DOI: 10.1038/s41598-025-91368-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 02/20/2025] [Indexed: 02/24/2025] Open
Abstract
The study of key Pseudomonas aeruginosa (PA) virulence factors, the molecular basis of pathogenicity, as well as their correlation with the immune response during exacerbations in patients with non-cystic fibrosis bronchiectasis can help to identify novel targets and biomarkers for clinical management. The objective was to compare P. aeruginosa virulence and the patient's immune response during stable phases and exacerbations of bronchiectasis. We used polymerase chain reaction (PCR) and real-time quantitative PCR (qRT-PCR) to perform molecular characterization of the genomic islands and virulence genes present in 42 P. aeruginosa strains obtained from the sputum of patients with bronchiectasis during stability and exacerbations. Immunoglobulin (Ig) and interleukin (IL) levels in 32 serum samples were analyze by ELISA and Luminex assay. A greater presence of the conjugative element pKLC102, specific virulence genes (exoS, exoY) and pyoverdine production characterize the P. aeruginosa strains obtained during exacerbations. The expression levels of type III secretion system (exoS, exoY) showed an important role in the humoral immune response during exacerbations. Exacerbations were associated with high levels of IL-6. The presence of specific genomic islands, virulence genes, and increased IL-6 levels provide an accurate characterization on bronchiectasis exacerbations. These targets could be useful in the prevention, management and treatment of these exacerbations.
Collapse
Affiliation(s)
- Roberto Cabrera
- Hospital Clínic, Cellex Laboratory, CIBERES (Center for Net Biomedical Research Respiratory Diseases, 06/06/0028), FCRB-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), School of Medicine, University of Barcelona, Barcelona, Spain
- Respiratory Intensive Care Unit, Pneumology Department, Hospital Clínic, Barcelona, Spain
| | - Nona Rovira-Ribalta
- Hospital Clínic, Cellex Laboratory, CIBERES (Center for Net Biomedical Research Respiratory Diseases, 06/06/0028), FCRB-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), School of Medicine, University of Barcelona, Barcelona, Spain
- Respiratory Intensive Care Unit, Pneumology Department, Hospital Clínic, Barcelona, Spain
| | - Anna Motos
- Hospital Clínic, Cellex Laboratory, CIBERES (Center for Net Biomedical Research Respiratory Diseases, 06/06/0028), FCRB-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), School of Medicine, University of Barcelona, Barcelona, Spain
- Respiratory Intensive Care Unit, Pneumology Department, Hospital Clínic, Barcelona, Spain
| | - Leticia Bueno-Freire
- Hospital Clínic, Cellex Laboratory, CIBERES (Center for Net Biomedical Research Respiratory Diseases, 06/06/0028), FCRB-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), School of Medicine, University of Barcelona, Barcelona, Spain
- Respiratory Intensive Care Unit, Pneumology Department, Hospital Clínic, Barcelona, Spain
| | - Nil Vázquez
- Hospital Clínic, Cellex Laboratory, CIBERES (Center for Net Biomedical Research Respiratory Diseases, 06/06/0028), FCRB-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), School of Medicine, University of Barcelona, Barcelona, Spain
- Respiratory Intensive Care Unit, Pneumology Department, Hospital Clínic, Barcelona, Spain
| | - Alba Soler-Comas
- Hospital Clínic, Cellex Laboratory, CIBERES (Center for Net Biomedical Research Respiratory Diseases, 06/06/0028), FCRB-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), School of Medicine, University of Barcelona, Barcelona, Spain
- Respiratory Intensive Care Unit, Pneumology Department, Hospital Clínic, Barcelona, Spain
| | - Victoria Alcaraz-Serrano
- Hospital Clínic, Cellex Laboratory, CIBERES (Center for Net Biomedical Research Respiratory Diseases, 06/06/0028), FCRB-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), School of Medicine, University of Barcelona, Barcelona, Spain
- Respiratory Intensive Care Unit, Pneumology Department, Hospital Clínic, Barcelona, Spain
| | - Rubén López-Aladid
- Hospital Clínic, Cellex Laboratory, CIBERES (Center for Net Biomedical Research Respiratory Diseases, 06/06/0028), FCRB-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), School of Medicine, University of Barcelona, Barcelona, Spain
- Respiratory Intensive Care Unit, Pneumology Department, Hospital Clínic, Barcelona, Spain
| | - Laura Muñoz
- Department of Clinical Microbiology, Hospital Clínic, CIBERINFEC, Barcelona Global Health Institute, University of Barcelona, Barcelona, Spain
| | - Jordi Vila
- Department of Clinical Microbiology, Hospital Clínic, CIBERINFEC, Barcelona Global Health Institute, University of Barcelona, Barcelona, Spain
| | - Antoni Torres
- Hospital Clínic, Cellex Laboratory, CIBERES (Center for Net Biomedical Research Respiratory Diseases, 06/06/0028), FCRB-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), School of Medicine, University of Barcelona, Barcelona, Spain.
- Respiratory Intensive Care Unit, Pneumology Department, Hospital Clínic, Barcelona, Spain.
| | - Laia Fernández-Barat
- Hospital Clínic, Cellex Laboratory, CIBERES (Center for Net Biomedical Research Respiratory Diseases, 06/06/0028), FCRB-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), School of Medicine, University of Barcelona, Barcelona, Spain.
- Respiratory Intensive Care Unit, Pneumology Department, Hospital Clínic, Barcelona, Spain.
| |
Collapse
|
15
|
Kar A, Mukherjee SK, Hossain ST. Regulatory role of PA3299.1 small RNA in Pseudomonas aeruginosa biofilm formation via modulation of algU and mucA expression. Biochem Biophys Res Commun 2025; 748:151348. [PMID: 39827548 DOI: 10.1016/j.bbrc.2025.151348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Small RNAs (sRNAs) have emerged as key regulators of transcriptional factors and components within regulatory networks that govern bacterial biofilm formation. This study aimed to explore the regulatory role of the PA3299.1 sRNA in controlling biofilm formation in P. aeruginosa. Results showed that PA3299.1 expression was significantly elevated in both substratum-attached and colony biofilms compared to planktonic growth. Further investigation revealed that strains overexpressing PA3299.1 exhibited enhanced biofilm formation, while its deletion resulted in a substantial reduction in biofilm development. PA3299.1 was found to regulate the expression of AlgU and MucA, the sigma and anti-sigma factors, integral to the biofilm developmental network. In summary, this research identifies PA3299.1 as a critical regulator of biofilm formation and potentially a contributor to the pathogenicity of P. aeruginosa, that could help to develop new therapeutic strategies to manage biofilm-associated infections.
Collapse
Affiliation(s)
- Amiya Kar
- Department of Microbiology, University of Kalyani, Kalyani, 741235, India
| | | | | |
Collapse
|
16
|
Al-Daghistani HI, Matalqah SM, Shadid KA, Abu-Niaaj LF, Zein S, Abo-Ali RM. Quorum Quenching of P. aeruginosa by Portulaca oleracea Methanolic Extract and Its Phytochemical Profile. Pathogens 2025; 14:163. [PMID: 40005538 PMCID: PMC11858189 DOI: 10.3390/pathogens14020163] [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: 12/27/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Quorum sensing (QS) is a molecular communication mechanism among bacterial cells. It is critical in regulating virulence factors, motility, antibiotic resistance, and biofilm formation. Pseudomonas aeruginosa is a Gram-negative opportunistic pathogen linked to healthcare-associated infections, food poisoning, and biofilm formation. Treating infections caused by pathogenic bacteria has become a challenge due to the development of multi-antibiotic resistance upon continuous exposure of bacteria to antibiotics. An alternative strategy to conventional antimicrobials to decrease the bacterial pathogenicity is QS inhibition, also known as quorum quenching. Using plant-derived compounds is an environmentally friendly strategy to block the bacterial QS and inhibit bacterial growth. Portulaca oleracea is a popular plant in different countries and is also used in traditional medicine. It is widely consumed raw in salads and as garnishes, though it can be cooked as a vegetarian dish. This study evaluates the antimicrobial activity of the methanolic extract of P. oleracea and its effectiveness in blocking or attenuating the QS of P. aeruginosa. The agar well diffusion method used for screening the antibacterial activity showed a significant growth inhibition of P. aeruginosa by the extract at 500 mg/mL with a minimum inhibitory concentration of 31.25 mg/mL. A bioindicator bacterium, Chromobacterium violaceum CV026, was used to determine the effect of the methanolic extract on the QS of P. aeruginosa. The results indicated a significant reduction in biofilm formation, pyocyanin production, and LasA staphylolytic activity. The phytochemical analysis by Gas Chromatography-Mass Spectrometry showed that the methanolic extract contained several phenols, alkaloids, esters, and other compounds previously reported to have antibacterial and antioxidant effects. These findings highlight the effectiveness of P. oleracea methanolic extract in attenuating the QS and virulence factors of P. aeruginosa. This study suggests that P. oleracea is an important source of natural antimicrobials and its use would be beneficial in food and pharmaceutical applications.
Collapse
Affiliation(s)
- Hala I. Al-Daghistani
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan;
| | - Sina M. Matalqah
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; (S.M.M.); (K.A.S.)
| | - Khalid A. Shadid
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; (S.M.M.); (K.A.S.)
| | - Lubna F. Abu-Niaaj
- Department of Agricultural and Life Sciences, College of Engineering, Science, Technology and Agriculture, Central State University, Wilberforce, OH 45384, USA
| | - Sima Zein
- Department of Pharmaceutical Biotechnology, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan;
| | - Raeda M. Abo-Ali
- Faculty of Nursing, Al-Balqa Applied University, Amman 19117, Jordan;
| |
Collapse
|
17
|
Maan SA, Faiesal AA, Gamar GM, El Dougdoug NK. Efficacy of bacteriophages with Aloe vera extract in formulated cosmetics to combat multidrug-resistant bacteria in skin diseases. Sci Rep 2025; 15:4335. [PMID: 39910123 PMCID: PMC11799309 DOI: 10.1038/s41598-025-86334-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 01/09/2025] [Indexed: 02/07/2025] Open
Abstract
Phage therapy offers a promising alternative to antibiotic treatment for combating illnesses caused by multidrug-resistant bacteria. In this study, pathogenic bacteria Staphylococcus aureus and Pseudomonas aeruginosa were isolated from pus and skin infected fluidsusing selective media. These bacterial isolates were biochemically identified as S. aureus and P. aeruginosa with probabilities of 98% and 99%, respectively, through VITEK2 system, and were confirmed as multidrug-resistant based on minimum inhibitory concentration test using colorimetric reagent cards. Lytic phages specific to these isolates were isolated, identified through plaque assays, transmission electron microscopy and classified morphologically according to the new International Committee on Taxonomy of Viruses classification as members of the Straboviridae, Drexlerviridae, and Autographiviridae families. A cosmetic gel formulation combining Aloe vera extract and the phage cocktail was prepared and tested. This gel significantly enhanced phage longevity and reduced bacterial growth by 95.5% compared to the reductions of 90.5% with Aloe Vera extract alone and 45.7% with the basic cosmetic gel. The phage remained effective for 4 to over 12 weeks after being preserved in the cosmetic formula, maintaining populations ranging from 5 × 103 to 25 × 104 PFU/mL in vitro. These findings highlight the potential of phage-based formulations, such as Vena Skin Gel, as innovative biotherapeutic tools for managing skin infections.
Collapse
Affiliation(s)
- Sodaf A Maan
- Department of Agricultural Microbiology, Faculty of Agriculture, Ain Shams University, Cairo, Egypt.
| | - Abeer A Faiesal
- Department of Basic and Applied Agriculture Sciences, Higher Institute for Agriculture Cooperation, Cairo, Egypt
| | - Gamar M Gamar
- Department of Life and Earth Sciences, Higher N'Djamena Institute for Training Teachers, N'Djamena, Chad
| | - Noha K El Dougdoug
- Department of Plant and Microbiology, Faculty of Science, Benha University, Banha, Egypt
| |
Collapse
|
18
|
Le Terrier C, Raro OHF, Saad AM, Nordmann P, Poirel L. In-vitro activity of newly-developed β-lactamase inhibitors avibactam, relebactam and vaborbactam in combination with anti-pseudomonal β-lactam antibiotics against AmpC-overproducing clinical Pseudomonas aeruginosa isolates. Eur J Clin Microbiol Infect Dis 2025; 44:277-284. [PMID: 39589655 PMCID: PMC11754317 DOI: 10.1007/s10096-024-04965-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/09/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE Overproduction of the intrinsic chromosomally-encoded AmpC β-lactamase is one of the main mechanisms responsible for broad-spectrum β-lactam resistance in Pseudomonas aeruginosa. Our study aimed to evaluate the in-vitro activity of anti-pseudomonal β-lactam molecules associated with the recently-developed and commercially-available β-lactamase inhibitors, namely avibactam, relebactam and vaborbactam, against P. aeruginosa isolates overproducing their AmpC. METHODS MIC values of ceftazidime, cefepime, meropenem, imipenem and ceftolozane with or without β-lactam inhibitor were determined for 50 AmpC-overproducing P. aeruginosa clinical isolates. MIC breakpoints for resistance were retained at 8 mg/L for β-lactams and β-lactam/β-lactamase inhibitor combinations containing ceftazidime, cefepime and meropenem, while 4 mg/L was used for those containing imipenem and ceftolozane. The concentration of all β-lactamases inhibitors was fixed at 4 mg/L, except for vaborbactam (8 mg/L). RESULTS The rates of isolates not being resistant to ceftazidime, cefepime, meropenem, imipenem and ceftolozane were found at 12%, 22%, 34%, 8% and 74%, respectively. When combined with avibactam, those rates increased to 60%, 62%, 60%, 46%, and 80%, respectively. The highest rates were found with relebactam-based combinations, being 76%, 64%, 66%, 76% and 84%, respectively. By contrast, associations with vaborbactam did not lead to significantly increased "non-resistance" rates. CONCLUSION Our results showed that all combinations including relebactam led to higher "non-resistance" rates against AmpC-overproducing P. aeruginosa clinical isolates. The best activity was achieved by combining ceftolozane and relebactam, that might therefore be considered as an excellent clinical alternative against AmpC overproducers.
