1
|
Subramaniyan Y, Khan A, Mujeeburahiman M, Ashmitha, Shivani M, Patil S, Lavanya N, Rekha PD. Urological metabolic conditions alter regulation of quorum-sensing system and virulence genes expression in uropathogenic Pseudomonasaeruginosa. Microb Pathog 2025; 203:107502. [PMID: 40127827 DOI: 10.1016/j.micpath.2025.107502] [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: 09/02/2024] [Revised: 02/24/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is responsible for complicated urinary tract infections (UTI) acquired from different transmission routes. Quorum sensing (QS) plays an important role in the regulation of virulence factors in P. aeruginosa and is dependent on the nutritional conditions. Under pathological conditions, urine contains elevated levels of metabolites such as glucose, creatinine, albumin and haem that may alter the QS behavior in P. aeruginosa. Hence, we investigated the role of the QS system in P. aeruginosa on growth and the expression of virulence genes under altered urinary metabolic compositions. We used P. aeruginosa strains isolated from UTI patients, along with PAO1 as a reference strain and grown under simulated metabolic conditions in the synthetic urine. Growth, biofilm formation, motility, siderophore production, rhamnolipid secretion, pyocyanin production, elastase and urease activity were quantified using standard laboratory methods. The expression levels of QS genes, such as lasI/R, rhlI/R, and pqsA/R, alginate synthesis genes (algD/R), the motility-regulating gene (pilA), and urease gene (ureC), were estimated using qRT-PCR for comparison between the growth conditions. The results showed significant differences in growth, biofilm formation and virulence factors between the metabolic conditions and were significantly higher (p<0.001) in simulated glycosuria, haematuria, and creatininuria conditions compared to the control. Under albuminuria, significantly reduced growth and virulence factor production were observed. Compared to control, higher expression levels of lasI/R, rhlI/R, pqsA/R, algD/R, pilA and ureC genes were observed in all conditions except albuminuria. The results highlighted the overexpression of QS-regulating genes and virulence in strains from similar metabolic environments in patients, suggesting possible niche adaptation. Such adaptation may pose considerable challenge in the management of chronic P. aeruginosa infections in urinary tract.
Collapse
Affiliation(s)
- Yuvarajan Subramaniyan
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), University Road, Deralakatte, Mangalore, 575018, India
| | - Altaf Khan
- Department of Urology, Yenepoya Medical College and Hospital, Yenepoya (Deemed to Be University), University Road, Deralakatte, Mangalore, 575018, India
| | - M Mujeeburahiman
- Department of Urology, Yenepoya Medical College and Hospital, Yenepoya (Deemed to Be University), University Road, Deralakatte, Mangalore, 575018, India
| | - Ashmitha
- Department of Biosciences, Mangalore University, Mangalagangotri, Mangalore, 574199, India
| | - M Shivani
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), University Road, Deralakatte, Mangalore, 575018, India
| | - Smitha Patil
- Department of Biosciences, Mangalore University, Mangalagangotri, Mangalore, 574199, India
| | - N Lavanya
- Department of Biosciences, Mangalore University, Mangalagangotri, Mangalore, 574199, India
| | - Punchappady Devasya Rekha
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), University Road, Deralakatte, Mangalore, 575018, India.
| |
Collapse
|
2
|
He L, Wang W, Chen H, Ma L, Yu L, Yang Y, Qu Y, Dai P, Wang D, Ma X. Gene expressions of clinical Pseudomonas aeruginosa harboring RND efflux pumps on chromosome and involving a novel integron on a plasmid. Microb Pathog 2025; 203:107512. [PMID: 40154852 DOI: 10.1016/j.micpath.2025.107512] [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: 11/14/2024] [Revised: 03/03/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
The clinical strain of Pseudomonas aeruginosa XM8 harbored multiple RND-type antibiotic efflux pump genes and a novel integron In4881 on its plasmid pXM8-2, rendering it resistant to nearly all conventional antibiotics except colistin. The resistance was primarily attributed to the inactivation of the oprD gene and overexpression of several efflux pump genes, including mexAB-oprM, mexCD-oprJ, oprN-mexFE, and mexXY. In this study, the XM8 strain was comprehensively characterized using various methods. Antimicrobial susceptibility testing was performed using the BioMerieux VITEK2 system and manual double dilution methods. Gene expression levels of efflux pump-related genes were analyzed via quantitative real-time PCR. The bacterial chromosome and plasmid were sequenced using both Illumina and Nanopore platforms, and bioinformatics tools were employed to analyze mobile genetic elements associated with antibiotic resistance. The pXM8-2 plasmid containsed multiple mobile genetic elements, including integrons (In4881, In334, In413) and transposons (Tn3, TnAs1, TnAs3). Notably, In4881 was reported for the first time in this study. The presence of these elements highlights the potential for horizontal gene transfer and further spread of antibiotic resistance. Given the strong resistance profile of the XM8 strain, effective measures should be implemented to prevent the dissemination and prevalence of such multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Long He
- Department of Clinical Laboratory Medicine, Wenling First People's Hospital, Taizhou, Zhejiang, 317500, China
| | - Wenji Wang
- Department of Central Laboratory, Taizhou Municipal Hospital (Taizhou Municipal Hospital Affiliated with Taizhou University), Taizhou, Zhejiang, 318000, China; School of Life Sciences, Taizhou University, Taizhou, Zhejiang, 318000, China
| | - Haiming Chen
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Xiamen University (Xiamen Key Laboratory of Genetic Testing), Xiamen, Fujian, 361003, China
| | - Liman Ma
- Department of Basic Medicine and Medical laboratory Science, School of Medicine, Taizhou University, Taizhou, Zhejiang, 318000, China
| | - Lianhua Yu
- Department of Clinical Laboratory Medicine, Taizhou Municipal Hospital (Taizhou Municipal Hospital Affiliated with Taizhou University), Taizhou, Zhejiang, 318000, China
| | - Yide Yang
- Department of Infectious Disease, Taizhou Municipal Hospital (Taizhou Municipal Hospital Affiliated with Taizhou University), Taizhou, Zhejiang, 318000, China
| | - Ying Qu
- Department of Clinical Laboratory Medicine, Taizhou Municipal Hospital (Taizhou Municipal Hospital Affiliated with Taizhou University), Taizhou, Zhejiang, 318000, China
| | - Piaopiao Dai
- Department of Clinical Laboratory Medicine, Taizhou Municipal Hospital (Taizhou Municipal Hospital Affiliated with Taizhou University), Taizhou, Zhejiang, 318000, China
| | - Dongguo Wang
- Department of Central Laboratory, Taizhou Municipal Hospital (Taizhou Municipal Hospital Affiliated with Taizhou University), Taizhou, Zhejiang, 318000, China.
| | - Xiaobo Ma
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Xiamen University (Xiamen Key Laboratory of Genetic Testing), Xiamen, Fujian, 361003, China.
| |
Collapse
|
3
|
Raya J, Montagut EJ, Marco MP. Analysing the integrated quorum sensing system its potential role in Pseudomonas aeruginosa pathogenesis. Front Cell Infect Microbiol 2025; 15:1575421. [PMID: 40438239 PMCID: PMC12116448 DOI: 10.3389/fcimb.2025.1575421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/02/2025] [Indexed: 06/01/2025] Open
Abstract
Since its discovery, Quorum Sensing (QS), a form of bacterial communication, has been the focus of numerous studies aimed at unravelling the mechanisms behind this intricate process. Bacterial QS relies on releasing low molecular weight signals known as autoinducers (AIs). When these AIs reach a threshold concentration, they activate coordinated genetic expression of pathogenic and bacterial survival mechanisms. Pseudomonas aeruginosa's QS has been extensively studied due to its incidence and clinical significance in a wide range of human infections. Several decades ago, three QS systems, named Las, Rhl, and Pqs, were identified and have since then become the focus of numerous research studies and the target of innovative diagnostic and therapeutic strategies. However, a fourth QS-related system was more recently proposed that it has been the subject of debate. Named "integrated quorum sensing" (Iqs), interconnects the previously mentioned systems with the phosphate stress response. The associated AI has been identified as 2-(2-hydroxyphenyl)-thiazole-4-carbaldehyde, also known as IQS. This discovery has sparked a controversial discussion about its biosynthetic origin and whether it truly functions as an intercellular communication system. In this review, we critically discuss the different hypotheses, and its biological relevance while presenting key findings of the Iqs system.
Collapse
Affiliation(s)
- Juan Raya
- Department of Chemical and Biomolecular Nanotechnology, Nanobiotechnology for Diagnostics (Nb4D), Institute for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - Enrique-J. Montagut
- Department of Chemical and Biomolecular Nanotechnology, Nanobiotechnology for Diagnostics (Nb4D), Institute for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - M.-Pilar Marco
- Department of Chemical and Biomolecular Nanotechnology, Nanobiotechnology for Diagnostics (Nb4D), Institute for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| |
Collapse
|
4
|
Maaskant A, Lee D, Ngo H, Montijn RC, Bakker J, Langermans JAM, Levin E. AI for rapid identification of major butyrate-producing bacteria in rhesus macaques (Macaca mulatta). Anim Microbiome 2025; 7:39. [PMID: 40275402 PMCID: PMC12020216 DOI: 10.1186/s42523-025-00410-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND The gut microbiome plays a crucial role in health and disease, influencing digestion, metabolism, and immune function. Traditional microbiome analysis methods are often expensive, time-consuming, and require specialized expertise, limiting their practical application in clinical settings. Evolving artificial intelligence (AI) technologies present opportunities for developing alternative methods. However, the lack of transparency in these technologies limits the ability of clinicians to incorporate AI-driven diagnostic tools into their healthcare systems. The aim of this study was to investigate an AI approach that rapidly predicts different bacterial genera and bacterial groups, specifically butyrate producers, from digital images of fecal smears of rhesus macaques (Macaca mulatta). In addition, to improve transparency, we employed explainability analysis to uncover the image features influencing the model's predictions. RESULTS By integrating fecal image data with corresponding metagenomic sequencing information, the deep learning (DL) and machine learning (ML) algorithms successfully predicted 16 individual bacterial genera (area under the curve (AUC) > 0.7) among the 50 most abundant genera in rhesus macaques (Macaca mulatta). The model was successful in predicting functional groups, major butyrate producers (AUC 0.75) and a mixed group including fermenters and short-chain fatty acid (SCFA) producers (AUC 0.81). For both models of butyrate producers and mixed fermenters, the explainability experiments revealed no decline in the AUC when random noise was added to the images. Increased blurring led to a gradual decline in the AUC. The model's performance was robust against the impact of fecal shape from smearing, with a stable AUC maintained until patch 4 for all groups, as assessed through scrambling. No significant correlation was detected between the prediction probabilities and the total fecal weight used in the smear; r = 0.30 ± 0.3 (p > 0.1) and r = 0.04 ± 0.36 (p > 0.8) for the butyrate producers and mixed fermenters, respectively. CONCLUSION Our approach demonstrated the ability to predict a wide range of clinically relevant microbial genera and microbial groups in the gut microbiome based on digital images from a fecal smear. The models proved to be robust to the smearing method, random noise and the amount of fecal matter. This study introduces a rapid, non-invasive, and cost-effective method for microbiome profiling, with potential applications in veterinary diagnostics.
Collapse
Affiliation(s)
- Annemiek Maaskant
- Biomedical Primate Research Centre, Lange Kleiweg 161, Rijswijk, 2288 GJ, Netherlands.
- Department Population Health Sciences, Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, Utrecht, 3584 CM, Netherlands.
| | - Donghyeok Lee
- HORAIZON Technology BV, Marshallaan 2, Delft, 2625 GZ, Netherlands
| | - Huy Ngo
- HORAIZON Technology BV, Marshallaan 2, Delft, 2625 GZ, Netherlands
| | - Roy C Montijn
- HORAIZON Technology BV, Marshallaan 2, Delft, 2625 GZ, Netherlands
| | - Jaco Bakker
- Biomedical Primate Research Centre, Lange Kleiweg 161, Rijswijk, 2288 GJ, Netherlands
| | - Jan A M Langermans
- Biomedical Primate Research Centre, Lange Kleiweg 161, Rijswijk, 2288 GJ, Netherlands
- Department Population Health Sciences, Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, Utrecht, 3584 CM, Netherlands
| | - Evgeni Levin
- HORAIZON Technology BV, Marshallaan 2, Delft, 2625 GZ, Netherlands.
| |
Collapse
|
5
|
Kennelly C, Prindle A. Substrate identification of putative NCS1 and NCS2 nucleobase transporters in Pseudomonas aeruginosa. mBio 2024; 15:e0243424. [PMID: 39475230 PMCID: PMC11633122 DOI: 10.1128/mbio.02434-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/03/2024] [Indexed: 12/12/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that can salvage nucleobases from the environment to conserve nutrients that would otherwise be spent on de novo nucleotide biosynthesis. However, little is known regarding the substrate specificity of the 13 putative nucleobase transporters in P. aeruginosa. Here, using a combination of genetic and chemical approaches, we report substrate identifications for 10 putative nucleobase transporters in P. aeruginosa. Specifically, we individually expressed each transporter in a genetic background lacking all 13 putative nucleobase transporters and quantified growth on a panel of 10 nucleobases as sole nitrogen sources. We confirmed these expression-based substrate identifications using targeted genetic knockouts. In a complementary approach, we utilized four toxic nucleobase antimetabolites to characterize antimicrobial activity in these same strains. We identified the sole allantoin transporter as well as transporters for guanine, xanthine, uric acid, cytosine, thymine, uracil, and dihydrouracil. Furthermore, we associated at least five nucleobase transporters with hypoxanthine, which has been recently reported to be an antibiofilm cue in P. aeruginosa. These results provide an initial characterization of the putative nucleobase transporters in P. aeruginosa, significantly advancing our understanding of nucleobase transport in this clinically relevant organism. IMPORTANCE Pseudomonas aeruginosa is a frequently multidrug-resistant opportunistic pathogen and one of the most common causes of healthcare-acquired infections. While nucleobases are known to support growth in nutrient-limited conditions, recent work showed that adenine and hypoxanthine can also decrease P. aeruginosa biofilm formation by disrupting c-di-GMP metabolism. Thus, nucleobase transport may be relevant to multiple aspects of P. aeruginosa biology and pathogenesis. However, there is currently little known about the transport of nucleobases in P. aeruginosa. Our work reports initial substrate identifications for 10 putative nucleobase transporters in P. aeruginosa, providing new tools to address previously difficult-to-test hypotheses relating to nucleobase transport in this organism.
Collapse
Affiliation(s)
- Corey Kennelly
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Center for Synthetic Biology, Northwestern University, Chicago, Illinois, USA
| | - Arthur Prindle
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Center for Synthetic Biology, Northwestern University, Chicago, Illinois, USA
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA
- Chan Zuckerberg Biohub Chicago, Chicago, Illinois, USA
| |
Collapse
|
6
|
Bisht K, Elmassry MM, Mahmud HA, Bhattacharjee S, Deonarine A, Black C, Francisco MJS, Hamood AN, Wakeman CA. Malonate is relevant to the lung environment and induces genome-wide stress responses in Pseudomonas aeruginosa. RESEARCH SQUARE 2024:rs.3.rs-4870062. [PMID: 39315254 PMCID: PMC11419262 DOI: 10.21203/rs.3.rs-4870062/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Versatility in carbon source utilization is a major contributor to niche adaptation in Pseudomonas aeruginosa. Malonate is among the abundant carbon sources in the lung airways, yet it is understudied. Recently, we characterized how malonate impacts quorum sensing regulation, antibiotic resistance, and virulence factor production in P. aeruginosa. Herein, we show that malonate as a carbon source supports more robust growth in comparison to glycerol in several cystic fibrosis isolates of P. aeruginosa. Furthermore, we show phenotypic responses to malonate were conserved among clinical strains, i.e., formation of biomineralized biofilm-like aggregates, increased tolerance to kanamycin, and increased susceptibility to norfloxacin. Moreover, we explored transcriptional adaptations of P. aeruginosa UCBPP-PA14 (PA14) in response to malonate versus glycerol as a sole carbon source using transcriptomics. Malonate utilization activated glyoxylate and methylcitrate cycles and induced several stress responses, including oxidative, anaerobic, and metal stress responses associated with increases in intracellular aluminum and strontium. We identified several genes that were required for optimal growth of P. aeruginosa in malonate. Our findings reveal important remodeling of P. aeruginosa gene expression during its growth on malonate as a sole carbon source that is accompanied by several important phenotypic changes. These findings add to the accumulating literature highlighting the role of different carbon sources in the physiology of P. aeruginosa and its niche adaptation.
Collapse
|
7
|
Dhingra S, Zhang Z, Lohans CT, Brewitz L, Schofield CJ. Substitution of 2-oxoglutarate alters reaction outcomes of the Pseudomonas savastanoi ethylene-forming enzyme. J Biol Chem 2024; 300:107546. [PMID: 38992435 PMCID: PMC11345546 DOI: 10.1016/j.jbc.2024.107546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024] Open
Abstract
In seeding plants, biosynthesis of the phytohormone ethylene, which regulates processes including fruit ripening and senescence, is catalyzed by 1-aminocyclopropane-1-carboxylic acid (ACC) oxidase. The plant pathogen Pseudomonas savastanoi (previously classified as: Pseudomonas syringae) employs a different type of ethylene-forming enzyme (psEFE), though from the same structural superfamily as ACC oxidase, to catalyze ethylene formation from 2-oxoglutarate (2OG) in an arginine dependent manner. psEFE also catalyzes the more typical oxidation of arginine to give L-Δ1-pyrroline-5-carboxylate (P5C), a reaction coupled to oxidative decarboxylation of 2OG giving succinate and CO2. We report on the effects of C3 and/or C4 substituted 2OG derivatives on the reaction modes of psEFE. 1H NMR assays, including using the pure shift method, reveal that, within our limits of detection, none of the tested 2OG derivatives is converted to an alkene; some are converted to the corresponding β-hydroxypropionate or succinate derivatives, with only the latter being coupled to arginine oxidation. The NMR results reveal that the nature of 2OG derivatization can affect the outcome of the bifurcating reaction, with some 2OG derivatives exclusively favoring the arginine oxidation pathway. Given that some of the tested 2OG derivatives are natural products, the results are of potential biological relevance. There are also opportunities for therapeutic or biocatalytic regulation of the outcomes of reactions catalyzed by 2OG-dependent oxygenases by the use of 2OG derivatives.
Collapse
Affiliation(s)
- Siddhant Dhingra
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, Oxford, United Kingdom
| | - Zhihong Zhang
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, Oxford, United Kingdom
| | - Christopher T Lohans
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, Oxford, United Kingdom
| | - Lennart Brewitz
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, Oxford, United Kingdom.
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
8
|
Jacobtorweihen J, Hartmann A, Hofer S, Spiegler V. Antibacterial Activities of the Algal Bromophenol Methylrhodomelol Against Pseudomonas aeruginosa. PLANTA MEDICA 2024; 90:469-481. [PMID: 38580306 DOI: 10.1055/a-2289-2423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
Methylrhodomelol (1: ) is a bromophenol from the red alga Vertebrata lanosa that has been associated with antimicrobial properties. The aim of the current study was, therefore, to assess the antimicrobial potential of this compound in more detail against the gram-negative pathogen Pseudomonas aeruginosa. 1: exerted weak bacteriostatic activity against different strains when grown in minimal medium, whereas other phenolics were inactive. In addition, 1: (35 and 10 µg/mL) markedly enhanced the susceptibility of multidrug-resistant P. aeruginosa toward the aminoglycoside gentamicin, while it did not affect the viability of Vero kidney cells up to 100 µM. Finally, pyoverdine release was reduced in bacteria treated at sub-inhibitory concentration, but no effect on other virulence factors was observed. Transcriptome analysis of treated versus untreated P. aeruginosa indicated an interference of 1: with bacterial carbon and energy metabolism, which was corroborated by RT-qPCR and decreased ATP-levels in treated bacteria. In summary, the current study characterized the antibacterial properties of methylrhodomelol, revealed its potential as an adjuvant to standard antibiotics, and generated a hypothesis on its mode of action.
Collapse
Affiliation(s)
- Joshua Jacobtorweihen
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Germany
| | - Anja Hartmann
- Institute of Pharmacy, Pharmacognosy, University of Innsbruck, Austria
| | - Stefanie Hofer
- Institute of Pharmacy, Pharmacognosy, University of Innsbruck, Austria
| | - Verena Spiegler
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Germany
| |
Collapse
|
9
|
Bisht K, Elmassry MM, Al Mahmud H, Bhattacharjee S, Deonarine A, Black C, San Francisco MJ, Hamood AN, Wakeman CA. Global stress response in Pseudomonas aeruginosa upon malonate utilization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586813. [PMID: 38585990 PMCID: PMC10996706 DOI: 10.1101/2024.03.26.586813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Versatility in carbon source utilization assists Pseudomonas aeruginosa in its adaptation to various niches. Recently, we characterized the role of malonate, an understudied carbon source, in quorum sensing regulation, antibiotic resistance, and virulence factor production in P. aeruginosa . These results indicate that global responses to malonate metabolism remain to be uncovered. We leveraged a publicly available metabolomic dataset on human airway and found malonate to be as abundant as glycerol, a common airway metabolite and carbon source for P. aeruginosa . Here, we explored and compared adaptations of P. aeruginosa UCBPP-PA14 (PA14) in response to malonate or glycerol as a sole carbon source using transcriptomics and phenotypic assays. Malonate utilization activated glyoxylate and methylcitrate cycles and induced several stress responses, including oxidative, anaerobic, and metal stress responses associated with increases in intracellular aluminum and strontium. Some induced genes were required for optimal growth of P. aeruginosa in malonate. To assess the conservation of malonate-associated responses among P. aeruginosa strains, we compared our findings in strain PA14 with other lab strains and cystic fibrosis isolates of P. aeruginosa . Most strains grew on malonate as a sole carbon source as efficiently as or better than glycerol. While not all responses to malonate were conserved among strains, formation of biomineralized biofilm-like aggregates, increased tolerance to kanamycin, and increased susceptibility to norfloxacin were the most frequently observed phenotypes. Our findings reveal global remodeling of P. aeruginosa gene expression during its growth on malonate as a sole carbon source that is accompanied by several important phenotypic changes. These findings add to accumulating literature highlighting the role of different carbon sources in the physiology of P. aeruginosa and its niche adaptation. Importance Pseudomonas aeruginosa is a notorious pathogen that causes local and systemic infections in immunocompromised individuals. Different carbon sources can uniquely modulate metabolic and virulence pathways in P. aeruginosa , highlighting the importance of the environment that the pathogen occupies. In this work, we used a combination of transcriptomic analysis and phenotypic assays to determine how malonate utilization impacts P. aeruginosa, as recent evidence indicates this carbon source may be relevant to certain niches associated within the human host. We found that malonate utilization can induce global stress responses, alter metabolic circuits, and influence various phenotypes of P. aeruginosa that could influence host colonization. Investigating the metabolism of malonate provides insight into P. aeruginosa adaptations to specific niches where this substrate is abundant, and how it can be leveraged in the development of much-needed antimicrobial agents or identification of new therapeutic targets of this difficult-to-eradicate pathogen.
Collapse
|
10
|
Gholami A, Minai-Tehrani D, Farewell A, Eriksson LA. Discovery of novel inhibitors for Pseudomonas aeruginosa lipase enzyme from in silico and in vitro studies. J Biomol Struct Dyn 2024; 42:2197-2210. [PMID: 37098781 DOI: 10.1080/07391102.2023.2203258] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 04/10/2023] [Indexed: 04/27/2023]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen prone to developing drug-resistance and is a major cause of infection for burn patients and patients suffering from cystic fibrosis or are hospitalized in intensive care units. One of the virulence factors of this bacterium is the lipase enzyme that degrades the extracellular matrix of the host tissue and promotes invasion. Bromhexine is a mucolytic drug and has recently been reported to function as a competitive inhibitor of lipase with an IC50 value of 49 µM. In the present study, an attempt was made to identify stronger inhibitors from the ChEMBL database of bioactive compounds, as compared to the reference compound Bromhexine. Following docking and MD simulations, four hit compounds (N1-N4) were selected that showed promising binding modes and low RMSD values indicative of stable protein-ligand complexes. From subsequent binding pose metadynamics (BPMD) simulations, two of these (N2 and N4) stood out as more potent than Bromhexine, displaying stable interactions with residues in the catalytic site of the enzyme. Biological investigations were performed for all four compounds. Among them, the same two hit compounds were found to be the most effective binders with IC50 values of 22.1 and 27.5 µM, respectively; i.e. roughly twice as efficient as the reference Bromhexine. Taken together, our results show that these hits can be promising new candidates to use as leads for the development of drugs targeting the P. aeruginosa lipase enzyme.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Asma Gholami
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, Sweden
| | - Dariush Minai-Tehrani
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Anne Farewell
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, Sweden
- Centre for Antibiotic Resistance Research, University of Gothenburg, Göteborg, Sweden
| | - Leif A Eriksson
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
11
|
Hu Y, Yuan M, Julian A, Tuz K, Juárez O. Identification of complex III, NQR, and SDH as primary bioenergetic enzymes during the stationary phase of Pseudomonas aeruginosa cultured in urine-like conditions. Front Microbiol 2024; 15:1347466. [PMID: 38468849 PMCID: PMC10926992 DOI: 10.3389/fmicb.2024.1347466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/08/2024] [Indexed: 03/13/2024] Open
Abstract
Pseudomonas aeruginosa is a common cause of urinary tract infections by strains that are often multidrug resistant, representing a major challenge to the world's health care system. This microorganism has a highly adaptable metabolism that allows it to colonize many environments, including the urinary tract. In this work, we have characterized the metabolic strategies used by stationary phase P. aeruginosa cells cultivated in urine-like media to understand the adaptations used by this microorganism to survive and produce disease. Our proteomics results show that cells rely on the Entner-Duodoroff pathway, pentose phosphate pathway, the Krebs cycle/ glyoxylate shunt and the aerobic oxidative phosphorylation to survive in urine-like media and other conditions. A deep characterization of the oxidative phosphorylation showed that the respiratory rate of stationary phase cells is increased 3-4 times compared to cells in the logarithmic phase of growth, indicating that the aerobic metabolism plays critical roles in the stationary phase of cells grown in urine like media. Moreover, the data show that respiratory complex III, succinate dehydrogenase and the NADH dehydrogenase NQR have important functions and could be used as targets to develop new antibiotics against this bacterium.