Collapse
Affiliation(s)
- Christophe Le Terrier
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology Unit, Department of Medicine, Faculty of Science, University of Fribourg, Chemin du Musée 18, Fribourg, CH-1700, Switzerland
- Division of Intensive Care Unit, University Hospitals of Geneva, Geneva, Switzerland
| | - Otávio Hallal Ferreira Raro
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology Unit, Department of Medicine, Faculty of Science, University of Fribourg, Chemin du Musée 18, Fribourg, CH-1700, Switzerland
| | - Alaaeldin Mohamed Saad
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology Unit, Department of Medicine, Faculty of Science, University of Fribourg, Chemin du Musée 18, Fribourg, CH-1700, Switzerland
- Department of Zoonoses, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Patrice Nordmann
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology Unit, Department of Medicine, Faculty of Science, University of Fribourg, Chemin du Musée 18, Fribourg, CH-1700, Switzerland
- Swiss National Reference Center for Emerging Antibiotic Resistance, Fribourg, Switzerland
| | - Laurent Poirel
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology Unit, Department of Medicine, Faculty of Science, University of Fribourg, Chemin du Musée 18, Fribourg, CH-1700, Switzerland.
- Swiss National Reference Center for Emerging Antibiotic Resistance, Fribourg, Switzerland.
| |
Collapse
|
19
|
Alabdali YAJ, Azeez DA, Munahi MG, Kuwait ZI. Molecular Analysis of Pseudomonas aeruginosa Isolates with Mutant gyrA Gene and Development of a New Ciprofloxacin Derivative for Antimicrobial Therapy. Mol Biotechnol 2025; 67:649-660. [PMID: 38302682 DOI: 10.1007/s12033-024-01076-y] [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] [Accepted: 01/12/2024] [Indexed: 02/03/2024]
Abstract
This study focuses on the prevalence of Pseudomonas aeruginosa in various medical specimens. In addition, the investigates of this research shows the genetic analysis of pathogen-resistant isolates and chemical modifications to ciprofloxacin. A total of 225 specimens from men and women aged 30 to 60 were carefully collected and examined, including samples from wound, burn, urine, sputum, and ear samples. The data were obtained from AL Muthanna hospitals. PCR-RFLP and gene expression analysis were used to identify resistant strains and explore the genetic basis of antibiotic resistance. A ciprofloxacin derivative was synthesized and confirmed through FT-IR, 1H-NMR, and mass spectroscopy techniques then it was tested as antibacterial agent. Also, molecular docking study was conducted to predict the mechanism of action for the synthesized derivative. The results demonstrated that wound samples had the highest positive rate (33.7%) of P. aeruginosa isolates. The PCR-RFLP testing correlated ciprofloxacin resistance with gyrA gene mutation. Gene expression analysis revealed significant changes in the gyrA gene expression in comparison to the reference rpsL gene subsequent to exposure to the synthesized derivative. Furthermore, the molecular docking investigation illustrated the strategic positioning of the ciprofloxacin derivative within the DNA-binding site of the gyrA enzyme. The examination of genetic expression patterns manifested diverse effects attributed to the CIP derivative on P. aeruginosa, thus portraying it as a viable candidate in the quest for the development of novel antimicrobial agents. Ciprofloxacin derivative may offer new antimicrobial therapeutic options for treating Pseudomonas aeruginosa infections in wound specimens, addressing resistance and gyrA gene mutations.
Collapse
Affiliation(s)
| | - Dhay Ali Azeez
- Department of Biology, College of Science, Al Muthanna University, Al Muthanna, Iraq
| | - Murad G Munahi
- Department of Biology, College of Science, Al Muthanna University, Al Muthanna, Iraq
| | - Zainab I Kuwait
- The Department of Chemistry, College of Science, Al Muthanna University, Al Muthanna, Iraq
| |
Collapse
|
20
|
Ebrahim H, Haldenby S, Moore M, Dashti A, Floyd R, Fothergill J. Genotypic and phenotypic analyses of two distinct sets of Pseudomonas aeruginosa urinary tract isolates. J Med Microbiol 2025; 74:001971. [PMID: 40013918 PMCID: PMC11868659 DOI: 10.1099/jmm.0.001971] [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: 05/27/2024] [Accepted: 01/18/2025] [Indexed: 02/28/2025] Open
Abstract
Introduction. Urinary tract infections (UTIs) are associated with a high burden of morbidity, mortality and cost. Pseudomonas aeruginosa employs a myriad of virulence factors, including biofilm formation and motility mechanisms, to cause infections including persistent UTIs. P. aeruginosa is highly resistant to antibiotics, and the World Health Organization has identified it as a pathogen for which novel antimicrobials are urgently required.Gap statement. Genotypic and phenotypic characterization of P. aeruginosa from UTIs is underreported. In addition, the rise of antimicrobial resistance (AMR) is a cause for concern, particularly in many countries where surveillance is severely lacking.Aim. To identify genotypic and phenotypic characteristics of P. aeruginosa UTI isolates sourced from the UK and the state of Kuwait, with an emphasis on genotypic diversity and AMR.Methods. Twenty-three P. aeruginosa UTI isolates were sourced from the UK and Kuwait. To establish the phenotypes of UK isolates, growth analysis, biofilm formation assays, motility assays and antibiotic disc diffusion assays were performed. Whole-genome sequencing, antimicrobial susceptibility assays and in silico detection of AMR-associated genes were conducted on both sets of isolates.Results. In terms of their phenotypic characteristics and genomic composition, the UTI isolates varied. Multiple resistance genes are associated with resistance to various classes of antibiotics, such as aminoglycosides, fluoroquinolones and β-lactams, particularly in isolates from Kuwait. Extreme antibiotic resistance was detected in the isolates obtained from Kuwait, indicating that the country may be an antibiotic resistance hotspot.Conclusion. This study highlights that isolates from UTIs are diverse and can display extremely high resistance. Surveillance in countries such as Kuwait is currently limited, and this study suggests the need for greater surveillance.
Collapse
Affiliation(s)
- H. Ebrahim
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- School of Arts and Sciences, American International University, Kuwait
| | - S. Haldenby
- Centre for Genomic Research, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - M.P. Moore
- School of Life Sciences, University of Warwick, Coventry, UK
| | - A.A. Dashti
- Department of Medical Laboratory Sciences, Faculty of Allied Health Science, Health Sciences Centre, Kuwait University, Kuwait
| | - R.V. Floyd
- School of Life Sciences, University of Liverpool, Liverpool, UK
| | - J.L. Fothergill
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
21
|
Golan N, Parizat A, Tabachnikov O, Barnea E, Olsen WP, Otzen DE, Landau M. Resilience and charge-dependent fibrillation of functional amyloid: Interactions of Pseudomonas biofilm-associated FapB and FapC amyloids. J Biol Chem 2025; 301:108096. [PMID: 39706277 PMCID: PMC11787515 DOI: 10.1016/j.jbc.2024.108096] [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: 09/25/2024] [Revised: 11/21/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024] Open
Abstract
FapC and FapB are biofilm-associated amyloids involved in the virulence of Pseudomonas and other bacteria. We herein demonstrate their exceptional thermal and chemical resilience, suggesting that their biofilm structures might withstand standard sterilization, thereby contributing to the persistence of Pseudomonas aeruginosa infections. Our findings also underscore the impact of environmental factors on functional amyloid in Pseudomonas (Fap) proteins, suggesting that orthologs in different Pseudomonas strains adapt to specific environments and roles. Challenging previous assumptions about a simple nucleation role for FapB in promoting FapC aggregation, the study shows a significant influence of FapC on FapB aggregation. The interaction between these FapB and FapC is intricate: FapB stabilizes FapC fibrils, while FapC slows down FapB fibrillation but can still serve as a cross-seeding template. This complex interplay is the key to understanding their roles in bacterial biofilms. Furthermore, the study highlights distinct differences between Fap and Escherichia coli's CsgA (curli) amyloid, where CsgB assumes a simple unidirectional role in nucleating CsgA fibrillation, emphasizing the importance of a comprehensive understanding of various amyloid systems. This knowledge is vital for developing effective intervention strategies against bacterial infections and leveraging the unique properties of these amyloids in technological applications such as novel bionanomaterials or protective coatings.
Collapse
Affiliation(s)
- Nimrod Golan
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Amit Parizat
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Orly Tabachnikov
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Eilon Barnea
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - William P Olsen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Meytal Landau
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel; CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany; The Center for Experimental Medicine, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany; European Molecular Biology Laboratory (EMBL), Hamburg, Germany.
| |
Collapse
|
22
|
Marchi J, Minh CNN, Debarbieux L, Weitz JS. Multi-strain phage induced clearance of bacterial infections. PLoS Comput Biol 2025; 21:e1012793. [PMID: 39903766 PMCID: PMC11828373 DOI: 10.1371/journal.pcbi.1012793] [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: 09/30/2024] [Revised: 02/14/2025] [Accepted: 01/14/2025] [Indexed: 02/06/2025] Open
Abstract
Bacteriophage (or 'phage' - viruses that infect and kill bacteria) are increasingly considered as a therapeutic alternative to treat antibiotic-resistant bacterial infections. However, bacteria can evolve resistance to phage, presenting a significant challenge to the near- and long-term success of phage therapeutics. Application of mixtures of multiple phages (i.e., 'cocktails') has been proposed to limit the emergence of phage-resistant bacterial mutants that could lead to therapeutic failure. Here, we combine theory and computational models of in vivo phage therapy to study the efficacy of a phage cocktail, composed of two complementary phages motivated by the example of Pseudomonas aeruginosa facing two phages that exploit different surface receptors, LUZ19v and PAK_P1. As confirmed in a Luria-Delbrück fluctuation test, this motivating example serves as a model for instances where bacteria are extremely unlikely to develop simultaneous resistance mutations against both phages. We then quantify therapeutic outcomes given single- or double-phage treatment models, as a function of phage traits and host immune strength. Building upon prior work showing monophage therapy efficacy in immunocompetent hosts, here we show that phage cocktails comprised of phage targeting independent bacterial receptors can improve treatment outcome in immunocompromised hosts and reduce the chance that pathogens simultaneously evolve resistance against phage combinations. The finding of phage cocktail efficacy is qualitatively robust to differences in virus-bacteria interactions and host immune dynamics. Altogether, the combined use of theory and computational analysis highlights the influence of viral life history traits and receptor complementarity when designing and deploying phage cocktails in immunocompetent and immunocompromised hosts.
Collapse
Affiliation(s)
- Jacopo Marchi
- Department of Biology, University of Maryland, College Park, Maryland, United States of America
| | - Chau Nguyen Ngoc Minh
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Bacteriophage Bacterium Host, Paris, France
- Sorbonne Université, Collége Doctoral, Paris, France
| | - Laurent Debarbieux
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Bacteriophage Bacterium Host, Paris, France
| | - Joshua S Weitz
- Department of Biology, University of Maryland, College Park, Maryland, United States of America
- Department of Physics, University of Maryland, College Park, Maryland, United States of America
- University of Maryland Institute for Health Computing, North Bethesda, Maryland, United States of America
| |
Collapse
|
23
|
Chen Y, Xu M, Pan J, Liao Y, Na J, Li P, Sun Y, Yu S, Zhao Y, Hu H. Moxifloxacin-Loaded Polymeric Nanoparticles for Overcoming Multidrug Resistance in Chronic Pulmonary Infections Caused by Pseudomonas aeruginosa. ACS APPLIED MATERIALS & INTERFACES 2025; 17:5695-5709. [PMID: 39804842 DOI: 10.1021/acsami.4c14991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) infections are increasingly challenging due to their propensity to form biofilms and low outer membrane permeability, especially in chronically infected patients with thick mucus. P. aeruginosa exhibits multiple drug resistance mechanisms, making it one of the most significant global public health threats. In this study, we found that moxifloxacin (MXC) and antibacterial peptides (ε-poly-l-lysine, ε-PLL) exhibited a synergistic effect against multidrug-resistant P. aeruginosa (MDR-P. aeruginosa). MXC was combined with ε-PLL to prepare lipase-responsive nanoparticles (MCIP/(PEG-PCL)/PLL NPs) with a weakly negative charge. The weakly negatively charged MCIP/(PEG-PCL)/PLL NPs demonstrated remarkable mucus and biofilm penetration capabilities, thereby overcoming one of the adaptive drug resistance mechanisms. MCIP/(PEG-PCL)/PLL NPs improved the outer and inner membrane permeability and inhibited the expression of the efflux pump MexAB-OprM gene in MDR-P. aeruginosa, thereby overcoming mechanisms of both intrinsic and acquired drug resistance. Meanwhile, the nanoparticles demonstrated an ability to reduce repeated infections with MDR-P. aeruginosa. Additionally, the bacterial burden in the lungs of mice treated with MCIP/(PEG-PCL)/PLL NPs was significantly lower than that in the MXC group, resulting in a 99% clearance rate. Notably, MCIP/(PEG-PCL)/PLL NPs showed no toxicity toward BEAS-2B cells or RAW 267.4 cells, nor did they adversely affect pulmonary function or major organs. This study demonstrated the potential of the nanodrug delivery system composed of the antibiotic moxifloxacin and the antibacterial peptide ε-PLL in addressing the clinical challenges of treating chronic pulmonary infections caused by MDR-P. aeruginosa.