Collapse
Affiliation(s)
| | | | | | | | - Oscar Juárez
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL, United States
| |
Collapse
|
12
|
Vinayavekhin N, Wattanophas T, Murphy MF, Vangnai AS, Hobbs G. Metabolomics responses and tolerance of Pseudomonas aeruginosa under acoustic vibration stress. PLoS One 2024; 19:e0297030. [PMID: 38285708 PMCID: PMC10824448 DOI: 10.1371/journal.pone.0297030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/25/2023] [Indexed: 01/31/2024] Open
Abstract
Sound has been shown to impact microbial behaviors. However, our understanding of the chemical and molecular mechanisms underlying these microbial responses to acoustic vibration is limited. In this study, we used untargeted metabolomics analysis to investigate the effects of 100-Hz acoustic vibration on the intra- and extracellular hydrophobic metabolites of P. aeruginosa PAO1. Our findings revealed increased levels of fatty acids and their derivatives, quinolones, and N-acylethanolamines upon sound exposure, while rhamnolipids (RLs) showed decreased levels. Further quantitative real-time polymerase chain reaction experiments showed slight downregulation of the rhlA gene (1.3-fold) and upregulation of fabY (1.5-fold), fadE (1.7-fold), and pqsA (1.4-fold) genes, which are associated with RL, fatty acid, and quinolone biosynthesis. However, no alterations in the genes related to the rpoS regulators or quorum-sensing networks were observed. Supplementing sodium oleate to P. aeruginosa cultures to simulate the effects of sound resulted in increased tolerance of P. aeruginosa in the presence of sound at 48 h, suggesting a potential novel response-tolerance correlation. In contrast, adding RL, which went against the response direction, did not affect its growth. Overall, these findings provide potential implications for the control and manipulation of virulence and bacterial characteristics for medical and industrial applications.
Collapse
Affiliation(s)
- Nawaporn Vinayavekhin
- Center of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Thanyaporn Wattanophas
- Center of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Mark Francis Murphy
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Alisa S. Vangnai
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Glyn Hobbs
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
13
|
Fortunato G, Vaz-Moreira I, Gajic I, Manaia CM. Insight into phylogenomic bias of bla VIM-2 or bla NDM-1 dissemination amongst carbapenem-resistant Pseudomonas aeruginosa. Int J Antimicrob Agents 2023; 61:106788. [PMID: 36924802 DOI: 10.1016/j.ijantimicag.2023.106788] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 02/06/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023]
Abstract
OBJECTIVES Pseudomonas aeruginosa (P. aeruginosa) are ubiquitous opportunistic pathogens that combine intrinsic and acquired multidrug resistance phenotypes. Due to different types of acquired genes, carbapenem resistance has been expanding in this species. This study hypothesised that the spread of carbapenem resistance among P. aeruginosa is influenced by phylogenomic features, being distinct for different genes. METHODS To test this hypothesis, the genomes of P. aeruginosa harbouring blaVIM-2 or blaNDM-1 genes were compared. The blaVIM-2 gene was selected because, although frequent, it is almost restricted to this species and blaNDM-1 gene due to its wide interspecies distribution. A group of genomes harbouring the genes blaVIM-2 (n = 116) or blaNDM-1 (n = 27), available in GenBank, was characterised based on core phylogenomic analysis, functional categories in the accessory genome and mobile genetic elements flanking the selected genes. RESULTS Most blaVIM-2 gene hosts belonged to multilocus sequence types (ST) ST111 (n = 32 of 116) and ST233 (n = 27 of 116) and were reported in Europe (n = 75 of 116). The blaNDM-1 gene hosts were distributed by different STs (ST38, ST773, ST235, ST357 and ST654), frequently from Asia (n = 11 of 27). Significant differences in the prevalence of functional protein/enzyme annotations per number of accessory genomes were observed between blaVIM-2+ and blaNDM-1+. The blaVIM-2 gene was frequently inserted in the Tn402-like and Tn21 transposons family and rarely in IS6100, while blaNDM-1 gene was preferentially flanked by ISAba125 and bleMBL genes or associated with IS91 insertion sequence. CONCLUSION The hypothesis that carbapenem resistance gene acquisition is not random among phylogenomic lineages was confirmed, suggesting the importance of phylogeny in the dissemination of antibiotic resistance genes.
Collapse
Affiliation(s)
- Gianuario Fortunato
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Ivone Vaz-Moreira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Ina Gajic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Célia M Manaia
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal.
| |
Collapse
|
14
|
Shrestha GS, Vijay AK, Stapleton F, White A, Pickford R, Carnt N. Human tear metabolites associated with nucleoside-signalling pathways in bacterial keratitis. Exp Eye Res 2023; 228:109409. [PMID: 36775205 DOI: 10.1016/j.exer.2023.109409] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/17/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023]
Abstract
OBJECTIVE The study aimed to profile and quantify tear metabolites associated with bacterial keratitis using both untargeted and targeted metabolomic platforms. METHODS Untargeted metabolomic analysis using liquid-chromatography-Q Exactive-HF mass-spectrometry explored tear metabolites significantly associated with bacterial keratitis (n = 6) compared to healthy participants (n = 6). Differential statistics and principal component analysis determined meaningful metabolite differences between cases and controls. Purines and nucleosides were further quantified and compared between 15 cases and 15 controls in the targeted metabolomic platform using TSQ quantum access triple quadrupole mass spectrometry. Compound quantification was done by plotting the calibration curves and the difference in the compound levels was evaluated using the Wilcoxon rank-sum test. RESULTS In the untargeted analysis, 49 tear metabolites (27 upregulated and 22 downregulated) were differentially expressed between cases and controls. The untargeted analysis indicated that the purine metabolism pathway was the most affected by bacterial keratitis. Metabolite quantification in the targeted analysis further confirmed the upregulation of xanthine (P = 0.02) and downregulation of adenine (P < 0.0001), adenosine (P < 0.0001) and cytidine (P < 0.0001) in the tears of participants with bacterial keratitis compared to that of healthy participants. CONCLUSIONS Bacterial keratitis significantly changes the tear metabolite profile, including five major compound classes such as indoles, amino acids, nucleosides, carbohydrates, and steroids. This study also indicates that tear fluids can be used to map the metabolic pathways and uncover metabolic markers associated with bacterial keratitis. Conceivably, the inhibition of nucleoside synthesis may contribute to the pathophysiology of bacterial keratitis because nucleosides are required for maintaining cellular energy homeostasis and immune adaptability.
Collapse
Affiliation(s)
| | | | - Fiona Stapleton
- School of Optometry and Vision Science, UNSW Sydney, Australia
| | - Andrew White
- Department of Ophthalmology, Westmead Hospital, University of Sydney, Australia; Westmead Institute for Medical Research, University of Sydney, Australia
| | - Russell Pickford
- Bioanalytical Mass Spectrometry Facility, UNSW Sydney, Australia
| | - Nicole Carnt
- School of Optometry and Vision Science, UNSW Sydney, Australia; Westmead Institute for Medical Research, University of Sydney, Australia; Institute of Ophthalmology, University College London, United Kingdom
| |
Collapse
|
15
|
Liu Y, Tang Y, Ren S, Chen L. Antibacterial Components and Modes of the Methanol-Phase Extract from Commelina communis Linn. PLANTS (BASEL, SWITZERLAND) 2023; 12:890. [PMID: 36840240 PMCID: PMC9966474 DOI: 10.3390/plants12040890] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
Infectious diseases caused by pathogenic bacteria severely threaten human health. Traditional Chinese herbs are potential sources of new or alternative medicine. In this study, we analyzed for the first time antibacterial substances in the methanol-phase extract from a traditional Chinese herb-Commelina communis Linn-which showed an inhibition rate of 58.33% against 24 species of common pathogenic bacteria. The extract was further purified using preparative high-performance liquid chromatography (Prep-HPLC), which generated four single fragments (Fragments 1 to 4). The results revealed that Fragment 1 significantly increased bacterial cell surface hydrophobicity and membrane permeability and decreased membrane fluidity, showing disruptive effects on cell integrity of Gram-positive and Gram-negative bacteria, such as Bacillus cereus, Enterococcus faecalis, Staphylococcus aureus, and Salmonella enterica subsp., compared to the control groups (p < 0.05). In sum, 65 compounds with known functions in Fragment 1 were identified using liquid chromatography and mass spectrometry (LC-MS), of which quercetin-3-o-glucuronide was predominant (19.35%). Comparative transcriptomic analysis revealed multiple altered metabolic pathways mediated by Fragment 1, such as inhibited ABC transporters, ribosome, citrate cycle and oxidative phosphorylation, and upregulated nitrogen metabolism and purine metabolism, thereby resulting in the repressed bacterial growth and even death (p < 0.05). Overall, the results of this study demonstrate that Fragment 1 from C. communis Linn is a promising candidate against common pathogenic bacteria.
Collapse
Affiliation(s)
- Yue Liu
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Shanghai 201306, China
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Yingping Tang
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Shanghai 201306, China
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Shunlin Ren
- Department of Internal Medicine, Virginia Commonwealth University/McGuire VA Medical Centre, Richmond, VA 23298, USA
| | - Lanming Chen
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Shanghai 201306, China
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
16
|
Bisht K, Luecke AR, Wakeman CA. Temperature-specific adaptations and genetic requirements in a biofilm formed by Pseudomonas aeruginosa. Front Microbiol 2023; 13:1032520. [PMID: 36687584 PMCID: PMC9853522 DOI: 10.3389/fmicb.2022.1032520] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/07/2022] [Indexed: 01/07/2023] Open
Abstract
Pseudomonas aeruginosa is a gram-negative opportunistic pathogen often associated with nosocomial infections that are made more severe by this bacterium's ability to form robust biofilms. A biofilm is a microbial community encompassing cells embedded within an extracellular polymeric substrate (EPS) matrix that is typically secreted by the encased microbial cells. Biofilm formation is influenced by several environmental cues, and temperature fluctuations are likely to be an important stimulus in the lifecycle of P. aeruginosa as it transitions between life in aquatic or soil environments to sites of infection in the human host. Previous work has demonstrated that human body temperature can induce a shift in the biofilm EPS relative to room temperature growth, resulting in an incorporation of a filamentous phage coat protein into the biofilm EPS. In this study, we sought to identify adaptations enabling biofilm formation at room temperature or temperatures mimicking the natural environment of P. aeruginosa (23°C and 30°C) relative to temperatures mimicking life in the human host (37°C and 40°C). We identified higher biofilm: biomass ratios at lower temperatures on certain substrates, which correlated with a higher relative abundance of apparent polysaccharide EPS content. However, the known genes for EPS polysaccharide production in P. aeruginosa PA14 did not appear to be specifically important for temperature-dependent biofilm adaptation, with the pelB gene appearing to be generally important and the algD gene being generally expendable in all conditions tested. Instead, we were able to identify two previously uncharacterized hypothetical proteins (PA14_50070 and PA14_67550) specifically required for biofilm formation at 23°C and/or 30°C relative to temperatures associated with the human host. These unstudied contributors to biofilm integrity may have been previously overlooked since most P. aeruginosa biofilm studies tend to use 37°C growth temperatures. Overall, our study demonstrates that temperature shifts can have dramatic impacts on biofilm structure and highlights the importance of studying environment-specific adaptations in biofilm physiology.