Collapse
Affiliation(s)
- Yujun Chen
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Mao Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jieyi Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuan Liao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Pengyu Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yingying Sun
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shihui Yu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yongxiang Zhao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Haiyan Hu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
24
|
Yang L, Liang E, Gao Y. Modeling and simulation of distribution and drug resistance of major pathogens in patients with respiratory system infections. BMC Infect Dis 2025; 25:138. [PMID: 39881259 PMCID: PMC11780821 DOI: 10.1186/s12879-025-10549-7] [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: 10/16/2024] [Accepted: 01/23/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Respiratory tract infections (RTIs) are one of the leading causes of morbidity and mortality worldwide. The increase in antimicrobial resistance in respiratory pathogens poses a major challenge to the effective management of these infections. OBJECTIVE To investigate the distribution of major pathogens of RTIs and their antimicrobial resistance patterns in a tertiary care hospital and to develop a mathematical model to explore the relationship between pathogen distribution and antimicrobial resistance. METHODS Five hundred patients with RTIs were included in the study and 475 bacterial strains were isolated from their respiratory specimens. Antimicrobial susceptibility testing and analysis of influencing factors were performed. A mathematical model was developed to simulate the relationship between pathogen distribution and drug resistance. RESULTS The most common pathogens were Streptococcus pneumoniae (30%), Haemophilus influenzae (20%), Pseudomonas aeruginosa (15%), Staphylococcus aureus (10%) and Klebsiella pneumoniae (10%). The distribution of pathogens varied according to age group and type of RTIs, with higher proportions of Pseudomonas aeruginosa and Staphylococcus aureus in hospital-acquired and ventilator-associated pneumonia. Isolated pathogens showed high and increasing rates of resistance to commonly used antibiotics. Model simulations suggest that a shift in the distribution of pathogens toward more resistant strains may lead to a significant increase in overall resistance rates, even if antibiotic use patterns remain unchanged. CONCLUSION This study emphasizes the importance of regular monitoring of respiratory pathogen distribution and antimicrobial resistance patterns and the need for a comprehensive approach to managing RTIs, including implementation of antibiotic stewardship programs, infection control measures, and development of new therapies.
Collapse
Affiliation(s)
- Li Yang
- Department of Respiratory Medicine, Anting Hospital of Jiading District, 1060 Hejing Road, Anting Town, Jiading District, Shanghai, 201805, China.
| | - Ermin Liang
- Department of Respiratory Medicine, Anting Hospital of Jiading District, 1060 Hejing Road, Anting Town, Jiading District, Shanghai, 201805, China
| | - Yali Gao
- Department of Respiratory Medicine, Anting Hospital of Jiading District, 1060 Hejing Road, Anting Town, Jiading District, Shanghai, 201805, China
| |
Collapse
|
25
|
Ilyina V, Gatina A, Trizna E, Siniagina M, Yadykova L, Ivannikova A, Ozhegov G, Zhuravleva D, Fedorova M, Gorshkova A, Evseev P, Drucker V, Bogachev M, Validov S, Kharitonova M, Kayumov A. New Bacteriophage Pseudomonas Phage Ka2 from a Tributary Stream of Lake Baikal. Viruses 2025; 17:189. [PMID: 40006944 PMCID: PMC11861027 DOI: 10.3390/v17020189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/20/2025] [Accepted: 01/25/2025] [Indexed: 02/27/2025] Open
Abstract
Pseudomonas aeruginosa, an opportunistic pathogen, causes various biofilm-associated infections like pneumonia, infections in cystic fibrosis patients, and urinary tract and burn infections with high morbidity and mortality, as well as low treatment efficacy due to the extremely wide spread of isolates with multidrug resistance. Here, we report the new bacteriophage Pseudomonas phage Ka2 isolated from a tributary stream of Lake Baikal and belonging to the Pbunavirus genus. Transmission electron microscopy resolved that Pseudomonas phage Ka2 has a capsid of 57 ± 9 nm and a contractile and inflexible tail of 115 ± 10 nm in the non-contracted state. The genome consists of 66,310 bp with a GC content of 55% and contains 96 coding sequences. Among them, 52 encode proteins have known functions, and none of them are potentially associated with lysogeny. The bacteriophage lyses 21 of 30 P. aeruginosa clinical isolates and decreases the MIC of amikacin, gentamicin, and cefepime up to 16-fold and the MIC of colistin up to 32-fold. When treating the biofilms with Ka2, the biomass was reduced by twice, and up to a 32-fold decrease in the antibiotics MBC against biofilm-embedded cells was achieved by the combination of Ka2 with cefepime for the PAO1 strain, along with a decrease of up to 16-fold with either amikacin or colistin for clinical isolates. Taken together, these data characterize the new Pseudomonas phage Ka2 as a promising tool for the combined treatment of infections associated with P. aeruginosa biofilms.
Collapse
Affiliation(s)
- Valeriya Ilyina
- Institute of Fundamental Biology and Medicine, Kazan Federal University, 420012 Kazan, Russia; (V.I.); (A.G.); (E.T.); (M.S.); (L.Y.); (A.I.); (G.O.); (D.Z.); (M.F.); (M.K.)
| | - Alina Gatina
- Institute of Fundamental Biology and Medicine, Kazan Federal University, 420012 Kazan, Russia; (V.I.); (A.G.); (E.T.); (M.S.); (L.Y.); (A.I.); (G.O.); (D.Z.); (M.F.); (M.K.)
| | - Elena Trizna
- Institute of Fundamental Biology and Medicine, Kazan Federal University, 420012 Kazan, Russia; (V.I.); (A.G.); (E.T.); (M.S.); (L.Y.); (A.I.); (G.O.); (D.Z.); (M.F.); (M.K.)
| | - Maria Siniagina
- Institute of Fundamental Biology and Medicine, Kazan Federal University, 420012 Kazan, Russia; (V.I.); (A.G.); (E.T.); (M.S.); (L.Y.); (A.I.); (G.O.); (D.Z.); (M.F.); (M.K.)
| | - Liudmila Yadykova
- Institute of Fundamental Biology and Medicine, Kazan Federal University, 420012 Kazan, Russia; (V.I.); (A.G.); (E.T.); (M.S.); (L.Y.); (A.I.); (G.O.); (D.Z.); (M.F.); (M.K.)
| | - Anastasiya Ivannikova
- Institute of Fundamental Biology and Medicine, Kazan Federal University, 420012 Kazan, Russia; (V.I.); (A.G.); (E.T.); (M.S.); (L.Y.); (A.I.); (G.O.); (D.Z.); (M.F.); (M.K.)
| | - Georgiy Ozhegov
- Institute of Fundamental Biology and Medicine, Kazan Federal University, 420012 Kazan, Russia; (V.I.); (A.G.); (E.T.); (M.S.); (L.Y.); (A.I.); (G.O.); (D.Z.); (M.F.); (M.K.)
| | - Daria Zhuravleva
- Institute of Fundamental Biology and Medicine, Kazan Federal University, 420012 Kazan, Russia; (V.I.); (A.G.); (E.T.); (M.S.); (L.Y.); (A.I.); (G.O.); (D.Z.); (M.F.); (M.K.)
| | - Marina Fedorova
- Institute of Fundamental Biology and Medicine, Kazan Federal University, 420012 Kazan, Russia; (V.I.); (A.G.); (E.T.); (M.S.); (L.Y.); (A.I.); (G.O.); (D.Z.); (M.F.); (M.K.)
| | - Anna Gorshkova
- Limnological Institute of the Siberian Branch of the Russian Academy of Sciences, 664033 Irkutsk, Russia; (A.G.); (V.D.)
| | - Peter Evseev
- Laboratory of Molecular Microbiology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Valentin Drucker
- Limnological Institute of the Siberian Branch of the Russian Academy of Sciences, 664033 Irkutsk, Russia; (A.G.); (V.D.)
| | - Mikhail Bogachev
- Biomedical Engineering Research Centre, St. Petersburg Electrotechnical University, 197022 St. Petersburg, Russia;
| | - Shamil Validov
- Laboratory of Molecular Genetics and Microbiology Methods, Kazan Scientific Center of the Russian Academy of Sciences, 420111 Kazan, Russia;
| | - Maya Kharitonova
- Institute of Fundamental Biology and Medicine, Kazan Federal University, 420012 Kazan, Russia; (V.I.); (A.G.); (E.T.); (M.S.); (L.Y.); (A.I.); (G.O.); (D.Z.); (M.F.); (M.K.)
| | - Airat Kayumov
- Institute of Fundamental Biology and Medicine, Kazan Federal University, 420012 Kazan, Russia; (V.I.); (A.G.); (E.T.); (M.S.); (L.Y.); (A.I.); (G.O.); (D.Z.); (M.F.); (M.K.)
| |
Collapse
|
26
|
Chang TH, Pourtois JD, Haddock NL, Furkuawa D, Kelly KE, Amanatullah DF, Burgener E, Milla C, Banaei N, Bollyky PL. Prophages are Infrequently Associated With Antibiotic Resistance in Pseudomonas aeruginosa Clinical Isolates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.02.595912. [PMID: 38895396 PMCID: PMC11185549 DOI: 10.1101/2024.06.02.595912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Lysogenic bacteriophages can integrate their genome into the bacterial chromosome in the form of a prophage and can promote genetic transfer between bacterial strains in vitro . However, the contribution of lysogenic phages to the incidence of antimicrobial resistance (AMR) in clinical settings is poorly understood. Here, in a set of 186 clinical isolates of Pseudomonas aeruginosa collected from respiratory cultures from 82 patients with cystic fibrosis (CF), we evaluate the links between prophage counts and both genomic and phenotypic resistance to six anti-pseudomonal antibiotics: tobramycin, colistin, ciprofloxacin, meropenem, aztreonam, and piperacillin-tazobactam. We identified 239 different prophages in total. We find that P. aeruginosa isolates contain on average 3.06 +/- 1.84 (SD) predicted prophages. We find no significant association between the number of prophages per isolate and the minimum inhibitory concentration (MIC) for any of these antibiotics. We then investigate the relationship between particular prophages and AMR. We identify a single lysogenic phage associated with phenotypic resistance to the antibiotic tobramycin and, consistent with this association, we observe that AMR genes associated with resistance to tobramycin are more likely to be found when this prophage is present. However we find that they are not encoded directly on prophage sequences. Additionally, we identify a single prophage statistically associated with ciprofloxacin resistance but do not identify any genes associated with ciprofloxacin phenotypic resistance. These findings suggest that prophages are only infrequently associated with the AMR genes in clinical isolates of P. aeruginosa . Importance Antibiotic-resistant infections of Pseudomonas aeruginosa , a leading pathogen in patients with Cystic Fibrosis (CF) are a global health threat. While lysogenic bacteriophages are known to facilitate horizontal gene transfer, their role in promoting antibiotic resistance in clinical settings remains poorly understood. In our analysis of 186 clinical isolates of P. aeruginosa from CF patients, we find that prophage abundance does not predict phenotypic resistance to key antibiotics but that specific prophages are infrequently associated with tobramycin resistance genes. In addition, we do not find antimicrobial resistance (AMR) genes encoded directly on prophages. These results highlight that while phages can be associated with AMR, phage-mediated AMR transfer may be rare in clinical isolates and difficult to identify. This work is important for future efforts on mitigating AMR in Cystic Fibrosis and other vulnerable populations affected by Pseudomonas aeruginosa infections and advances our understanding of bacterial-phage dynamics in clinical infections.
Collapse
|
27
|
Montero MM, Domene-Ochoa S, Prim N, Ferola E, López-Causapé C, Echeverria D, Morisaki MFA, Vega-Toribio V, Sorlí L, Luque S, Padilla E, Oliver A, Horcajada JP. Synergistic efficacy of ceftazidime/avibactam and aztreonam against carbapenemase-producing Pseudomonas aeruginosa: insights from the hollow-fiber infection model. Infect Dis (Lond) 2025; 57:81-88. [PMID: 39212630 DOI: 10.1080/23744235.2024.2396882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/09/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Combination therapy is an attractive therapeutic option for extensively drug-resistant (XDR) Pseudomonas aeruginosa infections. Existing data support the combination of aztreonam and ceftazidime/avibactam (CZA) against class serine-β-lactamase (SBL)- and metallo-β-lactamase (MBL) - producing Enterobacterales. However, data about that combination against SBL- and MBL-producing P. aeruginosa are scarce. The objective of the study was to assess the in vitro activity of CZA and aztreonam alone and in combination against SBL- and MBL-producing XDR P. aeruginosa isolates. METHODS The combination was analyzed by means of the hollow-fiber infection model in three selected carbapenemase-producing P. aeruginosa isolates that were representative of the three most common XDRP. aeruginosa high-risk clones (ST175, ST111, ST235) responsible for global nosocomial infection outbreaks. RESULTS The three isolates were nonsusceptible to CZA and nonsusceptible to aztreonam. In the dynamic hollow-fiber infection model, the combination of CZA plus aztreonam exerts a bactericidal effect on the isolates, regardless of their resistance mechanism and demonstrates synergistic interactions against three isolates, achieving a bacterial reduction of 5.07 log10 CFU/ml, 5.2 log10 CFU/ml and 4 log10 CFU/ml, respectively. CONCLUSION The combination of CZA and aztreonam significantly enhanced the in vitro efficacy against XDR P. aeruginosa isolates compared to each monotherapy. This improvement suggests that the combination could serve as a feasible treatment alternative for infections caused by carbapenemase-producing XDR P. aeruginosa, especially in scenarios where no other treatment options are available.
Collapse
Affiliation(s)
- María M Montero
- Infectious Diseases Service, Hospital del Mar, Barcelona, Spain. Infectious Pathology and Antimicrobials Research Group (IPAR), Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra Barcelona, Barcelona, Spain
- CIBER of Infectious Diseases (CIBERINFEC CB21/13/00002 and CB21/13/00099), Institute of Health Carlos III, Madrid, Spain
| | - Sandra Domene-Ochoa
- Infectious Diseases Service, Hospital del Mar, Barcelona, Spain. Infectious Pathology and Antimicrobials Research Group (IPAR), Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Núria Prim
- MicrobiologyService, Laboratori de Referència de Catalunya, Barcelona, Spain
| | - Eliana Ferola
- Infectious Diseases Service, Hospital del Mar, Barcelona, Spain. Infectious Pathology and Antimicrobials Research Group (IPAR), Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Carla López-Causapé
- Servicio de Microbiología y Unidad de Investigación, Hospital Son Espases, IdISBa, Palma de Mallorca, Spain
| | | | | | | | - Luisa Sorlí
- Infectious Diseases Service, Hospital del Mar, Barcelona, Spain. Infectious Pathology and Antimicrobials Research Group (IPAR), Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra Barcelona, Barcelona, Spain
- CIBER of Infectious Diseases (CIBERINFEC CB21/13/00002 and CB21/13/00099), Institute of Health Carlos III, Madrid, Spain
| | - Sonia Luque
- CIBER of Infectious Diseases (CIBERINFEC CB21/13/00002 and CB21/13/00099), Institute of Health Carlos III, Madrid, Spain
- PharmacyService, Hospital del Mar, Barcelona, Spain
| | - Eduardo Padilla
- MicrobiologyService, Laboratori de Referència de Catalunya, Barcelona, Spain
| | - Antonio Oliver
- CIBER of Infectious Diseases (CIBERINFEC CB21/13/00002 and CB21/13/00099), Institute of Health Carlos III, Madrid, Spain
- Servicio de Microbiología y Unidad de Investigación, Hospital Son Espases, IdISBa, Palma de Mallorca, Spain
| | - Juan P Horcajada
- Infectious Diseases Service, Hospital del Mar, Barcelona, Spain. Infectious Pathology and Antimicrobials Research Group (IPAR), Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra Barcelona, Barcelona, Spain
- CIBER of Infectious Diseases (CIBERINFEC CB21/13/00002 and CB21/13/00099), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
28
|
Xu X, Meng L, Li J, Zhang Y, Liu B, Jiang W, Hao C. Analysis of the potentially pathogenic bacteria of lower respiratory tract infections in children per-, during and post-COVID-19: a retrospective study. Eur J Clin Microbiol Infect Dis 2025; 44:167-180. [PMID: 39556174 DOI: 10.1007/s10096-024-04991-9] [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: 09/30/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic has caused significant changes in lower respiratory tract infections (LRTIs). This study aimed to characterize potentially pathogenic bacterial infections in paediatric patients hospitalized for LRTIs per-, during and post-COVID-19. METHODS Sputum culture data from 85,659 children with LRTIs at the Children's Hospital of Soochow University from January 2016 to May 2024 were analyzed for eight bacteria: Streptococcus pneumoniae, Haemophilus influenzae, Staphylococcus aureus, Moraxella catarrhalis, Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, and Acinetobacter baumannii. The data during the pandemic (2020-2022, during COVID-19) and after the pandemic (January 2023-May 2024, post-COVID-19) were compared with those before the pandemic (2016-2019). RESULTS Overall, 85,659 children with LRTIs were enrolled. Of these, 42,567 cases (49.7%) were diagnosed in the pre-COVID-19 period, 22,531 cases (26.3%) during the COVID-19 period and 20,561 cases (24.0%) in the post-COVID-19 period. The overall positive rate for pathogenic bacteria was 37.1%, with the top three being S. pneumoniae (14.5%), H. influenzae (12.1%) and S. aureus (6.5%). Compared to the average pre-COVID-19 levels, the bacterial pathogen positive rate decreased by 3.5% during the COVID-19 period (OR: 0.94, 95% CI: 0.91-0.98) and by 23.4% in the post-COVID-19 period (OR: 0.66, 95% CI: 0.64-0.69). During the COVID-19 period, the positive rates for S. pneumoniae, H. influenzae, E. coli, K. pneumoniae and mixed infections decreased by 11.7%, 35.3%, 22.2%, 33.3% and 45.7% respectively, while the positive rates for S. aureus, M. catarrhalis and P. aeruginosa increased by 21.7%, 44.7% and 25% respectively. In the post-COVID-19 period, the positive rates for S. pneumoniae, H. influenzae, E. coli, P. aeruginosa, K. pneumoniae, A. baumannii and mixed infections decreased by 50.0%, 7.4%, 22.2%, 50.0%, 44.4%, 60.0% and 32.6% respectively, while there was no statistical change in the positive rates for S. aureus and M. catarrhalis. Bacteria case detection decreases in 2020 (67.0%), 2021 (60.5%), 2022 (76.3%) and 2023 (72.7%) compared to predicted cases. CONCLUSIONS Measures to restrict COVID-19 as a driver of declining bacterial positive rates. Respiratory bacteria in children are change across COVID-19 phases, age groups and seasons. After COVID-19, clinicians should continue to increase surveillance for pathogenic bacteria, especially drug-resistant flora.
Collapse
Affiliation(s)
- Xuena Xu
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Lingjian Meng
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
- Department of Pediatrics, Xuzhou Medical University Affiliated Hospital, Xuzhou, China
| | - Jiaoyang Li
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Yizhu Zhang
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Bingjie Liu
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Wujun Jiang
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China.
| | - Chuangli Hao
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
29
|
Zahorska E, Denig LM, Lienenklaus S, Kuhaudomlarp S, Tschernig T, Lipp P, Munder A, Gillon E, Minervini S, Verkhova V, Imberty A, Wagner S, Titz A. High-Affinity Lectin Ligands Enable the Detection of Pathogenic Pseudomonas aeruginosa Biofilms: Implications for Diagnostics and Therapy. JACS AU 2024; 4:4715-4728. [PMID: 39735928 PMCID: PMC11672137 DOI: 10.1021/jacsau.4c00670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 12/31/2024]
Abstract
Pseudomonas aeruginosa is a critical priority pathogen and causes life-threatening acute and biofilm-associated chronic infections. The choice of suitable treatment for complicated infections requires lengthy culturing for species identification from swabs or an invasive biopsy. To date, no fast, pathogen-specific diagnostic tools for P. aeruginosa infections are available. Here, we present the noninvasive pathogen-specific detection of P. aeruginosa using novel fluorescent probes that target the bacterial biofilm-associated lectins LecA and LecB. Several glycomimetic probes were developed to target these extracellular lectins and demonstrated to stain P. aeruginosa biofilms in vitro. Importantly, for the targeting of LecA an activity boost to low-nanomolar affinity could be achieved, which is essential for in vivo application. In vitro, the nanomolar divalent LecA-targeted imaging probe accumulated effectively in biofilms under flow conditions, independent of the fluorophore identity. Investigation of these glycomimetic imaging probes in a murine lung infection model and fluorescence imaging revealed accumulation at the infection site. These findings demonstrate the use of LecA- and LecB-targeting probes for the imaging of P. aeruginosa infections and suggest their potential as pathogen-specific diagnostics to accelerate the start of the appropriate treatment.
Collapse
Affiliation(s)
- Eva Zahorska
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| | - Lisa Marie Denig
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| | - Stefan Lienenklaus
- Hannover
Medical School, Institute of Laboratory
Animal Science, Hannover 30625, Germany
| | - Sakonwan Kuhaudomlarp
- Université
Grenoble Alpes, CNRS, CERMAV, Grenoble 38000, France
- Department
of Biochemistry, Faculty of Science, Mahidol
University, Bangkok 10400, Thailand
- Center
for Excellence in Protein and Enzyme Technology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Thomas Tschernig
- Medical
Faculty of Saarland University, Institute
of Anatomy and Cell Biology, Homburg/Saar, D-66421, Germany
| | - Peter Lipp
- Center
for Molecular Signaling (PZMS), Medical
Faculty of Saarland University, Homburg/Saar D-66421, Germany
| | - Antje Munder
- Department
of Pediatric Pneumology, Allergology and
Neonatology, Hannover Medical School, Carl Neuberg-Str. 1, Hannover D-30625, Germany
- Biomedical
Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover D-30625, Germany
| | - Emilie Gillon
- Université
Grenoble Alpes, CNRS, CERMAV, Grenoble 38000, France
| | - Saverio Minervini
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
| | - Varvara Verkhova
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| | - Anne Imberty
- Université
Grenoble Alpes, CNRS, CERMAV, Grenoble 38000, France
| | - Stefanie Wagner
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| | - Alexander Titz
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| |
Collapse
|
30
|
Baumgarten J, Schneider P, Thiemann M, Zimmermann M, Diederich C, Blankenfeldt W, Kunick C. Substrate-Based Ligand Design for Phenazine Biosynthesis Enzyme PhzF. ChemMedChem 2024; 19:e202400466. [PMID: 39163032 DOI: 10.1002/cmdc.202400466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024]
Abstract
The phenazine pyocyanin is an important virulence factor of the pathogen Pseudomonas aeruginosa, which is on the WHO list of antibiotic resistant "priority pathogens". In this study the isomerase PhzF, a key bacterial enzyme of the pyocyanin biosynthetic pathway, was investigated as a pathoblocker target. The aim of the pathoblocker strategy is to reduce the virulence of the pathogen without killing it, thus preventing the rapid development of resistance. Based on crystal structures of PhzF, derivatives of the inhibitor 3-hydroxyanthranilic acid were designed. Co-crystal structures of the synthesized derivatives with PhzF revealed spacial limitations of the binding pocket of PhzF in the closed conformation. In contrast, ligands aligned to the open conformation of PhzF provided more room for structural modifications. The intrinsic fluorescence of small 3-hydroxyanthranilic acid derivatives enabled direct affinity determinations using FRET assays. The analysis of structure-activity relationships showed that the carboxylic acid moiety is essential for binding to the target enzyme. The results of this study provide fundamental structural insights that will be useful for the design of PhzF-inhibitors.
Collapse
Affiliation(s)
- Janosch Baumgarten
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstraße 55, Braunschweig, 38106, Germany
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Str. 35a, Braunschweig, 38106, Germany
- Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, Braunschweig, 38124, Germany
| | - Philipp Schneider
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Str. 35a, Braunschweig, 38106, Germany
- Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, Braunschweig, 38124, Germany
| | - Marie Thiemann
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstraße 55, Braunschweig, 38106, Germany
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Str. 35a, Braunschweig, 38106, Germany
- Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, Braunschweig, 38124, Germany
| | - Moritz Zimmermann
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstraße 55, Braunschweig, 38106, Germany
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Str. 35a, Braunschweig, 38106, Germany
- Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, Braunschweig, 38124, Germany
| | - Christina Diederich
- Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, Braunschweig, 38124, Germany
| | - Wulf Blankenfeldt
- Structure and Function of Proteins (SFPR), Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, Braunschweig, 38124, Germany
- Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universität Braunschweig, Rebenring 56, Braunschweig, 38106, Germany
| | - Conrad Kunick
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstraße 55, Braunschweig, 38106, Germany
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Str. 35a, Braunschweig, 38106, Germany
| |
Collapse
|
31
|
Tigabie M, Assefa M, Gashaw Y, Amare A, Ambachew A, Biset S, Moges F. Prevalence and antibiotic resistance patterns of Pseudomonas aeruginosa and Acinetobacter baumannii strains isolated from chicken droppings on poultry farms in Gondar city, Northwest Ethiopia. SCIENCE IN ONE HEALTH 2024; 4:100099. [PMID: 39926021 PMCID: PMC11803164 DOI: 10.1016/j.soh.2024.100099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/28/2024] [Indexed: 02/11/2025]
Abstract
Background Pseudomonas aeruginosa and Acinetobacter baumannii are common nosocomial pathogens in hospital settings. Recently, they have also been found in non-hospital environments, such as poultry farms. While most studies in Ethiopia have focused on these bacteria's antibiotic resistance patterns in hospitals, information regarding their prevalence and resistance in veterinary settings, particularly poultry farms, is limited. This study aimed to assess the prevalence and antibiotic resistance patterns of P. aeruginosa and A. baumannii isolated from chicken droppings on poultry farms. Methods A cross-sectional study was conducted from March 2022 to June 2022. A total of 87 poultry farms were included in this study, and pooled chicken dropping samples were collected. The samples were subsequently transferred to buffered peptone water and cultured on MacConkey agar. Species of the isolates were identified via routine biochemical tests, including oxidase, catalase, urease, Simon's citrate, sulfide indole motility medium, triple sugar iron agar and growth at temperatures of 37 °C and 42 °C. The Kirby-Bauer disk diffusion technique was used for antibiotic susceptibility testing. The data were entered into EpiData version 4.6 and then exported to SPSS version 26 for analysis. Fisher's exact test was used to observe an appropriate association between independent variables and the occurrence of isolates. The results are presented in the text, figures and tables. Results Among the 87 poultry farms, 41 (47.1 %) were positive for Pseudomonas and Acinetobacter. Among these strains, 24 (27.6 %) P. aeruginosa strains and 13 (14.9 %) A. baumannii strains were recovered. P. aeruginosa showed complete resistance to tetracycline (24, 100.0 %) and trimethoprim-sulfamethoxazole (24, 100.0 %). Additionally, there was a high rate of resistance to ciprofloxacin (13, 54.2 %) and amikacin (12, 50.0 %). Similarly, 13 (100.0 %) A. baumannii isolates were resistant to tetracycline, and 12 (92.3 %) were resistant to trimethoprim-sulfamethoxazole. However, both isolates presented lower resistance rates to piperacillin-tazobactam (4, 9.8 %) and cefepime (7, 17.1 %). Both A. baumannii and P. aeruginosa exhibited multidrug resistance in 10/13 (76.9 %) and 16/24 (66.7 %) of the strains, respectively. The overall prevalence of multidrug resistance in this study was 28/41 (68.3 %). Conclusion This study demonstrated that poultry farms may be potential reservoirs for P. aeruginosa and A. baumannii, including antibiotic-resistant strains. This is a significant concern to public health because poultry farmers may be contaminated, increasing their dissemination to the community. Therefore, poultry farmers should improve sanitation and reduce the misuse and overuse of antibiotics at poultry farms.