Collapse
Affiliation(s)
| | | | - Catherine A. Wakeman
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| |
Collapse
|
17
|
Frigoli M, Lowdon JW, Caldara M, Arreguin-Campos R, Sewall J, Cleij TJ, Diliën H, Eersels K, van Grinsven B. Thermal Pyocyanin Sensor Based on Molecularly Imprinted Polymers for the Indirect Detection of Pseudomonas aeruginosa. ACS Sens 2023; 8:353-362. [PMID: 36599088 PMCID: PMC9887650 DOI: 10.1021/acssensors.2c02345] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Pseudomonas aeruginosa is a ubiquitous multi-drug-resistant bacterium, capable of causing serious illnesses and infections. This research focuses on the development of a thermal sensor for the indirect detection of P. aeruginosa infection using molecularly imprinted polymers (MIPs). This was achieved by developing MIPs for the detection of pyocyanin, the main toxin secreted by P. aeruginosa. To this end, phenazine was used as a dummy template, evaluating several polymeric compositions to achieve a selective MIP for pyocyanin recognition. The sensitivity of the synthesized MIPs was investigated by UV-vis analysis, with the best composition having a maximum rebinding capacity of 30 μmol g-1 and an imprinting factor (IF) of 1.59. Subsequently, the MIP particles were immobilized onto planar aluminum chips using an adhesive layer, to perform thermal resistance measurements at clinically relevant concentrations of pyocyanin (1.4-9.8 μM), achieving a limit of detection (LoD) of 0.347 ± 0.027 μM. The selectivity of the sensor was also scrutinized by subjecting the receptor to potential interferents. Furthermore, the rebinding was demonstrated in King's A medium, highlighting the potential of the sensor for the indirect detection of P. aeruginosa in complex fluids. The research culminates in the demonstration of the MIP-based sensor's applicability for clinical diagnosis. To achieve this goal, an experiment was performed in which the sensor was exposed to pyocyanin-spiked saliva samples, achieving a limit of detection of 0.569 ± 0.063 μM and demonstrating that this technology is suitable to detect the presence of the toxin even at the very first stage of its production.
Collapse
|
18
|
Lerdsittikul V, Thongdee M, Chaiwattanarungruengpaisan S, Atithep T, Apiratwarrasakul S, Withatanung P, Clokie MRJ, Korbsrisate S. A novel virulent Litunavirus phage possesses therapeutic value against multidrug resistant Pseudomonas aeruginosa. Sci Rep 2022; 12:21193. [PMID: 36476652 PMCID: PMC9729221 DOI: 10.1038/s41598-022-25576-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Pseudomonas aeruginosa is a notable nosocomial pathogen that can cause severe infections in humans and animals. The emergence of multidrug resistant (MDR) P. aeruginosa has motivated the development of phages to treat the infections. In this study, a novel Pseudomonas phage, vB_PaeS_VL1 (VL1), was isolated from urban sewage. Phylogenetic analyses revealed that VL1 is a novel species in the genus Litunavirus of subfamily Migulavirinae. The VL1 is a virulent phage as no genes encoding lysogeny, toxins or antibiotic resistance were identified. The therapeutic potential of phage VL1 was investigated and revealed that approximately 56% (34/60 strains) of MDR P. aeruginosa strains, isolated from companion animal diseases, could be lysed by VL1. In contrast, VL1 did not lyse other Gram-negative and Gram-positive bacteria suggesting its specificity of infection. Phage VL1 demonstrated high efficiency to reduce bacterial load (~ 6 log cell number reduction) and ~ 75% reduction of biofilm in pre-formed biofilms of MDR P. aeruginosa. The result of two of the three MDR P. aeruginosa infected Galleria mellonella larvae showed that VL1 could significantly increase the survival rate of infected larvae. Taken together, phage VL1 has genetic and biological properties that make it a potential candidate for phage therapy against P. aeruginosa infections.
Collapse
Affiliation(s)
- Varintip Lerdsittikul
- grid.10223.320000 0004 1937 0490Veterinary Diagnostic Center, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Metawee Thongdee
- grid.10223.320000 0004 1937 0490The Monitoring and Surveillance Center for Zoonotic Diseases in Wildlife and Exotic Animals, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Somjit Chaiwattanarungruengpaisan
- grid.10223.320000 0004 1937 0490The Monitoring and Surveillance Center for Zoonotic Diseases in Wildlife and Exotic Animals, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Thassanant Atithep
- grid.494627.a0000 0004 4684 9800Frontier Research Center, Vidyasirimedhi Institute of Science and Technology, Rayong, Thailand
| | - Sukanya Apiratwarrasakul
- grid.10223.320000 0004 1937 0490Veterinary Diagnostic Center, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Patoo Withatanung
- grid.10223.320000 0004 1937 0490Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Martha R. J. Clokie
- grid.9918.90000 0004 1936 8411Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Sunee Korbsrisate
- grid.10223.320000 0004 1937 0490Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
19
|
Gholami A, Minai-Tehrani D, Eftekhar F. Bromhexine and its inhibitory effect on lipase - kinetics and structural study. Arch Physiol Biochem 2022; 128:1687-1692. [PMID: 32633559 DOI: 10.1080/13813455.2020.1788606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Lipase hydrolyses the ester bonds in triglyceride. It is an important enzyme in medicine and industry. Some pathogen bacteria use this exoenzyme to disrupt the extracellular matrix of host organisms. Pseudomonas uses various extracellular enzymes such as lipase to invade its host. In this report, for the first time, bromhexine was introduced as an inhibitor of lipase. Bromhexine is a mucolytic drug which is used in the treatment of respiratory tract disorders. The results showed that bromhexine inhibited the enzyme by competitive inhibition. IC50 and Ki values of the drug were 0.049 mM and 0.02 mM, respectively. Arrhenius plot showed that the drug reduced the activation energy. The enzyme was purified and SDS-PAGE showed that its molecular weight is 13 kDa. Fluorescence measurement revealed that binding of the drug to lipase could make structural changes in the enzyme. Inhibition of lipase by bromhexine could be applicable in medicine.
Collapse
Affiliation(s)
- Asma Gholami
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Dariush Minai-Tehrani
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Fereshteh Eftekhar
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
20
|
Growth Substrate and Prophage Induction Collectively Influence Metabolite and Lipid Profiles in a Marine Bacterium. mSystems 2022; 7:e0058522. [PMID: 35972149 PMCID: PMC9600351 DOI: 10.1128/msystems.00585-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Bacterial growth substrates influence a variety of biological functions, including the biosynthesis and regulation of lipid intermediates. The extent of this rewiring is not well understood nor has it been considered in the context of virally infected cells. Here, we used a one-host-two-temperate phage model system to probe the combined influence of growth substrate and phage infection on host carbon and lipid metabolism. Using untargeted metabolomics and lipidomics, we reported the detection of a suite of metabolites and lipid classes for two Sulfitobacter lysogens provided with three growth substrates of differing complexity and nutrient composition (yeast extract/tryptone [complex], glutamate and acetate). The growth medium led to dramatic differences in the detectable intracellular metabolites, with only 15% of 175 measured metabolites showing overlap across the three growth substrates. Between-strain differences were most evident in the cultures grown on acetate, followed by glutamate then complex medium. Lipid distribution profiles were also distinct between cultures grown on different substrates as well as between the two lysogens grown in the same medium. Five phospholipids, three aminolipid, and one class of unknown lipid-like features were identified. Most (≥94%) of these 75 lipids were quantifiable in all samples. Metabolite and lipid profiles were strongly determined by growth medium composition and modestly by strain type. Because fluctuations in availability and form of carbon substrates and nutrients, as well as virus pressure, are common features of natural systems, the influence of these intersecting factors will undoubtedly be imprinted in the metabolome and lipidome of resident bacteria. IMPORTANCE Community-level metabolomics approaches are increasingly used to characterize natural microbial populations. These approaches typically depend upon temporal snapshots from which the status and function of communities are often inferred. Such inferences are typically drawn from lab-based studies of select model organisms raised under limited growth conditions. To better interpret community-level data, the extent to which ecologically relevant bacteria demonstrate metabolic flexibility requires elucidation. Herein, we used an environmentally relevant model heterotrophic marine bacterium to assess the relationship between growth determinants and metabolome. We also aimed to assess the contribution of phage activity to the host metabolome. Striking differences in primary metabolite and lipid profiles appeared to be driven primarily by growth regime and, secondarily, by phage type. These findings demonstrated the malleable nature of metabolomes and lipidomes and lay the foundation for future studies that relate cellular composition with function in complex environmental microbial communities.
Collapse
|
21
|
Grace A, Sahu R, Owen DR, Dennis VA. Pseudomonas aeruginosa reference strains PAO1 and PA14: A genomic, phenotypic, and therapeutic review. Front Microbiol 2022; 13:1023523. [PMID: 36312971 PMCID: PMC9607943 DOI: 10.3389/fmicb.2022.1023523] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
Pseudomonas aeruginosa is a ubiquitous, motile, gram-negative bacterium that has been recently identified as a multi-drug resistant pathogen in critical need of novel therapeutics. Of the approximately 5,000 strains, PAO1 and PA14 are common laboratory reference strains, modeling moderately and hyper-virulent phenotypes, respectively. PAO1 and PA14 have been instrumental in facilitating the discovery of novel drug targets, testing novel therapeutics, and supplying critical genomic information on the bacterium. While the two strains have contributed to a wide breadth of knowledge on the natural behaviors and therapeutic susceptibilities of P. aeruginosa, they have demonstrated significant deviations from observations in human infections. Many of these deviations are related to experimental inconsistencies in laboratory strain environment that complicate and, at times, terminate translation from laboratory results to clinical applications. This review aims to provide a comparative analysis of the two strains and potential methods to improve their clinical relevance.
Collapse
Affiliation(s)
- Amber Grace
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
| | - Rajnish Sahu
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
| | | | - Vida A. Dennis
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
- *Correspondence: Vida A. Dennis,
| |
Collapse
|
22
|
Revealing a New Family of D-2-Hydroxyglutarate Dehydrogenases in Escherichia coli and Pantoea ananatis Encoded by ydiJ. Microorganisms 2022; 10:microorganisms10091766. [PMID: 36144368 PMCID: PMC9504171 DOI: 10.3390/microorganisms10091766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/24/2022] Open
Abstract
In E. coli and P. ananatis, L-serine biosynthesis is initiated by the action of D-3-phosphoglycerate dehydrogenase (SerA), which converts D-3-phosphoglycerate into 3-phosphohydroxypyruvate. SerA can concomitantly catalyze the production of D-2-hydroxyglutarate (D-2-HGA) from 2-ketoglutarate by oxidizing NADH to NAD+. Several bacterial D-2-hydroxyglutarate dehydrogenases (D2HGDHs) have recently been identified, which convert D-2-HGA back to 2-ketoglutarate. However, knowledge about the enzymes that can metabolize D-2-HGA is lacking in bacteria belonging to the Enterobacteriaceae family. We found that ydiJ encodes novel D2HGDHs in P. ananatis and E. coli, which were assigned as D2HGDHPa and D2HGDHEc, respectively. Inactivation of ydiJ in P. ananatis and E. coli led to the significant accumulation of D-2-HGA. Recombinant D2HGDHEc and D2HGDHPa were purified to homogeneity and characterized. D2HGDHEc and D2HGDHPa are homotetrameric with a subunit molecular mass of 110 kDa. The pH optimum was 7.5 for D2HGDHPa and 8.0 for D2HGDHEc. The Km for D-2-HGA was 208 μM for D2HGDHPa and 83 μM for D2HGDHEc. The enzymes have strict substrate specificity towards D-2-HGA and displayed maximal activity at 45 °C. Their activity was completely inhibited by 0.5 mM Mn2+, Ni2+ or Co2+. The discovery of a novel family of D2HGDHs may provide fundamental information for the metabolic engineering of microbial chassis with desired properties.