Collapse
Affiliation(s)
- Mitkie Tigabie
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Ethiopia
| | - Muluneh Assefa
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Ethiopia
| | - Yalewayker Gashaw
- Department of Medical Laboratory Sciences, College of Health Sciences, Woldia University, Ethiopia
| | - Azanaw Amare
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Ethiopia
| | - Aklilu Ambachew
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Ethiopia
| | - Sirak Biset
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Ethiopia
| | - Feleke Moges
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Ethiopia
| |
Collapse
|
32
|
Alimoghadam S, Eslami A, Alimoghadam R, Bahrami Mianrood I, Azizmohammad Looha M, Khodadadi S, Shokouhi S, Alavi Darazam I. The frequency of AmpC overproduction, OprD downregulation and OprM efflux pump expression in Pseudomonas aeruginosa: A comprehensive meta-analysis. J Glob Antimicrob Resist 2024; 39:159-169. [PMID: 39303871 DOI: 10.1016/j.jgar.2024.08.014] [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: 08/20/2023] [Revised: 06/06/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024] Open
Abstract
OBJECTIVES Pseudomonas aeruginosa is a major opportunistic pathogen responsible for a wide range of infections. The emergence of antibiotic resistance in this pathogen poses a significant public health challenge. This study aims to conduct a comprehensive meta-analysis of studies conducted in Iran to determine the frequency of key antibiotic resistance mechanisms in Pseudomonas aeruginosa and their association with multidrug-resistant and extensively drug-resistant strains or pandrug-resistant strains. METHODS Systematic database searches encompassing literature up to June 2023 were undertaken. The selected studies centered on OprD downregulation, efflux pump (mexAB-OprM, mexXY-OprM) expression, and AmpC overproduction. Extracted data were synthesised in a meta-analysis for pooled frequency determination of each resistance mechanism. RESULTS In total, 24 studies were included. OprD downregulation exhibited a pooled frequency of 61%. Efflux pump component frequency ranged from 48% to 77.5%. AmpC overproduction was identified in 29.1% of isolates. Polymyxin B and colistin demonstrated lower antibiotic resistance rates, with pooled frequency of 1% and 1.6%, respectively. Conversely, resistance to other antibiotics ranged widely, with pooled frequency spanning 38.4% to 98.2%. CONCLUSIONS This study underscores the concerning frequency of diverse antibiotic resistance mechanisms in Pseudomonas aeruginosa strains from Iran. Concurrent OprD downregulation, mexAB, mexXY, OprM expression, and AmpC overproduction highlight the urgent need for stringent infection control and prudent antibiotic usage to curb the dissemination of these resistant strains. PROSPERO CRD42022379311.
Collapse
Affiliation(s)
- Shaya Alimoghadam
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Infectious Diseases, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Arvin Eslami
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Infectious Diseases, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Rojina Alimoghadam
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Infectious Diseases, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ibrahim Bahrami Mianrood
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Infectious Diseases, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mehdi Azizmohammad Looha
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Sanaz Khodadadi
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Infectious Diseases, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Shervin Shokouhi
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Infectious Diseases, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Research Center for Antibiotic Stewardship and Antimicrobial Resistance, Tehran University of Medical Sciences, Tehran, Iran
| | - Ilad Alavi Darazam
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Infectious Diseases, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Research Center for Antibiotic Stewardship and Antimicrobial Resistance, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Edward EA, El Shehawy MR, Abouelfetouh A, Aboulmagd E. Phenotypic and molecular characterization of extended spectrum- and metallo- beta lactamase producing Pseudomonas aeruginosa clinical isolates from Egypt. Infection 2024; 52:2399-2414. [PMID: 38824475 PMCID: PMC11621155 DOI: 10.1007/s15010-024-02297-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/13/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Antimicrobial resistance among Pseudomonas aeruginosa (P. aeruginosa), a leading cause of nosocomial infections worldwide, is escalating. This study investigated the prevalence of extended-spectrum β-lactamases (ESBLs) and metallo-β-lactamases (MBLs) among 104 P. aeruginosa clinical isolates from Alexandria Main University Hospital, Alexandria, Egypt. METHODS Antimicrobial susceptibility testing was performed using agar dilution technique, or broth microdilution method in case of colistin. ESBL and MBL prevalence was assessed phenotypically and genotypically using polymerase chain reaction (PCR). The role of plasmids in mediating resistance to extended-spectrum β-lactams was studied via transformation technique using plasmids isolated from ceftazidime-resistant isolates. RESULTS Antimicrobial susceptibility testing revealed alarming resistance rates to carbapenems, cephalosporins, and fluoroquinolones. Using PCR as the gold standard, phenotypic methods underestimated ESBL production while overestimating MBL production. Eighty-five isolates (81.7%) possessed only ESBL encoding genes, among which 69 isolates harbored a single ESBL gene [blaOXA-10 (n = 67) and blaPER (n = 2)]. Four ESBL-genotype combinations were detected: blaPER + blaOXA-10 (n = 8), blaVEB-1 + blaOXA-10 (n = 6), blaPSE + blaOXA-10 (n = 1), and blaPER + blaVEB-1 + blaOXA-10 (n = 1). Three isolates (2.9%) possessed only the MBL encoding gene blaVIM. Three ESBL + MBL- genotype combinations: blaOXA-10 + blaAIM, blaOXA-10 + blaVIM, and blaPER + blaOXA-10 + blaAIM were detected in 2, 1 and 1 isolate(s), respectively. Five plasmid preparations harboring blaVEB-1 and blaOXA-10 were successfully transformed into chemically competent Escherichia coli DH5α with transformation efficiencies ranging between 6.8 × 10 3 and 3.7 × 10 4 CFU/μg DNA plasmid. Selected tested transformants were ceftazidime-resistant and harbored plasmids carrying blaOXA-10. CONCLUSIONS The study highlights the importance of the expeditious characterization of ESBLs and MBLs using genotypic methods among P. aeruginosa clinical isolates to hinder the development and dissemination of multidrug resistant strains.
Collapse
Affiliation(s)
- Eva A Edward
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, El-Khartoom Square, Azarita, Alexandria, Egypt.
| | - Marwa R El Shehawy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, El-Khartoom Square, Azarita, Alexandria, Egypt
| | - Alaa Abouelfetouh
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, El-Khartoom Square, Azarita, Alexandria, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Elsayed Aboulmagd
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, El-Khartoom Square, Azarita, Alexandria, Egypt
- College of Pharmacy, Arab Academy for Science, Technology and Maritime, Alamein Branch, Alamein, Egypt
| |
Collapse
|
34
|
Li R, Zhu X, Zhang P, Wu X, Jin Q, Pan J. Ser/Thr protein kinase Stk1 phosphorylates the key transcriptional regulator AlgR to modulate virulence and resistance in Pseudomonas aeruginosa. Virulence 2024; 15:2367649. [PMID: 38898809 PMCID: PMC11197903 DOI: 10.1080/21505594.2024.2367649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/09/2024] [Indexed: 06/21/2024] Open
Abstract
Pseudomonas aeruginosa is one of the leading causes of nosocomial infections worldwide and has emerged as a serious public health threat, due in large part to its multiple virulence factors and remarkable resistance capabilities. Stk1, a eukaryotic-type Ser/Thr protein kinase, has been shown in our previous work to be involved in the regulation of several signalling pathways and biological processes. Here, we demonstrate that deletion of stk1 leads to alterations in several virulence- and resistance-related physiological functions, including reduced pyocyanin and pyoverdine production, attenuated twitching motility, and enhanced biofilm production, extracellular polysaccharide secretion, and antibiotic resistance. Moreover, we identified AlgR, an important transcriptional regulator, as a substrate for Stk1, with its phosphorylation at the Ser143 site catalysed by Stk1. Intriguingly, both the deletion of stk1 and the mutation of Ser143 of AlgR to Ala result in similar changes in the above-mentioned physiological functions. Furthermore, assays of algR expression in these strains suggest that changes in the phosphorylation state of AlgR, rather than its expression level, underlie changes in these physiological functions. These findings uncover Stk1-mediated phosphorylation of AlgR as an important mechanism for regulating virulence and resistance in P. aeruginosa.
Collapse
Affiliation(s)
- Rui Li
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xuan Zhu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Pengfei Zhang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xuan Wu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Qian Jin
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jianyi Pan
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
35
|
Ferrusca Bernal D, Mosqueda J, Pérez-Sánchez G, Chávez JAC, Neri Martínez M, Rodríguez A, Carvajal-Gamez B. Loop-Mediated Isothermal Amplification Coupled with Reverse Line Blot Hybridization for the Detection of Pseudomonas aeruginosa. Microorganisms 2024; 12:2316. [PMID: 39597705 PMCID: PMC11596522 DOI: 10.3390/microorganisms12112316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/04/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Pseudomonas aeruginosa is a pathogen of critical priority importance according to the WHO. Due to its multi-resistance and expression of various virulence factors, it is the causal agent of severe healthcare-acquired infections (HAIs). Effective strategies to control infections caused by P. aeruginosa must include early and specific detection of the pathogen for early and timely antibiotic prescription. The need to develop a specific and reproducible diagnostic technique is urgent, which must often be more sensitive and faster than current clinical diagnostic methods. In this study, we implement and standardize the loop-mediated isothermal amplification (LAMP) technique, coupled with the reverse line blot hybridization (RLBH) technique for the detection of P. aeruginosa. A set of primers and probes was designed to amplify a specific region of the P. aeruginosa 16s rRNA gene. The sensitivity of the LAMP-RLBH method was 3 × 10-4 ng/μL, 1000 times more sensitive than the PCR and LAMP technique (this work), with a sensitivity of 3 × 10-3 ng/μL. The LAMP-RLBH and LAMP techniques showed specific amplification and no cross-reaction with members of the ESKAPE group and other Pseudomonas species. The present investigation provides a technique that can be easily performed in less time, achieving a faster and more reliable alternative compared to traditional microbial diagnostic methods for the detection of P. aeruginosa.
Collapse
Affiliation(s)
- Daniel Ferrusca Bernal
- Immunology and Vaccines Laboratory, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76010, Mexico; (D.F.B.); (J.M.); (M.N.M.)
| | - Juan Mosqueda
- Immunology and Vaccines Laboratory, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76010, Mexico; (D.F.B.); (J.M.); (M.N.M.)
- Cuerpo Académico, Salud Animal y Microbiología Ambiental, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76010, Mexico;
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico;
| | | | - Mónica Neri Martínez
- Immunology and Vaccines Laboratory, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76010, Mexico; (D.F.B.); (J.M.); (M.N.M.)
- Proteogenomic and Molecular Diagnosis Laboratory, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76010, Mexico
| | - Angelina Rodríguez
- Cuerpo Académico, Salud Animal y Microbiología Ambiental, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76010, Mexico;
- Maestría en Química Clínica Diagnóstica, Facultad de Química, Autonomous University of Queretaro, Queretaro 76010, Mexico
| | - Bertha Carvajal-Gamez
- Immunology and Vaccines Laboratory, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76010, Mexico; (D.F.B.); (J.M.); (M.N.M.)
- Cuerpo Académico, Salud Animal y Microbiología Ambiental, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76010, Mexico;
- Proteogenomic and Molecular Diagnosis Laboratory, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76010, Mexico
- Maestría en Química Clínica Diagnóstica, Facultad de Química, Autonomous University of Queretaro, Queretaro 76010, Mexico
| |
Collapse
|
36
|
Sanchez C, Vargas-Cuebas GG, Michaud ME, Allen RA, Morrison-Lewis KR, Siddiqui S, Minbiole KPC, Wuest WM. Highly Effective Biocides against Pseudomonas aeruginosa Reveal New Mechanistic Insights Across Gram-Negative Bacteria. ACS Infect Dis 2024; 10:3868-3879. [PMID: 39440866 PMCID: PMC11555683 DOI: 10.1021/acsinfecdis.4c00433] [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: 05/27/2024] [Revised: 08/28/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
Pseudomonas aeruginosa is a major nosocomial pathogen that persists in healthcare settings despite rigorous disinfection protocols due to intrinsic mechanisms conferring resistance. We sought to systematically assess cationic biocide efficacy against this pathogen using a panel of multidrug-resistant P. aeruginosa clinical isolates. Our studies revealed widespread resistance to commercial cationic disinfectants that are the current standard of care, raising concerns about their efficacy. To address this shortcoming, we highlight a new class of quaternary phosphonium compounds that are highly effective against all members of the panel. To understand the difference in efficacy, mechanism of action studies were carried out, which identified a discrete inner-membrane selective target. Resistance selection studies implicated the SmvRA efflux system (a transcriptionally regulated, inner membrane-associated efflux system) as a major determinant of resistance. This system is also implicated in resistance to two commercial bolaamphiphile antiseptics, octenidine and chlorhexidine, which was further validated herein. In sum, this work highlights, for the first time, a discrete inner-membrane specific mechanism for the bolaamphiphile class of disinfectants that contrasts with the prevailing model of indiscriminate membrane interactions of commercial amphiphiles paving the way for future innovations in disinfectant research.
Collapse
Affiliation(s)
- Christian
A. Sanchez
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Germán G. Vargas-Cuebas
- Department
of Microbiology and Immunology, Emory University, Atlanta, Georgia 30322, United States
| | - Marina E. Michaud
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Ryan A. Allen
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | | | - Shehreen Siddiqui
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Kevin P. C. Minbiole
- Department
of Chemistry, Villanova University, Villanova, Pennsylvania 19085, United States
| | - William M. Wuest
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
37
|
Dong L, Sun L, Yang Y, Yuan L, Gao W, Yu D, Meng Q, Shi W, Wang Q, Li Y, Zhang Y, You X, Yao K. Non-antibiotic pharmaceutical phenylbutazone binding to MexR reduces the antibiotic susceptibility of Pseudomonas aeruginosa. Microbiol Res 2024; 288:127872. [PMID: 39146705 DOI: 10.1016/j.micres.2024.127872] [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: 05/12/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024]
Abstract
Antimicrobial resistance has been an increasingly serious threat to global public health. The contribution of non-antibiotic pharmaceuticals to the development of antibiotic resistance has been overlooked. Our study found that the anti-inflammatory drug phenylbutazone could protect P. aeruginosa against antibiotic mediated killing by binding to the efflux pump regulator MexR. In this study, antibiotic activity against P. aeruginosa alone or in combination with phenylbutazone was evaluated in vitro and in vivo. Resazurin accumulation assay, transcriptomic sequencing, and PISA assay were conducted to explore the underlying mechanism for the reduced antibiotic susceptibility caused by phenylbutazone. Then EMSA, ITC, molecular dynamic simulations, and amino acid substitutions were used to investigate the interactions between phenylbutazone and MexR. We found that phenylbutazone could reduce the susceptibility of P. aeruginosa to multiple antibiotics, including parts of β-lactams, fluoroquinolones, tetracyclines, and macrolides. Phenylbutazone could directly bind to MexR, then promote MexR dissociating from the mexA-mexR intergenic region and de-repress the expression of MexAB-OprM efflux pump. The overexpressed MexAB-OprM pump resulted in the reduced antibiotic susceptibility. And the His41 and Arg21 residues of MexR were involved in the phenylbutazone-MexR interaction. We hope this study would imply the potential risk of antibiotic resistance caused by non-antibiotic pharmaceuticals.
Collapse
Affiliation(s)
- Limin Dong
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Lang Sun
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yan Yang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lin Yuan
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Wei Gao
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Dan Yu
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Qinghong Meng
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Wei Shi
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Qing Wang
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Yue Li
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Youwen Zhang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Xuefu You
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Kaihu Yao
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics, Laboratory of Infection and Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| |
Collapse
|
38
|
Gao L, Zhang K, Wang Y, Qin C, Zhang Y, Liu Y, Liu C, Wan Y. Curcumin-mediated photodynamic disinfection strategy with specific spectral range for mucoid Pseudomonas Aeruginosa from hospital water. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 260:113035. [PMID: 39303620 DOI: 10.1016/j.jphotobiol.2024.113035] [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: 05/21/2024] [Revised: 08/22/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Hospital water systems represent critical environments for the transmission of pathogens, including multidrug-resistant strains like mucoid Pseudomonas aeruginosa (M-PA). Conventional disinfection methods often struggle to eradicate these pathogens effectively, highlighting the need for innovative approaches. OBJECTIVE This study aimed to develop an enhanced photodynamic disinfection strategy targeting M-PA from hospital water systems, using curcumin-mediated photodynamic inactivation (PDI) with specific spectral range. METHODS An M-PA strain isolated from hospital water was subjected to photodynamic treatment using curcumin as the photosensitizer. The efficacy of different wavelengths of light and varying concentrations of curcumin, with and without Tris-EDTA adjuvants, was evaluated through bacterial enumeration, ROS level measurements, transcriptome analysis, and assessment of virulence factors and biofilm formation. In vivo experiments utilizing a DSS-induced colitis mouse model assessed the protective effects of the photodynamic treatment against M-PA infection. RESULTS Our findings demonstrated that the combination of curcumin-mediated PDI with specific spectral range effectively reduced M-PA counts in water, particularly when supplemented with Tris-EDTA. Transcriptome analysis revealed significant downregulation of virulence-related genes under sublethal photodynamic conditions. Furthermore, photodynamic treatment inhibited pyocyanin production and biofilm formation in M-PA, highlighting its potential to disrupt pathogenicity mechanisms. In vivo experiments showed that PDI attenuated M-PA-induced colitis in mice, indicating its protective efficacy. CONCLUSION This study presents a promising photodynamic disinfection strategy for combating M-PA from hospital water. By optimizing curcumin-mediated PDI with specific spectral range and adjuvants, our approach demonstrates substantial efficacy in reducing bacterial counts, inhibiting virulence factors, and preventing M-PA-associated colitis.
Collapse
Affiliation(s)
- Lei Gao
- Microbiology Institute of Shaanxi, No.76 Xiying Road, Xi'an 710043, PR China
| | - Kun Zhang
- Microbiology Institute of Shaanxi, No.76 Xiying Road, Xi'an 710043, PR China
| | - Yan Wang
- Microbiology Institute of Shaanxi, No.76 Xiying Road, Xi'an 710043, PR China
| | - Chuan Qin
- Microbiology Institute of Shaanxi, No.76 Xiying Road, Xi'an 710043, PR China
| | - Yuejuan Zhang
- Microbiology Institute of Shaanxi, No.76 Xiying Road, Xi'an 710043, PR China
| | - Ying Liu
- Microbiology Institute of Shaanxi, No.76 Xiying Road, Xi'an 710043, PR China
| | - Chengcheng Liu
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, PR China.
| | - Yi Wan
- Microbiology Institute of Shaanxi, No.76 Xiying Road, Xi'an 710043, PR China.
| |
Collapse
|
39
|
Rodriguez-Gonzalez RA, Balacheff Q, Debarbieux L, Marchi J, Weitz JS. Metapopulation model of phage therapy of an acute Pseudomonas aeruginosa lung infection. mSystems 2024; 9:e0017124. [PMID: 39230264 PMCID: PMC11562898 DOI: 10.1128/msystems.00171-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/24/2024] [Indexed: 09/05/2024] Open
Abstract
Infections caused by multidrug resistant (MDR) pathogenic bacteria are a global health threat. Bacteriophages ("phage") are increasingly used as alternative or last-resort therapeutics to treat patients infected by MDR bacteria. However, the therapeutic outcomes of phage therapy may be limited by the emergence of phage resistance during treatment and/or by physical constraints that impede phage-bacteria interactions in vivo. In this work, we evaluate the role of lung spatial structure on the efficacy of phage therapy for Pseudomonas aeruginosa infections. To do so, we developed a spatially structured metapopulation network model based on the geometry of the bronchial tree, including host innate immune responses and the emergence of phage-resistant bacterial mutants. We model the ecological interactions between bacteria, phage, and the host innate immune system at the airway (node) level. The model predicts the synergistic elimination of a P. aeruginosa infection due to the combined effects of phage and neutrophils, given the sufficient innate immune activity and efficient phage-induced lysis. The metapopulation model simulations also predict that MDR bacteria are cleared faster at distal nodes of the bronchial tree. Notably, image analysis of lung tissue time series from wild-type and lymphocyte-depleted mice revealed a concordant, statistically significant pattern: infection intensity cleared in the bottom before the top of the lungs. Overall, the combined use of simulations and image analysis of in vivo experiments further supports the use of phage therapy for treating acute lung infections caused by P. aeruginosa, while highlighting potential limits to therapy in a spatially structured environment given impaired innate immune responses and/or inefficient phage-induced lysis. IMPORTANCE Phage therapy is increasingly employed as a compassionate treatment for severe infections caused by multidrug-resistant (MDR) bacteria. However, the mixed outcomes observed in larger clinical studies highlight a gap in understanding when phage therapy succeeds or fails. Previous research from our team, using in vivo experiments and single-compartment mathematical models, demonstrated the synergistic clearance of acute P. aeruginosa pneumonia by phage and neutrophils despite the emergence of phage-resistant bacteria. In fact, the lung environment is highly structured, prompting the question of whether immunophage synergy explains the curative treatment of P. aeruginosa when incorporating realistic physical connectivity. To address this, we developed a metapopulation network model mimicking the lung branching structure to assess phage therapy efficacy for MDR P. aeruginosa pneumonia. The model predicts the synergistic elimination of P. aeruginosa by phage and neutrophils but emphasizes potential challenges in spatially structured environments, suggesting that higher innate immune levels may be required for successful bacterial clearance. Model simulations reveal a spatial pattern in pathogen clearance where P. aeruginosa are cleared faster at distal nodes of the bronchial tree than in primary nodes. Interestingly, image analysis of infected mice reveals a concordant and statistically significant pattern: infection intensity clears in the bottom before the top of the lungs. The combined use of modeling and image analysis supports the application of phage therapy for acute P. aeruginosa pneumonia while emphasizing potential challenges to curative success in spatially structured in vivo environments, including impaired innate immune responses and reduced phage efficacy.
Collapse
Affiliation(s)
- Rogelio A. Rodriguez-Gonzalez
- Interdisciplinary
Graduate Program in Quantitative Biosciences,Georgia Institute of
Technology, Atlanta,
Georgia, USA
- School of Biological
Sciences, Georgia Institute of
Technology, Atlanta,
Georgia, USA
| | - Quentin Balacheff
- CHU Félix
Guyon, Service des maladies
respiratoires, La
Réunion, France
| | - Laurent Debarbieux
- Department of
Microbiology, Institut Pasteur, Université Paris Cité,
CNRS UMR6047, Bacteriophage Bacterium
Host, Paris,
France
| | - Jacopo Marchi
- Department of Biology,
University of Maryland, College
Park, Maryland, USA
| | - Joshua S. Weitz
- Department of Biology,
University of Maryland, College
Park, Maryland, USA
- Department of Physics,
University of Maryland, College
Park, Maryland, USA
- Institut de Biologie,
École Normale Supérieure,
Paris, France
| |
Collapse
|
40
|
Rodrigues YC, Silva MJA, dos Reis HS, dos Santos PAS, Sardinha DM, Gouveia MIM, dos Santos CS, Marcon DJ, Aires CAM, Souza CDO, Quaresma AJPG, Lima LNGC, Brasiliense DM, Lima KVB. Molecular Epidemiology of Pseudomonas aeruginosa in Brazil: A Systematic Review and Meta-Analysis. Antibiotics (Basel) 2024; 13:983. [PMID: 39452249 PMCID: PMC11504043 DOI: 10.3390/antibiotics13100983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Globally, Pseudomonas aeruginosa is a high-priority opportunistic pathogen which displays several intrinsic and acquired antimicrobial resistance (AMR) mechanisms, leading to challenging treatments and mortality of patients. Moreover, its wide virulence arsenal, particularly the type III secretion system (T3SS) exoU+ virulotype, plays a crucial role in pathogenicity and poor outcome of infections. In depth insights into the molecular epidemiology of P. aeruginosa, especially the prevalence of high-risk clones (HRCs), are crucial for the comprehension of virulence and AMR features and their dissemination among distinct strains. This study aims to evaluate the prevalence and distribution of HRCs and non-HRCs among Brazilian isolates of P. aeruginosa. METHODS A systematic review and meta-analysis were conducted on studies published between 2011 and 2023, focusing on the prevalence of P. aeruginosa clones determined by multilocus sequence typing (MLST) in Brazil. Data were extracted from retrospective cross-sectional and case-control studies, encompassing clinical and non-clinical samples. The analysis included calculating the prevalence rates of various sequence types (STs) and assessing the regional variability in the distribution of HRCs and non-HRCs. RESULTS A total of 872 samples were analyzed within all studies, of which 298 (34.17%) were MLST typed, identifying 78 unique STs. HRCs accounted for 48.90% of the MLST-typed isolates, with ST277 being the most prevalent (100/298-33.55%), followed by ST244 (29/298-9.73%), ST235 (13/298-4.36%), ST111 (2/298-0.67%), and ST357 (2/298-0.67%). Significant regional variability was observed, with the Southeast region showing a high prevalence of ST277, while the North region shows a high prevalence of MLST-typed samples and HRCs. CONCLUSIONS Finally, this systematic review and meta-analysis highlight the role of P. aeruginosa clones in critical issue of AMR in P. aeruginosa in Brazil and the need of integration of comprehensive data from individual studies.