Collapse
|
23
|
Iglesias-Fernandez M, Buxadera-Palomero J, Sadowska JM, Espanol M, Ginebra MP. Implementation of bactericidal topographies on biomimetic calcium phosphates and the potential effect of its reactivity. BIOMATERIALS ADVANCES 2022; 136:212797. [PMID: 35929296 DOI: 10.1016/j.bioadv.2022.212797] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 06/15/2023]
Abstract
Since the discovery that nanostructured surfaces were able to kill bacteria, many works have been published focusing on the design of nanopatterned surfaces with antimicrobial properties. Synthetic bone grafts, based on calcium phosphate (CaP) formulations, can greatly benefit from this discovery if adequate nanotopographies can be developed. However, CaP are reactive materials and experience ionic exchanges when placed into aqueous solutions which may in turn affect cell behaviour and complicate the interpretation of the bactericidal results. The present study explores the bactericidal potential of two nanopillared CaP prepared by hydrolysis of two different sizes of α-tricalcium phosphate (α-TCP) powders under biomimetic or hydrothermal conditions. A more lethal bactericidal response toward Pseudomonas aeruginosa (~75% killing efficiency of adhered bacteria) was obtained from the hydrothermally treated CaP which consisted in a more irregular topography in terms of pillar size (radius: 20-60 nm), interpillar distances (100-1500 nm) and pillar distribution (pillar groups forming bouquets) than the biomimetically treated one (radius: 20-40 nm and interpillar distances: 50-200 nm with a homogeneous pillar distribution). The material reactivity was greatly influenced by the type of medium (nutrient-rich versus nutrient-free) and the presence or not of bacteria. A lower reactivity and superior bacterial attachment were observed in the nutrient-free medium while a lower attachment was observed for the nutrient rich medium which was explained by a superior reactivity of the material paired with the lower tendency of planktonic bacteria to adhere on surfaces in the presence of nutrients. Importantly, the ionic exchanges produced by the presence of materials were not toxic to planktonic cells. Thus, we can conclude that topography was the main contributor to mortality in the bacterial adhesion tests.
Collapse
Affiliation(s)
- Marc Iglesias-Fernandez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain; Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain
| | - Judit Buxadera-Palomero
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain; Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain
| | - Joanna-Maria Sadowska
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain; Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain
| | - Montserrat Espanol
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain; Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain.
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain; Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain; Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| |
Collapse
|
24
|
Tang XK, Su YB, Ye HQ, Dai ZY, Yi H, Yang KX, Zhang TT, Chen ZG. Glucose-Potentiated Amikacin Killing of Cefoperazone/Sulbactam Resistant Pseudomonas aeruginosa. Front Microbiol 2022; 12:800442. [PMID: 35310395 PMCID: PMC8928219 DOI: 10.3389/fmicb.2021.800442] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
Multidrug-resistant Pseudomonas aeruginosa has become one of global threat pathogens for human health due to insensitivity to antibiotics. Recently developed reprogramming metabolomics can identify biomarkers, and then, the biomarkers were used to revert the insensitivity and elevate antibiotic-mediated killing. Here, the methodology was used to study cefoperazone/sulbactam (SCF)-resistant P. aeruginosa (PA-RSCF) and identified reduced glycolysis and pyruvate cycle, a recent clarified cycle providing respiratory energy in bacteria, as the most key enriched pathways and the depressed glucose as one of the most crucial biomarkers. Further experiments showed that the depression of glucose was attributed to reduction of glucose transport. However, exogenous glucose reverted the reduction to elevate intracellular glucose via activating glucose transport. The elevated glucose fluxed to the glycolysis, pyruvate cycle, and electron transport chain to promote downstream proton motive force (PMF). Consistently, exogenous glucose did not promote SCF-mediated elimination but potentiated aminoglycosides-mediated killing since aminoglycosides uptake is PMF-dependent, where amikacin was the best one. The glucose-potentiated amikacin-mediated killing was effective to both lab-evolved PA-RSCF and clinical multidrug-resistant P. aeruginosa. These results reveal the depressed glucose uptake causes the reduced intracellular glucose and expand the application of metabolome-reprogramming on selecting conventional antibiotics to achieve the best killing efficacy.
Collapse
Affiliation(s)
- Xi-kang Tang
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu-bin Su
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Hui-qing Ye
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhen-yuan Dai
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huan Yi
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ke-xin Yang
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tian-tuo Zhang
- Department of Pulmonary & Critical Care Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhuang-gui Chen
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Zhuang-gui Chen,
| |
Collapse
|
25
|
Postbiotics of Lactobacillus casei target virulence and biofilm formation of Pseudomonas aeruginosa by modulating quorum sensing. Arch Microbiol 2022; 204:157. [PMID: 35106661 DOI: 10.1007/s00203-022-02770-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/03/2022] [Accepted: 01/17/2022] [Indexed: 11/02/2022]
Abstract
Among various anti-virulence aspects, the efficacy of the bioactive constituents of probiotics, referred to as postbiotics, to affect quorum sensing (QS)-modulated signaling of pathogens, is considered as a safe natural approach. The present study investigated the potential QS-inhibitory activity of lyophilized postbiotics from Lactobacillus casei sub sp. casei PTCC 1608 on virulence phenotypes and biofilm of two strains and three clinical isolates of Pseudomonas aeruginosa. The effect of L. casei postbiotics (LCP) at sub-minimum inhibitory concentration on the expression of QS genes including lasR/I, rhlR/I, pqsA, pqsR and virulence genes including pelF (pellicle/biofilm glycosyltransferase PelF), lasB (elastase LasB) and toxA (exotoxin A) was evaluated. The viability of mouse fibroblastic NIH/3T3 cell line treated with sub-MICS of LCP was also investigated. Postbiotics were characterized using mass spectrometry-based analyses. The QS-attenuation effect of pure lactic acid as the major constituent of LCP was determined on P. aeruginosa strains. Neutralized postbiotics and crude bacteriocin did not exhibit any antibacterial activity. It was found that sub-MICS of LCP could more drastically attenuate the tested virulence phenotypes and biofilm formation than lactic acid. Biofilm inhibition was confirmed using scanning electron microscopy. The rhlI, rhlR, and pelF genes were down-regulated after treatment with LCP. No cytotoxicity effect was observed on NIH/3T3 cell line. The findings demonstrated that postbiotics of L. casei could reduce the virulence and biofilm development of P. aeruginosa and suggested a novel safe natural source for the expansion of anti-virulence treatments.
Collapse
|
26
|
Pseudomonas aeruginosa Pangenome: Core and Accessory Genes of a Highly Resourceful Opportunistic Pathogen. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1386:3-28. [DOI: 10.1007/978-3-031-08491-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
27
|
Tümmler B. What Makes Pseudomonas aeruginosa a Pathogen? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1386:283-301. [DOI: 10.1007/978-3-031-08491-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
28
|
Comparison of bacteria disintegration methods and their influence on data analysis in metabolomics. Sci Rep 2021; 11:20859. [PMID: 34675363 PMCID: PMC8531443 DOI: 10.1038/s41598-021-99873-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 09/13/2021] [Indexed: 11/16/2022] Open
Abstract
Metabolomic experiments usually contain many different steps, each of which can strongly influence the obtained results. In this work, metabolic analyses of six bacterial strains were performed in light of three different bacterial cell disintegration methods. Three strains were gram-negative (Pseudomonas aeruginosa, Escherichia coli, and Klebsiella pneumoniae), and three were gram-positive (Corynebacterium glutamicum, Bacillus cereus, and Enterococcus faecalis). For extraction, the methanol–water extraction method (1:1) was chosen. To compare the efficiency of different cell disintegration methods, sonication, sand mill, and tissue lyser were used. For bacterial extract metabolite analysis, 1H NMR together with univariate and multivariate analyses were applied. The obtained results showed that metabolite concentrations are strongly dependent on the cell lysing methodology used and are different for various bacterial strains. The results clearly show that one of the disruption methods gives the highest concentration for most identified compounds (e. g. sand mill for E. faecalis and tissue lyser for B. cereus). This study indicated that the comparison of samples prepared by different procedures can lead to false or imprecise results, leaving an imprint of the disintegration method. Furthermore, the presented results showed that NMR might be a useful bacterial strain identification and differentiation method. In addition to disintegration method comparison, the metabolic profiles of each elaborated strain were analyzed, and each exhibited its metabolic profile. Some metabolites were identified by the 1H NMR method in only one strain. The results of multivariate data analyses (PCA) show that regardless of the disintegration method used, the strain group can be identified. Presented results can be significant for all types of microbial studies containing the metabolomic targeted and non-targeted analysis.
Collapse
|
29
|
Oxidative Stress Response in Pseudomonas aeruginosa. Pathogens 2021; 10:pathogens10091187. [PMID: 34578219 PMCID: PMC8466533 DOI: 10.3390/pathogens10091187] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/17/2022] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative environmental and human opportunistic pathogen highly adapted to many different environmental conditions. It can cause a wide range of serious infections, including wounds, lungs, the urinary tract, and systemic infections. The high versatility and pathogenicity of this bacterium is attributed to its genomic complexity, the expression of several virulence factors, and its intrinsic resistance to various antimicrobials. However, to thrive and establish infection, P. aeruginosa must overcome several barriers. One of these barriers is the presence of oxidizing agents (e.g., hydrogen peroxide, superoxide, and hypochlorous acid) produced by the host immune system or that are commonly used as disinfectants in a variety of different environments including hospitals. These agents damage several cellular molecules and can cause cell death. Therefore, bacteria adapt to these harsh conditions by altering gene expression and eliciting several stress responses to survive under oxidative stress. Here, we used PubMed to evaluate the current knowledge on the oxidative stress responses adopted by P. aeruginosa. We will describe the genes that are often differently expressed under oxidative stress conditions, the pathways and proteins employed to sense and respond to oxidative stress, and how these changes in gene expression influence pathogenicity and the virulence of P. aeruginosa. Understanding these responses and changes in gene expression is critical to controlling bacterial pathogenicity and developing new therapeutic agents.
Collapse
|
30
|
Bacterial Resistance against Heavy Metals in Pseudomonas aeruginosa RW9 Involving Hexavalent Chromium Removal. SUSTAINABILITY 2021. [DOI: 10.3390/su13179797] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Pseudomonas aeruginosa RW9 is a promising candidate for the bioremediation of chromium hexavalent (Cr(VI)) pollution, as it resists a high concentration of up to 60 mg/L of Cr(VI). Leaving cells exposed to Cr(VI) has large bioreduction potential, implying its capacity to extract the ions from the contaminated medium. In this study, the tolerance for and distribution of Cr(VI) were investigated to identify the cells’ adaptation and removal strategies. Micro-characterization analysis was conducted to assess the effect of Cr(VI) on the cells. The cells’ elongation was observed at higher Cr(VI) concentrations, signifying their adaptation to DNA damage caused by Cr(VI) toxicity. Cr(VI) distribution analysis showed that the strain developed a complex mechanism to adapt to Cr(VI), based on surface-bound (0.46 mg/L), intracellularly accumulated (1.24 mg/L) and extracellular sequestration (6.74 mg/L), which accounted for 85% of the removal efficiency. The extracellular sequestration might be attributable to the production of metabolites, in accordance with the fourier-transform infrared spectroscopy (FTIR) spectra and orcinol analysis that confirmed the presence of a glycolipid biosurfactant, rhamnolipid. Remarkably, the rhamnolipid was slightly induced in the presence of Cr(VI). From the data obtained, it was confirmed that this local strain is well equipped to survive high doses of Cr(VI) and has great potential for application in Cr(VI) bioremediation.
Collapse
|
31
|
Manzo G, Gianfanti F, Hind CK, Allison L, Clarke M, Hohenbichler J, Limantoro I, Martin B, Do Carmo Silva P, Ferguson PM, Hodgson-Casson AC, Fleck RA, Sutton JM, Phoenix DA, Mason AJ. Impacts of Metabolism and Organic Acids on Cell Wall Composition and Pseudomonas aeruginosa Susceptibility to Membrane Active Antimicrobials. ACS Infect Dis 2021; 7:2310-2323. [PMID: 34329558 DOI: 10.1021/acsinfecdis.1c00002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Reliable antimicrobial susceptibility testing is essential in informing both clinical antibiotic therapy decisions and the development of new antibiotics. Mammalian cell culture media have been proposed as an alternative to bacteriological media, potentially representing some critical aspects of the infection environment more accurately. Here, we use a combination of NMR metabolomics and electron microscopy to investigate the response of Escherichia coli and Pseudomonas aeruginosa to growth in differing rich media to determine whether and how this determines metabolic strategies, the composition of the cell wall, and consequently susceptibility to membrane active antimicrobials including colistin and tobramycin. The NMR metabolomic approach is first validated by characterizing the expected E. coli acid stress response to fermentation and the accompanying changes in the cell wall composition, when cultured in glucose rich mammalian cell culture media. Glucose is not a major carbon source for P. aeruginosa but is associated with a response to osmotic stress and a modest increase in colistin tolerance. Growth of P. aeruginosa in a range of bacteriological media is supported by consumption of formate, an important electron donor in anaerobic respiration. In mammalian cell culture media, however, the overall metabolic strategy of P. aeruginosa is instead dependent on consumption of glutamine and lactate. Formate doping of mammalian cell culture media does not alter the overall metabolic strategy but is associated with polyamine catabolism, remodelling of both inner and outer membranes, and a modest sensitization of P. aeruginosa PAO1 to colistin. Further, in a panel of P. aeruginosa isolates an increase between 2- and 3-fold in sensitivity to tobramycin is achieved through doping with other organic acids, notably propionate which also similarly enhances the activity of colistin. Organic acids are therefore capable of nonspecifically influencing the potency of membrane active antimicrobials.
Collapse
Affiliation(s)
- Giorgia Manzo
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Federico Gianfanti
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Charlotte K. Hind
- Technology Development Group, National Infection Service, Public Health England, Salisbury SP4 0JG United Kingdom
| | - Leanne Allison
- Centre for Ultrastructural Imaging, Guy’s Campus, King’s College London, London SE1 1UL, United Kingdom
| | - Maria Clarke
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Julia Hohenbichler
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Ilene Limantoro
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Bethany Martin
- Technology Development Group, National Infection Service, Public Health England, Salisbury SP4 0JG United Kingdom
| | - Phoebe Do Carmo Silva
- Technology Development Group, National Infection Service, Public Health England, Salisbury SP4 0JG United Kingdom
| | - Philip M. Ferguson
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Alice C. Hodgson-Casson
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Roland A. Fleck
- Centre for Ultrastructural Imaging, Guy’s Campus, King’s College London, London SE1 1UL, United Kingdom
| | - J. Mark Sutton
- Technology Development Group, National Infection Service, Public Health England, Salisbury SP4 0JG United Kingdom
| | - David A. Phoenix
- School of Applied Science, London South Bank University, 103 Borough Road, London SE1 0AA, United Kingdom
| | - A. James Mason
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| |
Collapse
|
32
|
Dunphy LJ, Grimes KL, Wase N, Kolling GL, Papin JA. Untargeted Metabolomics Reveals Species-Specific Metabolite Production and Shared Nutrient Consumption by Pseudomonas aeruginosa and Staphylococcus aureus. mSystems 2021; 6:e0048021. [PMID: 34156287 PMCID: PMC8269234 DOI: 10.1128/msystems.00480-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/02/2021] [Indexed: 11/20/2022] Open
Abstract
While bacterial metabolism is known to impact antibiotic efficacy and virulence, the metabolic capacities of individual microbes in cystic fibrosis lung infections are difficult to disentangle from sputum samples. Here, we show that untargeted metabolomic profiling of supernatants of multiple strains of Pseudomonas aeruginosa and Staphylococcus aureus grown in monoculture in synthetic cystic fibrosis media (SCFM) reveals distinct species-specific metabolic signatures despite intraspecies metabolic variability. We identify a set of 15 metabolites that were significantly consumed by both P. aeruginosa and S. aureus, suggesting that nutrient competition has the potential to impact community dynamics even in the absence of other pathogen-pathogen interactions. Finally, metabolites that were uniquely produced by one species or the other were identified. Specifically, the virulence factor precursor anthranilic acid, as well as the quinoline 2,4-quinolinediol (DHQ), were robustly produced across all tested strains of P. aeruginosa. Through the direct comparison of the extracellular metabolism of P. aeruginosa and S. aureus in a physiologically relevant environment, this work provides insight toward the potential for metabolic interactions in vivo and supports the development of species-specific diagnostic markers of infection. IMPORTANCE Interactions between P. aeruginosa and S. aureus can impact pathogenicity and antimicrobial efficacy. In this study, we aim to better understand the potential for metabolic interactions between P. aeruginosa and S. aureus in an environment resembling the cystic fibrosis lung. We find that S. aureus and P. aeruginosa consume many of the same nutrients, suggesting that metabolic competition may play an important role in community dynamics during coinfection. We further identify metabolites uniquely produced by either organism with the potential to be developed into species-specific biomarkers of infection in the cystic fibrosis lung.
Collapse
Affiliation(s)
- Laura J. Dunphy
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Kassandra L. Grimes
- Department of Engineering Systems and Environment, University of Virginia, Charlottesville, Virginia, USA
| | - Nishikant Wase
- Biomolecular Analysis Facility, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Glynis L. Kolling
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Jason A. Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
33
|
Alford MA, Baquir B, An A, Choi KYG, Hancock REW. NtrBC Selectively Regulates Host-Pathogen Interactions, Virulence, and Ciprofloxacin Susceptibility of Pseudomonas aeruginosa. Front Cell Infect Microbiol 2021; 11:694789. [PMID: 34249781 PMCID: PMC8264665 DOI: 10.3389/fcimb.2021.694789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022] Open
Abstract
Pseudomonas aeruginosa is a metabolically versatile opportunistic pathogen capable of infecting distinct niches of the human body, including skin wounds and the lungs of cystic fibrosis patients. Eradication of P. aeruginosa infection is becoming increasingly difficult due to the numerous resistance mechanisms it employs. Adaptive resistance is characterized by a transient state of decreased susceptibility to antibiotic therapy that is distinct from acquired or intrinsic resistance, can be triggered by various environmental stimuli and reverted by removal of the stimulus. Further, adaptive resistance is intrinsically linked to lifestyles such as swarming motility and biofilm formation, both of which are important in infections and lead to multi-drug adaptive resistance. Here, we demonstrated that NtrBC, the master of nitrogen control, had a selective role in host colonization and a substantial role in determining intrinsic resistance to ciprofloxacin. P. aeruginosa mutant strains (ΔntrB, ΔntrC and ΔntrBC) colonized the skin but not the respiratory tract of mice as well as WT and, unlike WT, could be reduced or eradicated from the skin by ciprofloxacin. We hypothesized that nutrient availability contributed to these phenomena and found that susceptibility to ciprofloxacin was impacted by nitrogen source in laboratory media. P. aeruginosa ΔntrB, ΔntrC and ΔntrBC also exhibited distinct host interactions, including modestly increased cytotoxicity toward human bronchial epithelial cells, reduced virulence factor production and 10-fold increased uptake by macrophages. These data might explain why NtrBC mutants were less adept at colonizing the upper respiratory tract of mice. Thus, NtrBC represents a link between nitrogen metabolism, adaptation and virulence of the pathogen P. aeruginosa, and could represent a target for eradication of recalcitrant infections in situ.
Collapse
Affiliation(s)
- Morgan A Alford
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Beverlie Baquir
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Andy An
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Ka-Yee G Choi
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
34
|
Pseudomonas aeruginosa adaptation and evolution in patients with cystic fibrosis. Nat Rev Microbiol 2021; 19:331-342. [PMID: 33214718 DOI: 10.1038/s41579-020-00477-5] [Citation(s) in RCA: 275] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2020] [Indexed: 01/29/2023]
Abstract
Intense genome sequencing of Pseudomonas aeruginosa isolates from cystic fibrosis (CF) airways has shown inefficient eradication of the infecting bacteria, as well as previously undocumented patient-to-patient transmission of adapted clones. However, genome sequencing has limited potential as a predictor of chronic infection and of the adaptive state during infection, and thus there is increasing interest in linking phenotypic traits to the genome sequences. Phenotypic information ranges from genome-wide transcriptomic analysis of patient samples to determination of more specific traits associated with metabolic changes, stress responses, antibiotic resistance and tolerance, biofilm formation and slow growth. Environmental conditions in the CF lung shape both genetic and phenotypic changes of P. aeruginosa during infection. In this Review, we discuss the adaptive and evolutionary trajectories that lead to early diversification and late convergence, which enable P. aeruginosa to succeed in this niche, and we point out how knowledge of these biological features may be used to guide diagnosis and therapy.
Collapse
|
35
|
Zhao ZC, Xie GJ, Liu BF, Xing DF, Ding J, Han HJ, Ren NQ. A review of quorum sensing improving partial nitritation-anammox process: Functions, mechanisms and prospects. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 765:142703. [PMID: 33069466 DOI: 10.1016/j.scitotenv.2020.142703] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 06/11/2023]
Abstract
Partial nitritation-anammox (PNA) is a promising and energy-efficient process for the sustainable nitrogen removal. However, its wide applications are still limited by the long start-up period and instability of long-term operation. Quorum sensing (QS), as a way of cell-to-cell communication generally regulating various microbial behaviors, has been increasingly investigated in PNA process, because QS may substantially manipulate the metabolism of microorganisms and overcome the limitations of PNA process. This critical review provides a comprehensive analysis of QS in PNA systems, and identifies the challenges and opportunities for the optimization of PNA process based on QS. The analysis is grouped based on the configurations of PNA process, including partial nitritation, anammox and single-stage PNA systems. QS is confirmed to regulate various properties of PNA systems, including microbial activity, microbial growth rate, microbial aggregation, microbial interactions and the robustness under adverse conditions. Major challenges in the mechanisms of QS, such as QS circuits, target genes and the response to environmental inputs, are identified. Potential applications of QS, such as short-term addition of certain acyl-homoserine lactones (AHLs) or substances containing AHLs, transient unfavorable conditions to stimulate the secretion of AHLs, are also proposed. This review focuses on the theoretical and practical cognation for QS in PNA systems, and serves as a stepping stone for further QS-based strategies to enhance nitrogen removal through PNA process.
Collapse
Affiliation(s)
- Zhi-Cheng Zhao
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, China
| | - Guo-Jun Xie
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, China.
| | - Bing-Feng Liu
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, China
| | - De-Feng Xing
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, China
| | - Jie Ding
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, China
| | - Hong-Jun Han
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, China
| | - Nan-Qi Ren
- State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin 150090, China
| |
Collapse
|
36
|
Yaeger LN, Coles VE, Chan DCK, Burrows LL. How to kill Pseudomonas-emerging therapies for a challenging pathogen. Ann N Y Acad Sci 2021; 1496:59-81. [PMID: 33830543 DOI: 10.1111/nyas.14596] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 12/16/2022]
Abstract
As the number of effective antibiotics dwindled, antibiotic resistance (AR) became a pressing concern. Some Pseudomonas aeruginosa isolates are resistant to all available antibiotics. In this review, we identify the mechanisms that P. aeruginosa uses to evade antibiotics, including intrinsic, acquired, and adaptive resistance. Our review summarizes many different approaches to overcome resistance. Antimicrobial peptides have potential as therapeutics with low levels of resistance evolution. Rationally designed bacteriophage therapy can circumvent and direct evolution of AR and virulence. Vaccines and monoclonal antibodies are highlighted as immune-based treatments targeting specific P. aeruginosa antigens. This review also identifies promising drug combinations, antivirulence therapies, and considerations for new antipseudomonal discovery. Finally, we provide an update on the clinical pipeline for antipseudomonal therapies and recommend future avenues for research.
Collapse
Affiliation(s)
- Luke N Yaeger
- Department of Biochemistry and Biomedical Sciences and M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Victoria E Coles
- Department of Biochemistry and Biomedical Sciences and M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Derek C K Chan
- Department of Biochemistry and Biomedical Sciences and M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Lori L Burrows
- Department of Biochemistry and Biomedical Sciences and M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
37
|
Hreha TN, Foreman S, Duran-Pinedo A, Morris AR, Diaz-Rodriguez P, Jones JA, Ferrara K, Bourges A, Rodriguez L, Koffas MAG, Hahn M, Hauser AR, Barquera B. The three NADH dehydrogenases of Pseudomonas aeruginosa: Their roles in energy metabolism and links to virulence. PLoS One 2021; 16:e0244142. [PMID: 33534802 PMCID: PMC7857637 DOI: 10.1371/journal.pone.0244142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 12/03/2020] [Indexed: 11/19/2022] Open
Abstract
Pseudomonas aeruginosa is a ubiquitous opportunistic pathogen which relies on a highly adaptable metabolism to achieve broad pathogenesis. In one example of this flexibility, to catalyze the NADH:quinone oxidoreductase step of the respiratory chain, P. aeruginosa has three different enzymes: NUO, NQR and NDH2, all of which carry out the same redox function but have different energy conservation and ion transport properties. In order to better understand the roles of these enzymes, we constructed two series of mutants: (i) three single deletion mutants, each of which lacks one NADH dehydrogenase and (ii) three double deletion mutants, each of which retains only one of the three enzymes. All of the mutants grew approximately as well as wild type, when tested in rich and minimal medium and in a range of pH and [Na+] conditions, except that the strain with only NUO (ΔnqrFΔndh) has an extended lag phase. During exponential phase, the NADH dehydrogenases contribute to total wild-type activity in the following order: NQR > NDH2 > NUO. Some mutants, including the strain without NQR (ΔnqrF) had increased biofilm formation, pyocyanin production, and killed more efficiently in both macrophage and mouse infection models. Consistent with this, ΔnqrF showed increased transcription of genes involved in pyocyanin production.