Collapse
Affiliation(s)
- Yan Corrêa Rodrigues
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | - Marcos Jessé Abrahão Silva
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Herald Souza dos Reis
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
| | - Pabllo Antonny Silva dos Santos
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Daniele Melo Sardinha
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
| | - Maria Isabel Montoril Gouveia
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | - Carolynne Silva dos Santos
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Davi Josué Marcon
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Caio Augusto Martins Aires
- Department of Health Sciences (DCS), Federal Rural University of the Semi-Arid Region (UFERSA), Av. Francisco Mota, 572-Bairro Costa e Silva, Mossoró 59625-900, RN, Brazil;
| | - Cintya de Oliveira Souza
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
| | - Ana Judith Pires Garcia Quaresma
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Luana Nepomuceno Gondim Costa Lima
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Danielle Murici Brasiliense
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| | - Karla Valéria Batista Lima
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil; (H.S.d.R.); (P.A.S.d.S.); (D.M.S.); (M.I.M.G.); (C.S.d.S.); (D.J.M.); (C.d.O.S.); (A.J.P.G.Q.); (L.N.G.C.L.); (D.M.B.)
- Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ministry of Health, Ananindeua 67030-000, PA, Brazil
- Program in Parasitic Biology in the Amazon Region (PPGBPA), State University of Pará (UEPA), Tv. Perebebuí, 2623-Marco, Belém 66087-662, PA, Brazil
| |
Collapse
|
41
|
Khan F. Multifaceted strategies for alleviating Pseudomonas aeruginosa infection by targeting protease activity: Natural and synthetic molecules. Int J Biol Macromol 2024; 278:134533. [PMID: 39116989 DOI: 10.1016/j.ijbiomac.2024.134533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/29/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024]
Abstract
Pseudomonas aeruginosa has become a top-priority pathogen in the health sector because it is ubiquitous, has high metabolic/genetic versatility, and is identified as an opportunistic pathogen. The production of numerous virulence factors by P. aeruginosa was reported to act individually or cooperatively to make them robots invasion, adherences, persistence, proliferation, and protection against host immune systems. P. aeruginosa produces various kinds of extracellular proteases such as alkaline protease, protease IV, elastase A, elastase B, large protease A, Pseudomonas small protease, P. aeruginosa aminopeptidase, and MucD. These proteases effectively allow the cells to invade and destroy host cells. Thus, inhibiting these protease activities has been recognized as a promising approach to controlling the infection caused by P. aeruginosa. The present review discussed in detail the characteristics of these proteases and their role in infection to the host system. The second part of the review discussed the recent updates on the multiple strategies for attenuating or inhibiting protease activity. These strategies include the application of natural and synthetic molecules, as well as metallic/polymeric nanomaterials. It has also been reported that a propeptide present in the middle domain of protease IV also attenuates the virulence properties and infection ability of P. aeruginosa.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
42
|
Marchi J, Ngoc Minh CN, Debarbieux L, Weitz JS. Multi-strain phage induced clearance of bacterial infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.07.611814. [PMID: 39282405 PMCID: PMC11398464 DOI: 10.1101/2024.09.07.611814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Bacteriophage (or 'phage' - viruses that infect and kill bacteria) are increasingly considered as a therapeutic alternative to treat antibiotic-resistant bacterial infections. However, bacteria can evolve resistance to phage, presenting a significant challenge to the near- and long-term success of phage therapeutics. Application of mixtures of multiple phage (i.e., 'cocktails') have been proposed to limit the emergence of phage-resistant bacterial mutants that could lead to therapeutic failure. Here, we combine theory and computational models of in vivo phage therapy to study the efficacy of a phage cocktail, composed of two complementary phages motivated by the example of Pseudomonas aeruginosa facing two phages that exploit different surface receptors, LUZ19v and PAK_P1. As confirmed in a Luria-Delbrück fluctuation test, this motivating example serves as a model for instances where bacteria are extremely unlikely to develop simultaneous resistance mutations against both phages. We then quantify therapeutic outcomes given single- or double-phage treatment models, as a function of phage traits and host immune strength. Building upon prior work showing monophage therapy efficacy in immunocompetent hosts, here we show that phage cocktails comprised of phage targeting independent bacterial receptors can improve treatment outcome in immunocompromised hosts and reduce the chance that pathogens simultaneously evolve resistance against phage combinations. The finding of phage cocktail efficacy is qualitatively robust to differences in virus-bacteria interactions and host immune dynamics. Altogether, the combined use of theory and computational analysis highlights the influence of viral life history traits and receptor complementarity when designing and deploying phage cocktails in immunocompetent and immunocompromised hosts.
Collapse
Affiliation(s)
- Jacopo Marchi
- Department of Biology, University of Maryland, College Park, MD, USA
| | - Chau Nguyen Ngoc Minh
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Bacteriophage Bacterium Host, Paris, France and Sorbonne Université, Collège Doctoral, Paris, France
| | - Laurent Debarbieux
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Bacteriophage Bacterium Host, Paris, France
| | - Joshua S. Weitz
- Department of Biology, University of Maryland, College Park, MD USA
- Department of Physics, University of Maryland, College Park, MD USA
- University of Maryland Institute for Health Computing, North Bethesda, MD and Institut de Biologie, École Normale Supérieure, Paris, France
| |
Collapse
|
43
|
Sachdeva C, Satyamoorthy K, Murali TS. Pseudomonas aeruginosa: metabolic allies and adversaries in the world of polymicrobial infections. Crit Rev Microbiol 2024:1-20. [PMID: 39225080 DOI: 10.1080/1040841x.2024.2397359] [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: 09/25/2023] [Revised: 08/10/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Pseudomonas aeruginosa (PA), an opportunistic human pathogen that is frequently linked with chronic infections in immunocompromised individuals, is also metabolically versatile, and thrives in diverse environments. Additionally, studies report that PA can interact with other microorganisms, such as bacteria, and fungi, producing unique metabolites that can modulate the host immune response, and contribute to disease pathogenesis. This review summarizes the current knowledge related to the metabolic interactions of PA with other microorganisms (Staphylococcus, Acinetobacter, Klebsiella, Enterococcus, and Candida) and human hosts, and the importance of these interactions in a polymicrobial context. Further, we highlight the potential applications of studying these metabolic interactions toward designing better diagnostic tools, and therapeutic strategies to prevent, and treat infections caused by this pathogen.
Collapse
Affiliation(s)
- Chandni Sachdeva
- Department of Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kapaettu Satyamoorthy
- Department of Cell & Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
- SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara (SDM) University, Sattur, Karnataka, India
| | - Thokur Sreepathy Murali
- Department of Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
44
|
Flores-Maldonado O, Lezcano-Domínguez CI, Dávila-Aviña J, González GM, Ríos-López AL. Methyl gallate attenuates virulence and decreases antibiotic resistance in extensively drug-resistant Pseudomonasaeruginosa. Microb Pathog 2024; 194:106830. [PMID: 39084307 DOI: 10.1016/j.micpath.2024.106830] [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: 04/04/2024] [Revised: 07/19/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024]
Abstract
Pseudomonas aeruginosa infections have become a serious threat to public health due to the increasing emergence of extensively antibiotic-resistant strains and high mortality rates. Therefore, the search for new therapeutic alternatives has become crucial. In this study, the antivirulence and antibacterial activity of methyl gallate was evaluated against six clinical isolates of extensively antibiotic-resistant P. aeruginosa. Methyl gallate exhibited minimal inhibitory concentrations of 256-384 μg/mL; moreover, the use of subinhibitory concentrations of the compound inhibited biofilm formation, swimming, swarming, proteolytic activity, and pyocyanin production. Methyl gallate plus antipseudomonal antibiotics showed a synergistic effect by reduced the MICs of ceftazidime, gentamicin and meropenem. Furthermore, the potential therapeutic effect of methyl gallate was demonstrated in an infection model. This study evidenced the antivirulence and antimicrobial activity of methyl gallate as a therapeutic alternative against P. aeruginosa.
Collapse
Affiliation(s)
- Orlando Flores-Maldonado
- Universidad Autónoma de Nuevo León, Departamento de Microbiología, Facultad de Medicina y Hospital Universitario "Dr. José Eleuterio González", Monterrey, 64460, Nuevo León, Mexico
| | - Cristina I Lezcano-Domínguez
- Universidad Autónoma de Nuevo León, Departamento de Microbiología, Facultad de Medicina y Hospital Universitario "Dr. José Eleuterio González", Monterrey, 64460, Nuevo León, Mexico
| | - Jorge Dávila-Aviña
- Universidad Autónoma de Nuevo León, Departamento de Microbiología e Inmunología, Facultad de Ciencias Biológicas, San Nicolas de los Garza, 66455, Nuevo León, Mexico
| | - Gloria M González
- Universidad Autónoma de Nuevo León, Departamento de Microbiología, Facultad de Medicina y Hospital Universitario "Dr. José Eleuterio González", Monterrey, 64460, Nuevo León, Mexico
| | - Ana L Ríos-López
- Universidad Autónoma de Nuevo León, Departamento de Microbiología, Facultad de Medicina y Hospital Universitario "Dr. José Eleuterio González", Monterrey, 64460, Nuevo León, Mexico.
| |
Collapse
|
45
|
Rickard H, Cloutman-Green E, Ciric L. A microbiological survey approach to understanding the virulence factors of Pseudomonas species in healthcare sinks. J Hosp Infect 2024; 151:84-91. [PMID: 38992838 DOI: 10.1016/j.jhin.2024.06.012] [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: 02/21/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND Hospital water is involved in both the prevention and spread of healthcare-associated infections (HCAIs). Handwashing is key to reducing the transmission of pathogens, yet numerous outbreaks have been found to be caused by organisms within sinks, taps and showers. Pseudomonas aeruginosa and increasingly non-aeruginosa Pseudomonas cause waterborne HCAI, however, little is known about the virulence potential of Pseudomonas species found within hospital environments. METHODS Swabs were taken from 62 sinks within two newly opened wards at Great Ormond Street Hospital, samples were taken before and after the wards opened to understand the impact of patient occupancy on sink micro-organisms. Culturable bacteria were identified by MALDI-TOF and virulence factors assessed through phenotypic methods. RESULTS A total of 106 bacterial isolates were recovered including 24 Pseudomonas isolates. Of these 25% were identified as P. oleovorans, 21% P. aeruginosa, 17% P. composti, 13% P. alicalipha, 8% P. monteilii, 4% P. putida, 4% P. stutzeri and 8% could only be identified to genus level by MALDI-TOF. Differences were seen in both the number of Pseudomonas isolates and virulence production between the two wards, overall 25% of the Pseudomonas isolates produced pigment, 58% were capable of haemolysis, 87.5% were able to swim, 83.3% were capable of twitching motility, 33.3% produced alkaline protease and 8.3% produced gelatinase. CONCLUSIONS Results suggest that patients may be back-contaminating sinks with colonizing organisms which has ongoing implications for infection prevention and control. Additionally, this work highlights the ability of non-aeruginosa Pseudomonas to produce virulence factors traditionally associated with P. aeruginosa.
Collapse
Affiliation(s)
- H Rickard
- Healthy Infrastructure Research Group, Department of Civil, Environmental and Geomatic Engineering, University College London, London, UK.
| | - E Cloutman-Green
- Healthy Infrastructure Research Group, Department of Civil, Environmental and Geomatic Engineering, University College London, London, UK; Camelia Botnar Laboratories, Department of Microbiology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - L Ciric
- Healthy Infrastructure Research Group, Department of Civil, Environmental and Geomatic Engineering, University College London, London, UK
| |
Collapse
|
46
|
Cordery C, Craddock J, Malý M, Basavaraja K, Webb JS, Walsh MA, Tews I. Control of phosphodiesterase activity in the regulator of biofilm dispersal RbdA from Pseudomonas aeruginosa. RSC Chem Biol 2024:d4cb00113c. [PMID: 39247681 PMCID: PMC11372557 DOI: 10.1039/d4cb00113c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024] Open
Abstract
The switch between planktonic and biofilm lifestyle correlates with intracellular concentration of the second messenger bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP). While bacteria possess cyclase and phosphodiesterase enzymes to catalyse formation or hydrolysis of c-di-GMP, both enzymatic domains often occur in a single protein. It is tacitly assumed that one of the two enzymatic activities is dominant, and that additional domains and protein interactions enable responses to environmental conditions and control activity. Here we report the structure of the phosphodiesterase domain of the membrane protein RbdA (regulator of biofilm dispersal) in a dimeric, activated state and show that phosphodiesterase activity is controlled by the linked cyclase. The phosphodiesterase region around helices α5/α6 forms the dimer interface, providing a rationale for activation, as this region was seen in contact with the cyclase domain in an auto-inhibited structure previously described. Kinetic analysis supports this model, as the activity of the phosphodiesterase alone is lower when linked to the cyclase. Analysis of a computed model of the RbdA periplasmatic domain reveals an all-helical architecture with a large binding pocket that could accommodate putative ligands. Unravelling the regulatory circuits in multi-domain phosphodiesterases like RbdA is important to develop strategies to manipulate or disperse bacterial biofilms.