Collapse
Affiliation(s)
- Teri N. Hreha
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Sara Foreman
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Ana Duran-Pinedo
- Department of Oral Biology, University of Florida, College of Dentistry, Gainesville, Florida, United States of America
| | - Andrew R. Morris
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Patricia Diaz-Rodriguez
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - J. Andrew Jones
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Kristina Ferrara
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Anais Bourges
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Lauren Rodriguez
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Mattheos A. G. Koffas
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Mariah Hahn
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Alan R. Hauser
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Blanca Barquera
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- * E-mail:
| |
Collapse
|
38
|
McGill SL, Yung Y, Hunt KA, Henson MA, Hanley L, Carlson RP. Pseudomonas aeruginosa reverse diauxie is a multidimensional, optimized, resource utilization strategy. Sci Rep 2021; 11:1457. [PMID: 33446818 PMCID: PMC7809481 DOI: 10.1038/s41598-020-80522-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022] Open
Abstract
Pseudomonas aeruginosa is a globally-distributed bacterium often found in medical infections. The opportunistic pathogen uses a different, carbon catabolite repression (CCR) strategy than many, model microorganisms. It does not utilize a classic diauxie phenotype, nor does it follow common systems biology assumptions including preferential consumption of glucose with an 'overflow' metabolism. Despite these contradictions, P. aeruginosa is competitive in many, disparate environments underscoring knowledge gaps in microbial ecology and systems biology. Physiological, omics, and in silico analyses were used to quantify the P. aeruginosa CCR strategy known as 'reverse diauxie'. An ecological basis of reverse diauxie was identified using a genome-scale, metabolic model interrogated with in vitro omics data. Reverse diauxie preference for lower energy, nonfermentable carbon sources, such as acetate or succinate over glucose, was predicted using a multidimensional strategy which minimized resource investment into central metabolism while completely oxidizing substrates. Application of a common, in silico optimization criterion, which maximizes growth rate, did not predict the reverse diauxie phenotypes. This study quantifies P. aeruginosa metabolic strategies foundational to its wide distribution and virulence including its potentially, mutualistic interactions with microorganisms found commonly in the environment and in medical infections.
Collapse
Affiliation(s)
- S Lee McGill
- Department of Chemical and Biological Engineering, Center for Biofilm Engineering, Montana State University, Bozeman, MT, 59717, USA.,Department of Microbiology and Immunology, Montana State University, Bozeman, MT, 59717, USA
| | - Yeni Yung
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Kristopher A Hunt
- Department of Chemical and Biological Engineering, Center for Biofilm Engineering, Montana State University, Bozeman, MT, 59717, USA.,Department of Civil and Environmental Engineering, University of Washington, Seattle, WA, 98115, USA
| | - Michael A Henson
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Luke Hanley
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Ross P Carlson
- Department of Chemical and Biological Engineering, Center for Biofilm Engineering, Montana State University, Bozeman, MT, 59717, USA. .,Department of Microbiology and Immunology, Montana State University, Bozeman, MT, 59717, USA.
| |
Collapse
|
39
|
Latour X. The Evanescent GacS Signal. Microorganisms 2020; 8:microorganisms8111746. [PMID: 33172195 PMCID: PMC7695008 DOI: 10.3390/microorganisms8111746] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 12/18/2022] Open
Abstract
The GacS histidine kinase is the membrane sensor of the major upstream two-component system of the regulatory Gac/Rsm signal transduction pathway. This pathway governs the expression of a wide range of genes in pseudomonads and controls bacterial fitness and motility, tolerance to stress, biofilm formation, and virulence or plant protection. Despite the importance of these roles, the ligands binding to the sensor domain of GacS remain unknown, and their identification is an exciting challenge in this domain. At high population densities, the GacS signal triggers a switch from primary to secondary metabolism and a change in bacterial lifestyle. It has been suggested, based on these observations, that the GacS signal is a marker of the emergence of nutritional stress and competition. Biochemical investigations have yet to characterize the GacS signal fully. However, they portray this cue as a low-molecular weight, relatively simple and moderately apolar metabolite possibly resembling, but nevertheless different, from the aliphatic organic acids acting as quorum-sensing signaling molecules in other Proteobacteria. Significant progress in the development of metabolomic tools and new databases dedicated to Pseudomonas metabolism should help to unlock some of the last remaining secrets of GacS induction, making it possible to control the Gac/Rsm pathway.
Collapse
Affiliation(s)
- Xavier Latour
- Laboratory of Microbiology Signals and Microenvironment (LMSM EA 4312), Normandy University (University of Rouen Normandy), 55 rue Saint-Germain, 27000 Evreux, France;
- Research Federation NORVEGE Fed4277, Normandy University, F-76821 Mont-Saint-Aignan, France
| |
Collapse
|
40
|
Quantitative proteomic reveals gallium maltolate induces an iron-limited stress response and reduced quorum-sensing in Pseudomonas aeruginosa. J Biol Inorg Chem 2020; 25:1153-1165. [DOI: 10.1007/s00775-020-01831-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/21/2020] [Indexed: 01/12/2023]
|
41
|
Evaluation of the Biodegradation Efficiency of Four Various Types of Plastics by Pseudomonas aeruginosa Isolated from the Gut Extract of Superworms. Microorganisms 2020; 8:microorganisms8091341. [PMID: 32887431 PMCID: PMC7563362 DOI: 10.3390/microorganisms8091341] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/31/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
Plastic waste worldwide is becoming a serious pollution problem for the planet. Various physical and chemical methods have been tested in attempts to remove plastic dumps. However, these have usually resulted in secondary pollution issues. Recently, the biodegradation of plastic by fungal and bacterial strains has been spotlighted as a promising solution to remove plastic wastes without generating secondary pollution. We have previously reported that a Pseudomonas aeruginosa strain isolated from the gut of a superworm is capable of biodegrading polystyrene (PS) and polyphenylene sulfide (PPS). Herein, we demonstrate the extraordinary biodegradative power of P. aeruginosa in efficiently depolymerizing four different types of plastics: PS, PPS, polyethylene (PE) and polypropylene (PP). We further compared biodegradation rates for these four plastic types and found that PE was biodegraded fastest, whereas the biodegradation of PP was the slowest. Moreover, the growth rates of P. aeruginosa were not always proportional to biodegradation rates, suggesting that the rate of bacterial growth could be influenced by the composition and properties of intermediate molecules produced during plastic biodegradation, and these may supply useful cellular precursors and energy. In conclusion, an initial screening system to select the most suitable bacterial strain to biodegrade certain types of plastic is particularly important and may be necessary to solve plastic waste problems both presently and in the future.
Collapse
|
42
|
Wang S, Huang Z, Wan Q, Feng S, Xie X, Zhang R, Zhang Z. Comparative Genomic and Metabolomic Analyses of Two Pseudomonas aeruginosa Strains With Different Antifungal Activities. Front Microbiol 2020; 11:1841. [PMID: 32849439 PMCID: PMC7412747 DOI: 10.3389/fmicb.2020.01841] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022] Open
Abstract
Pseudomonas aeruginosa isolated from the plant rhizosphere has been widely used as an effective strain in biological control against plant disease. This bacterium promotes plant growth and protect plants against various phytopathogens through the production of phenazine metabolites. In this study, the strain P. aeruginosa Y12 with anti-Beauveria bassiana activity was isolated from the gut of housefly larvae. It was comparatively analyzed with the strain P. aeruginosa P18, which showed no anti-B. bassiana activity. Genomic and metabolomic methods were used to obtain a comprehensive understanding of the antimicrobial mechanism of Y12. After whole-genome resequencing of the two strains, a total of 7,087 non-synonymous single-nucleotide polymorphisms (nsSNPs), 1079 insertions and deletions (InDels), 62 copy-number variations (CNVs) and 42 structural variations (SV) were found in both strains. We analyzed the differentially abundant metabolites between Y12 and P18, and identified six bioactive compounds that could be associated with the antimicrobial activity of Y12. Additionally, we found that, unlike other previously reported rhizospheric P. aeruginosa strains, Y12 could produce both phenazine-1,6-dicarboxylic acid (PDC) and pyocyanin (PYO) at significantly higher concentrations than P18. As B. bassiana is an effective biological insecticide that can cause high mortality in adult houseflies but has little effect on housefly larvae, we believe that P. aeruginosa Y12, identified in housefly larvae but not in adults, were beneficial for the development of housefly larvae and could protect them from B. bassiana infection through the production of toxic metabolites.
Collapse
Affiliation(s)
- Shumin Wang
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Zhendong Huang
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Qing Wan
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Shuo Feng
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Xiaochen Xie
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Ruiling Zhang
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Zhong Zhang
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Sciences, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| |
Collapse
|
43
|
Vitale A, Pessi G, Urfer M, Locher HH, Zerbe K, Obrecht D, Robinson JA, Eberl L. Identification of Genes Required for Resistance to Peptidomimetic Antibiotics by Transposon Sequencing. Front Microbiol 2020; 11:1681. [PMID: 32793157 PMCID: PMC7390954 DOI: 10.3389/fmicb.2020.01681] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/26/2020] [Indexed: 12/27/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogen and a leading cause of nosocomial infections. Due to its high intrinsic and adaptive resistance to antibiotics, infections caused by this organism are difficult to treat and new therapeutic options are urgently needed. Novel peptidomimetic antibiotics that target outer membrane (OM) proteins have shown great promise for the treatment of P. aeruginosa infections. Here, we have performed genome-wide mutant fitness profiling using transposon sequencing (Tn-Seq) to identify resistance determinants against the recently described peptidomimetics L27-11, compounds 3 and 4, as well as polymyxin B2 (PMB) and colistin (COL). We identified a set of 13 core genes that affected resistance to all tested antibiotics, many of which encode enzymes involved in the modification of the lipopolysaccharide (LPS) or control their expression. We also identified fitness determinants that are specific for antibiotics with similar structures that may indicate differences in their modes of action. These results provide new insights into resistance mechanisms against these peptide antibiotics, which will be important for future clinical development and efforts to further improve their potency.
Collapse
Affiliation(s)
- Alessandra Vitale
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Gabriella Pessi
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | | | | | - Katja Zerbe
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | | | - John A Robinson
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Leo Eberl
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
44
|
Depke T, Thöming JG, Kordes A, Häussler S, Brönstrup M. Untargeted LC-MS Metabolomics Differentiates Between Virulent and Avirulent Clinical Strains of Pseudomonas aeruginosa. Biomolecules 2020; 10:biom10071041. [PMID: 32668735 PMCID: PMC7407980 DOI: 10.3390/biom10071041] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/27/2020] [Accepted: 07/07/2020] [Indexed: 01/02/2023] Open
Abstract
Pseudomonas aeruginosa is a facultative pathogen that can cause, inter alia, acute or chronic pneumonia in predisposed individuals. The gram-negative bacterium displays considerable genomic and phenotypic diversity that is also shaped by small molecule secondary metabolites. The discrimination of virulence phenotypes is highly relevant to the diagnosis and prognosis of P. aeruginosa infections. In order to discover small molecule metabolites that distinguish different virulence phenotypes of P. aeruginosa, 35 clinical strains were cultivated under standard conditions, characterized in terms of virulence and biofilm phenotype, and their metabolomes were investigated by untargeted liquid chromatography-mass spectrometry. The data was both mined for individual candidate markers as well as used to construct statistical models to infer the virulence phenotype from metabolomics data. We found that clinical strains that differed in their virulence and biofilm phenotype also had pronounced divergence in their metabolomes, as underlined by 332 features that were significantly differentially abundant with fold changes greater than 1.5 in both directions. Important virulence-associated secondary metabolites like rhamnolipids, alkyl quinolones or phenazines were found to be strongly upregulated in virulent strains. In contrast, we observed little change in primary metabolism. A hitherto novel cationic metabolite with a sum formula of C12H15N2 could be identified as a candidate biomarker. A random forest model was able to classify strains according to their virulence and biofilm phenotype with an area under the Receiver Operation Characteristics curve of 0.84. These findings demonstrate that untargeted metabolomics is a valuable tool to characterize P. aeruginosa virulence, and to explore interrelations between clinically important phenotypic traits and the bacterial metabolome.
Collapse
Affiliation(s)
- Tobias Depke
- Department of Chemical Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;
| | - Janne Gesine Thöming
- Institute of Molecular Bacteriology, Twincore, Centre for Clinical and Experimental Infection Research, 30625 Hannover, Germany; (J.G.T.); (A.K.); (S.H.)
| | - Adrian Kordes
- Institute of Molecular Bacteriology, Twincore, Centre for Clinical and Experimental Infection Research, 30625 Hannover, Germany; (J.G.T.); (A.K.); (S.H.)
| | - Susanne Häussler
- Institute of Molecular Bacteriology, Twincore, Centre for Clinical and Experimental Infection Research, 30625 Hannover, Germany; (J.G.T.); (A.K.); (S.H.)
- Department of Molecular Bacteriology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
- Correspondence:
| |
Collapse
|
45
|
Ismail NS, Subbiah SK, Taib NM. Application of Phenotype Microarray for Profiling Carbon Sources Utilization between Biofilm and Non-Biofilm of Pseudomonas aeruginosa from Clinical Isolates. Curr Pharm Biotechnol 2020; 21:1539-1550. [PMID: 32598252 DOI: 10.2174/1389201021666200629145217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/17/2020] [Accepted: 05/04/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND This is the fastest work in obtaining the metabolic profiles of Pseudomonas aeruginosa in order to combat the infection diseases which leads to high morbidity and mortality rates. Pseudomonas aeruginosa is a high versatility of gram-negative bacteria that can undergo aerobic and anaerobic respiration. Capabilities in deploying different carbon sources, energy metabolism and regulatory system, ensure the survival of this microorganism in the diverse environment condition. Determination of differences in carbon sources utilization among biofilm and non-biofilm of Pseudomonas aeruginosa provides a platform in understanding the metabolic activity of the microorganism. METHODS The study was carried out from September 2017 to February 2019. Four archive isolates forming strong and intermediate biofilm and non-biofilms producer were subcultured from archive isolates. ATCC 27853 P. aeruginosa was used as a negative control or non-biofilm producing microorganism. Biofilm formation was confirmed by Crystal Violet Assay (CVA) and Congo Red Agar (CRA). Metabolic profiles of the biofilm and non-biofilms isolates were determined by phenotype microarrays (Biolog Omnilog). RESULTS AND DISCUSSION In this study, Pseudomonas aeruginosa biofilm isolates utilized uridine, L-threonine and L-serine while non-biofilm utilized adenosine, inosine, monomethyl, sorbic acid and succinamic acid. CONCLUSION The outcome of this result will be used for future studies to improve detection or inhibit the growth of P. aeruginosa biofilm and non-biofilm respectively.
Collapse
Affiliation(s)
- Nur S Ismail
- Department of Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Suresh K Subbiah
- Department of Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Niazlin M Taib
- Department of Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
46
|
Cramer N, Fischer S, Hedtfeld S, Dorda M, Tümmler B. Intraclonal competitive fitness of longitudinal cystic fibrosis Pseudomonas aeruginosa airway isolates in liquid cultures. Environ Microbiol 2020; 22:2536-2549. [PMID: 31985137 DOI: 10.1111/1462-2920.14924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 01/22/2020] [Indexed: 12/31/2022]
Abstract
The metabolically versatile Pseudomonas aeruginosa inhabits biotic and abiotic environments including the niche of cystic fibrosis (CF) airways. This study investigated how the adaptation to CF lungs affects the within-clone fitness of P. aeruginosa to grow and persist in liquid cultures in the presence of the clonal ancestors. Longitudinal clonal P. aeruginosa isolates that had been collected from 12 CF donors since the onset of colonization for up to 30 years was subjected to within-clone competition experiments. The relative quantities of individual strains were determined by marker-free amplicon sequencing of multiplex PCR products of strain-specific nucleotide sequence variants, a novel method that is generally applicable to studies in evolutionary genetics and microbial ecology with real-world strain collections. For 10 of the 12 examined patient courses, P. aeruginosa isolates of the first years of colonization grew faster in the presence of their clonal progeny than alone. Single growth of individual strains showed no temporal trend with colonization time, but in co-culture, the early isolates out-competed their clonal progeny. Irrespective of the genetic make-up of the clone and its genomic microevolution in CF lungs, the early isolates expressed fitness traits to win the within-clone competition that were absent in their progeny.
Collapse
Affiliation(s)
- Nina Cramer
- Clinic for Paediatric Pneumology, Allergology and Neonatology, Clinical Research Group 'Pseudomonas Genomics', Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, Hannover, Germany
| | - Sebastian Fischer
- Clinic for Paediatric Pneumology, Allergology and Neonatology, Clinical Research Group 'Pseudomonas Genomics', Hannover, Germany
| | - Silke Hedtfeld
- Clinic for Paediatric Pneumology, Allergology and Neonatology, Clinical Research Group 'Pseudomonas Genomics', Hannover, Germany
| | - Marie Dorda
- Research Core Unit Genomics, Hannover, Germany
| | - Burkhard Tümmler
- Clinic for Paediatric Pneumology, Allergology and Neonatology, Clinical Research Group 'Pseudomonas Genomics', Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, Hannover, Germany
| |
Collapse
|
47
|
Park H, McGill SL, Arnold AD, Carlson RP. Pseudomonad reverse carbon catabolite repression, interspecies metabolite exchange, and consortial division of labor. Cell Mol Life Sci 2020; 77:395-413. [PMID: 31768608 PMCID: PMC7015805 DOI: 10.1007/s00018-019-03377-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/04/2019] [Accepted: 11/12/2019] [Indexed: 10/25/2022]
Abstract
Microorganisms acquire energy and nutrients from dynamic environments, where substrates vary in both type and abundance. The regulatory system responsible for prioritizing preferred substrates is known as carbon catabolite repression (CCR). Two broad classes of CCR have been documented in the literature. The best described CCR strategy, referred to here as classic CCR (cCCR), has been experimentally and theoretically studied using model organisms such as Escherichia coli. cCCR phenotypes are often used to generalize universal strategies for fitness, sometimes incorrectly. For instance, extremely competitive microorganisms, such as Pseudomonads, which arguably have broader global distributions than E. coli, have achieved their success using metabolic strategies that are nearly opposite of cCCR. These organisms utilize a CCR strategy termed 'reverse CCR' (rCCR), because the order of preferred substrates is nearly reverse that of cCCR. rCCR phenotypes prefer organic acids over glucose, may or may not select preferred substrates to optimize growth rates, and do not allocate intracellular resources in a manner that produces an overflow metabolism. cCCR and rCCR have traditionally been interpreted from the perspective of monocultures, even though most microorganisms live in consortia. Here, we review the basic tenets of the two CCR strategies and consider these phenotypes from the perspective of resource acquisition in consortia, a scenario that surely influenced the evolution of cCCR and rCCR. For instance, cCCR and rCCR metabolism are near mirror images of each other; when considered from a consortium basis, the complementary properties of the two strategies can mitigate direct competition for energy and nutrients and instead establish cooperative division of labor.
Collapse
Affiliation(s)
- Heejoon Park
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, USA
- Center for Biofilm Engineering, Montana State University, Bozeman, USA
| | - S Lee McGill
- Department of Microbiology and Immunology, Montana State University, Bozeman, USA
- Center for Biofilm Engineering, Montana State University, Bozeman, USA
| | - Adrienne D Arnold
- Department of Microbiology and Immunology, Montana State University, Bozeman, USA
- Center for Biofilm Engineering, Montana State University, Bozeman, USA
| | - Ross P Carlson
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, USA.
- Department of Microbiology and Immunology, Montana State University, Bozeman, USA.
- Center for Biofilm Engineering, Montana State University, Bozeman, USA.
| |
Collapse
|
48
|
Interdependency of Respiratory Metabolism and Phenazine-Associated Physiology in Pseudomonas aeruginosa PA14. J Bacteriol 2020; 202:JB.00700-19. [PMID: 31767778 DOI: 10.1128/jb.00700-19] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 11/19/2019] [Indexed: 12/16/2022] Open
Abstract
Extracellular electron transfer (EET), the reduction of compounds that shuttle electrons to distal oxidants, can support bacterial survival when preferred oxidants are not directly accessible. EET has been shown to contribute to virulence in some pathogenic organisms and is required for current generation in mediator-based fuel cells. In several species, components of the electron transport chain (ETC) have been implicated in electron shuttle reduction, raising the question of how shuttling-based metabolism is integrated with primary routes of metabolic electron flow. The clinically relevant bacterium Pseudomonas aeruginosa can utilize carbon sources (i.e., electron donors) covering a broad range of reducing potentials and possesses a branched ETC that can be modulated to optimize respiratory efficiency. It also produces electron shuttles called phenazines that facilitate intracellular redox balancing, increasing the complexity of its metabolic potential. In this study, we investigated the reciprocal influence of respiratory metabolism and phenazine-associated physiology in P. aeruginosa PA14. We found that phenazine production affects respiratory activity and terminal oxidase gene expression and that carbon source identity influences the mechanisms enabling phenazine reduction. Furthermore, we found that growth in biofilms, a condition for which phenazine metabolism is critical to normal development and redox balancing, affects the composition of the P. aeruginosa phenazine pool. Together, these findings can aid interpretation of P. aeruginosa behavior during host infection and provide inroads to understanding the cross talk between primary metabolism and shuttling-based physiology in the diverse bacteria that carry out EET.IMPORTANCE The clinically relevant pathogen Pseudomonas aeruginosa uses diverse organic compounds as electron donors and possesses multiple enzymes that transfer electrons from central metabolism to O2 These pathways support a balanced intracellular redox state and produce cellular energy. P. aeruginosa also reduces secondary metabolites called phenazines to promote redox homeostasis and virulence. In this study, we examined the reciprocal relationship between these primary and secondary routes of electron flow. We found that phenazines affect respiratory function and that the complement of phenazines produced is strongly affected by growth in assemblages called biofilms. These results provide a more nuanced understanding of P. aeruginosa redox metabolism and may inform strategies for treating persistent infections caused by this bacterium.
Collapse
|
49
|
Magalhães AP, Jorge P, Pereira MO. Pseudomonas aeruginosa and Staphylococcus aureus communication in biofilm infections: insights through network and database construction. Crit Rev Microbiol 2019; 45:712-728. [DOI: 10.1080/1040841x.2019.1700209] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Andreia Patrícia Magalhães
- CEB - Centre of Biological Engineering, LIBRO - Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
| | - Paula Jorge
- CEB - Centre of Biological Engineering, LIBRO - Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, LIBRO - Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
| |
Collapse
|
50
|
Dethlefsen S, Jäger C, Klockgether J, Schomburg D, Tümmler B. Metabolite profiling of the cold adaptation of Pseudomonas putida KT2440 and cold-sensitive mutants. ENVIRONMENTAL MICROBIOLOGY REPORTS 2019; 11:777-783. [PMID: 31503400 DOI: 10.1111/1758-2229.12793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/02/2019] [Accepted: 09/08/2019] [Indexed: 06/10/2023]
Abstract
Free-living bacteria such as Pseudomonas putida are frequently exposed to temperature shifts and non-optimal growth conditions. We compared the transcriptome and metabolome of the cold adaptation of P. putida KT2440 and isogenic cold-sensitive transposon mutants carrying transposons in their cbrA, cbrB, pcnB, vacB, and bipA genes. Pseudomonas putida changes the mRNA expression of about 43% of all annotated open reading frames during this initial phase of cold adaptation, but only a small number of 6-93 genes were differentially expressed at 10°C between the wild-type strain and the individual mutants. The spectrum of metabolites underwent major changes during cold adaptation particularly in the mutants. Both the KT2440 strain and the mutants increased the levels of the most abundant sugars and amino acids which were more pronounced in the cold-sensitive mutants. All mutants depleted their pools for core metabolites of aromatic and sugar metabolism, but increased their pool of polar amino acids which should be advantageous to cope with the cold stress.
Collapse
Affiliation(s)
- Sarah Dethlefsen
- Molecular Pathology of Cystic Fibrosis and Pseudomonas Genomics, Clinical Research Group, Hannover Medical School, 30625, Hannover, Germany
| | - Christian Jäger
- Department of Biochemistry and Bioinformatics, Institute for Biochemistry and Biotechnology, Technische Universität Braunschweig, 38106, Braunschweig, Germany
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jens Klockgether
- Molecular Pathology of Cystic Fibrosis and Pseudomonas Genomics, Clinical Research Group, Hannover Medical School, 30625, Hannover, Germany
| | - Dietmar Schomburg
- Department of Biochemistry and Bioinformatics, Institute for Biochemistry and Biotechnology, Technische Universität Braunschweig, 38106, Braunschweig, Germany
| | - Burkhard Tümmler
- Molecular Pathology of Cystic Fibrosis and Pseudomonas Genomics, Clinical Research Group, Hannover Medical School, 30625, Hannover, Germany
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research, Hannover, Germany
| |
Collapse
|