Collapse
Affiliation(s)
- Charlotte Cordery
- Biological Sciences, Institute for Life Sciences, University of Southampton Southampton SO17 1BJ UK
- National Biofilms Innovation Centre, University of Southampton Southampton SO17 1BJ UK
- Diamond Light Source, Harwell Science and Innovation Campus Didcot Oxfordshire OX11 0DE UK
- Research Complex at Harwell, Harwell Science and Innovation Campus Didcot Oxfordshire OX11 0FA UK
| | - Jack Craddock
- Biological Sciences, Institute for Life Sciences, University of Southampton Southampton SO17 1BJ UK
- National Biofilms Innovation Centre, University of Southampton Southampton SO17 1BJ UK
| | - Martin Malý
- Biological Sciences, Institute for Life Sciences, University of Southampton Southampton SO17 1BJ UK
| | - Kieran Basavaraja
- Biological Sciences, Institute for Life Sciences, University of Southampton Southampton SO17 1BJ UK
- National Biofilms Innovation Centre, University of Southampton Southampton SO17 1BJ UK
- Diamond Light Source, Harwell Science and Innovation Campus Didcot Oxfordshire OX11 0DE UK
- Research Complex at Harwell, Harwell Science and Innovation Campus Didcot Oxfordshire OX11 0FA UK
| | - Jeremy S Webb
- Biological Sciences, Institute for Life Sciences, University of Southampton Southampton SO17 1BJ UK
- National Biofilms Innovation Centre, University of Southampton Southampton SO17 1BJ UK
| | - Martin A Walsh
- Diamond Light Source, Harwell Science and Innovation Campus Didcot Oxfordshire OX11 0DE UK
- Research Complex at Harwell, Harwell Science and Innovation Campus Didcot Oxfordshire OX11 0FA UK
| | - Ivo Tews
- Biological Sciences, Institute for Life Sciences, University of Southampton Southampton SO17 1BJ UK
- National Biofilms Innovation Centre, University of Southampton Southampton SO17 1BJ UK
| |
Collapse
|
47
|
Montelongo-Martínez LF, Díaz-Guerrero M, Flores-Vega VR, Soto-Aceves MP, Rosales-Reyes R, Quiroz-Morales SE, González-Pedrajo B, Soberón-Chávez G, Cocotl-Yañez M. The quorum sensing regulator RhlR positively controls the expression of the type III secretion system in Pseudomonas aeruginosa PAO1. PLoS One 2024; 19:e0307174. [PMID: 39146292 PMCID: PMC11326643 DOI: 10.1371/journal.pone.0307174] [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: 01/08/2024] [Accepted: 07/01/2024] [Indexed: 08/17/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunist bacterium that causes acute and chronic infections. During acute infections, the type III secretion system (T3SS) plays a pivotal role in allowing the bacteria to translocate effectors such as ExoS, ExoT, and ExoY into host cells for colonization. Previous research on the involvement of quorum sensing systems Las and Rhl in controlling the T3SS gene expression produced ambiguous results. In this study, we determined the role of the Las and Rhl systems and the PqsE protein on T3SS expression. Our results show that in the wild-type PAO1 strain, the deletion of lasR or pqsE do not affect the secretion of ExoS. However, rhlI inactivation increases the expression of T3SS genes. In contrast to the rhlI deletion, rhlR inactivation decreases both T3SS genes expression and ExoS secreted protein levels, and this phenotype is restored when this mutant is complemented with the exsA gene, which codes for the master regulator of the T3SS. Additionally, cytotoxicity is affected in the rhlR mutant strain compared with its PAO1 parental strain. Overall, our results indicate that neither the Las system nor PqsE are involved in regulating the T3SS. Moreover, the Rhl system components have opposite effects, RhlI participates in negatively controlling the T3SS expression, while RhlR does it in a positive way, and this regulation is independent of C4 or PqsE. Finally, we show that rhlR, rhlI, or pqsE inactivation abolished pyocyanin production in T3SS-induction conditions. The ability of RhlR to act as a positive T3SS regulator in the absence of its cognate autoinducer and PqsE shows that it is a versatile regulator that controls different virulence traits allowing P. aeruginosa to compete for a niche.
Collapse
Affiliation(s)
- Luis Fernando Montelongo-Martínez
- Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Miguel Díaz-Guerrero
- Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad Nacional Autónoma de México, Ciudad de México, México
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, México
| | - Verónica Roxana Flores-Vega
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Martín Paolo Soto-Aceves
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Roberto Rosales-Reyes
- Facultad de Medicina, Unidad de Medicina Experimental, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Sara Elizabeth Quiroz-Morales
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Bertha González-Pedrajo
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, México
| | - Gloria Soberón-Chávez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Miguel Cocotl-Yañez
- Facultad de Medicina, Departamento de Microbiología y Parasitología, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
48
|
Price CTD, Hanford HE, Al-Quadan T, Santic M, Shin CJ, Da'as MSJ, Abu Kwaik Y. Amoebae as training grounds for microbial pathogens. mBio 2024; 15:e0082724. [PMID: 38975782 PMCID: PMC11323580 DOI: 10.1128/mbio.00827-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024] Open
Abstract
Grazing of amoebae on microorganisms represents one of the oldest predator-prey dynamic relationships in nature. It represents a genetic "melting pot" for an ancient and continuous multi-directional inter- and intra-kingdom horizontal gene transfer between amoebae and its preys, intracellular microbial residents, endosymbionts, and giant viruses, which has shaped the evolution, selection, and adaptation of microbes that evade degradation by predatory amoeba. Unicellular phagocytic amoebae are thought to be the ancient ancestors of macrophages with highly conserved eukaryotic processes. Selection and evolution of microbes within amoeba through their evolution to target highly conserved eukaryotic processes have facilitated the expansion of their host range to mammals, causing various infectious diseases. Legionella and environmental Chlamydia harbor an immense number of eukaryotic-like proteins that are involved in ubiquitin-related processes or are tandem repeats-containing proteins involved in protein-protein and protein-chromatin interactions. Some of these eukaryotic-like proteins exhibit novel domain architecture and novel enzymatic functions absent in mammalian cells, such as ubiquitin ligases, likely acquired from amoebae. Mammalian cells and amoebae may respond similarly to microbial factors that target highly conserved eukaryotic processes, but mammalian cells may undergo an accidental response to amoeba-adapted microbial factors. We discuss specific examples of microbes that have evolved to evade amoeba predation, including the bacterial pathogens- Legionella, Chlamydia, Coxiella, Rickettssia, Francisella, Mycobacteria, Salmonella, Bartonella, Rhodococcus, Pseudomonas, Vibrio, Helicobacter, Campylobacter, and Aliarcobacter. We also discuss the fungi Cryptococcus, and Asperigillus, as well as amoebae mimiviruses/giant viruses. We propose that amoeba-microbe interactions will continue to be a major "training ground" for the evolution, selection, adaptation, and emergence of microbial pathogens equipped with unique pathogenic tools to infect mammalian hosts. However, our progress will continue to be highly dependent on additional genomic, biochemical, and cellular data of unicellular eukaryotes.
Collapse
Affiliation(s)
- Christopher T. D. Price
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Hannah E. Hanford
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Tasneem Al-Quadan
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | | | - Cheon J. Shin
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Manal S. J. Da'as
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
| | - Yousef Abu Kwaik
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
- Center for Predictive Medicine, College of Medicine, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
49
|
Østergaard MZ, Nielsen FD, Meinfeldt MH, Kirkpatrick CL. The uncharacterized PA3040-3042 operon is part of the cell envelope stress response and a tobramycin resistance determinant in a clinical isolate of Pseudomonas aeruginosa. Microbiol Spectr 2024; 12:e0387523. [PMID: 38949386 PMCID: PMC11302039 DOI: 10.1128/spectrum.03875-23] [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: 11/09/2023] [Accepted: 05/22/2024] [Indexed: 07/02/2024] Open
Abstract
Bacteriophages (hereafter "phages") are ubiquitous predators of bacteria in the natural world, but interest is growing in their development into antibacterial therapy as complement or replacement for antibiotics. However, bacteria have evolved a huge variety of antiphage defense systems allowing them to resist phage lysis to a greater or lesser extent. In addition to dedicated phage defense systems, some aspects of the general stress response also impact phage susceptibility, but the details of this are not well known. In order to elucidate these factors in the opportunistic pathogen Pseudomonas aeruginosa, we used the laboratory-conditioned strain PAO1 as host for phage infection experiments as it is naturally poor in dedicated phage defense systems. Screening by transposon insertion sequencing indicated that the uncharacterized operon PA3040-PA3042 was potentially associated with resistance to lytic phages. However, we found that its primary role appeared to be in regulating biofilm formation, particularly in a clinical isolate of P. aeruginosa in which it also altered tobramycin resistance. Its expression was highly growth-phase dependent and responsive to phage infection and cell envelope stress. Our results suggest that this operon may be a cryptic but important locus for P. aeruginosa stress tolerance. IMPORTANCE An important category of bacterial stress response systems is bacteriophage defense, where systems are triggered by bacteriophage infection and activate a response which may either destroy the phage genome or destroy the infected cell so that the rest of the population survives. In some bacteria, the cell envelope stress response is activated by bacteriophage infection, but it is unknown whether this contributes to the survival of the infection. We have found that a conserved uncharacterized operon (PA3040-PA3042) of the cell envelope stress regulon in Pseudomonas aeruginosa, which has very few dedicated phage defense systems, responds to phage infection and stationary phase as well as envelope stress and is important for growth and biofilm formation in a clinical isolate of P. aeruginosa, even in the absence of phages. As homologs of these genes are found in other bacteria, they may be a novel component of the general stress response.
Collapse
Affiliation(s)
- Magnus Z. Østergaard
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Flemming D. Nielsen
- Department of Clinical Microbiology, Odense University Hospital, Odense, Denmark
| | - Mette H. Meinfeldt
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Clare L. Kirkpatrick
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
50
|
Sendra E, Fernández-Muñoz A, Zamorano L, Oliver A, Horcajada JP, Juan C, Gómez-Zorrilla S. Impact of multidrug resistance on the virulence and fitness of Pseudomonas aeruginosa: a microbiological and clinical perspective. Infection 2024; 52:1235-1268. [PMID: 38954392 PMCID: PMC11289218 DOI: 10.1007/s15010-024-02313-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/30/2024] [Indexed: 07/04/2024]
Abstract
Pseudomonas aeruginosa is one of the most common nosocomial pathogens and part of the top emergent species associated with antimicrobial resistance that has become one of the greatest threat to public health in the twenty-first century. This bacterium is provided with a wide set of virulence factors that contribute to pathogenesis in acute and chronic infections. This review aims to summarize the impact of multidrug resistance on the virulence and fitness of P. aeruginosa. Although it is generally assumed that acquisition of resistant determinants is associated with a fitness cost, several studies support that resistance mutations may not be associated with a decrease in virulence and/or that certain compensatory mutations may allow multidrug resistance strains to recover their initial fitness. We discuss the interplay between resistance profiles and virulence from a microbiological perspective but also the clinical consequences in outcomes and the economic impact.
Collapse
Affiliation(s)
- Elena Sendra
- Infectious Diseases Service, Hospital del Mar, Infectious Pathology and Antimicrobials Research Group (IPAR), Hospital del Mar Research Institute, Universitat Autònoma de Barcelona (UAB), CEXS-Universitat Pompeu Fabra, Passeig Marítim 25-27, 08003, Barcelona, Spain
| | - Almudena Fernández-Muñoz
- Research Unit, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Microbiology Department, University Hospital Son Espases, Crtra. Valldemossa 79, 07010, Palma, Spain
| | - Laura Zamorano
- Research Unit, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Microbiology Department, University Hospital Son Espases, Crtra. Valldemossa 79, 07010, Palma, Spain
| | - Antonio Oliver
- Research Unit, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Microbiology Department, University Hospital Son Espases, Crtra. Valldemossa 79, 07010, Palma, Spain
- Center for Biomedical Research in Infectious Diseases Network (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Pablo Horcajada
- Infectious Diseases Service, Hospital del Mar, Infectious Pathology and Antimicrobials Research Group (IPAR), Hospital del Mar Research Institute, Universitat Autònoma de Barcelona (UAB), CEXS-Universitat Pompeu Fabra, Passeig Marítim 25-27, 08003, Barcelona, Spain
- Center for Biomedical Research in Infectious Diseases Network (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Juan
- Research Unit, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Microbiology Department, University Hospital Son Espases, Crtra. Valldemossa 79, 07010, Palma, Spain.
- Center for Biomedical Research in Infectious Diseases Network (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| | - Silvia Gómez-Zorrilla
- Infectious Diseases Service, Hospital del Mar, Infectious Pathology and Antimicrobials Research Group (IPAR), Hospital del Mar Research Institute, Universitat Autònoma de Barcelona (UAB), CEXS-Universitat Pompeu Fabra, Passeig Marítim 25-27, 08003, Barcelona, Spain.
- Center for Biomedical Research in Infectious Diseases Network (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|