1
|
Ma W, Luo J, Liu H, Du Q, Hao T, Jiang Y, Huang Z, Lan L, Li Z, Li T. Chemoenzymatic Synthesis of Highly O-Glycosylated MUC7 Glycopeptides for Probing Inhibitory Activity against Pseudomonas aeruginosa Biofilm Formation. Angew Chem Int Ed Engl 2025; 64:e202424312. [PMID: 39996424 DOI: 10.1002/anie.202424312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 02/26/2025]
Abstract
MUC7, a highly glycosylated protein in saliva and respiratory tract, plays potential roles in facilitating bacterial clearance and preventing microbial invasion. The complexity of glycan structures and multiplicity of glycosylation sites of MUC7 make it very difficult to explore accurate biofunctions against pathogens. Here, we report an efficiently convergent chemoenzymatic approach to firstly synthesize highly O-glycosylated MUC7 glycopeptides with nine glycosylation sites bearing various glycoforms via the combined use of hydrophobic tag-assisted liquid-phase peptide synthesis and enzymatic-catalyzed glycan elongation. Biological evaluations reveal that different glycoforms of synthetic MUC7 glycopeptides mediate unique activities against biofilm formation of Pseudomonas aeruginosa, among which sialylated MUC7 glycopeptide exhibits better inhibitory activity and has the potential to develop antibacterial drugs.
Collapse
Affiliation(s)
- Wenjing Ma
- State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junyuan Luo
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, China
| | - Huan Liu
- State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Qi Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, NHC Key Laboratory of Glycoconjugates Research, and Liver Cancer Institute of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Tianhui Hao
- State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yinyu Jiang
- State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhengwei Huang
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Lefu Lan
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- Anhui Province Key Laboratory of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Zhonghua Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, NHC Key Laboratory of Glycoconjugates Research, and Liver Cancer Institute of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Tiehai Li
- State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
2
|
Sherafati S, Gholami M, Ebrahimzadeh MA, Goli HR. Inhibitory effect of naringin, naringenin, and crocin on biofilm formation and lecA gene expression in Pseudomonas aeruginosa clinical isolates. Microb Pathog 2025; 205:107652. [PMID: 40311942 DOI: 10.1016/j.micpath.2025.107652] [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: 05/14/2024] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/03/2025]
Abstract
Biofilm formation by Pseudomonas aeruginosa is a considerable challenge in treating infections. We aimed to investigate the inhibitory effect of crocin, naringin, and naringenin on biofilm formation capacity and the expression of the lecA gene by this organism. One hundred unrepeated P. aeruginosa isolates were collected from hospitalized patients and were identified. The antibiotic resistance pattern of the isolates was determined using the disk agar diffusion method. The minimum inhibitory concentration (MIC) of naringin, naringenin, and crocin was determined by a micro broth dilution test. Then, the biofilm production ability of the isolates was evaluated before and after treatment with the investigated flavonoids using the microtiter plate test. Finally, the lecA gene expression of the isolates was checked before and after treatment with investigated flavonoids using the Real-time PCR method. Among 89 biofilm-producer isolates, 48 (53.93 %), 17 (19.1 %), and 24 (26.96 %) showed a strong, moderate, and weak biofilm formation ability. Biofilm-positive isolates were more resistant to all tested antibiotics. Also, among 41 multidrug-resistant (MDR) isolates, 33 (80.48 %) were strong biofilm producers (P-value = 0.01). A strong correlation was observed between the lecA gene expression and the biofilm production ability of the isolates (P-value = 0.000). The investigated flavonoids were significantly effective on biofilm production by P. aeruginosa. Among 10 strong-biofilm producers, all (100 %) showed a moderate ability to form biofilm after treatment with crocin (P-value = 0.02), and 6 (60 %) isolates had lost their ability to produce biofilm after treatment with the simultaneous use of crocin with ciprofloxacin or tobramycin (P-value = 0.000). Also, one isolate was grouped as biofilm-negative after treatment with naringin (P-value = 0.012). The crocin, naringin, and naringenin and their concurrent use of antibiotics decreased 2-8-fold of the lecA gene expression in strong biofilm-producer isolates (P-value˂0.05). Crocin, naringin, and naringenin can be used separately or simultaneously with antibiotics to inhibit biofilm and reduce the expression of virulence factors effective in biofilm production in P. aeruginosa.
Collapse
Affiliation(s)
- Sara Sherafati
- Molecular and Cell Biology Research Centre, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehrdad Gholami
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ali Ebrahimzadeh
- Pharmaceutical Sciences Research Center, School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamid Reza Goli
- Molecular and Cell Biology Research Centre, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
3
|
Fares M, Imberty A, Titz A. Bacterial lectins: multifunctional tools in pathogenesis and possible drug targets. Trends Microbiol 2025:S0966-842X(25)00083-6. [PMID: 40307096 DOI: 10.1016/j.tim.2025.03.007] [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: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 05/02/2025]
Abstract
Glycans are vital macromolecules with diverse biological roles, decoded by lectins - specialized carbohydrate-binding proteins crucial in pathogenesis. The WHO identifies bacterial antimicrobial resistance (AMR) as a critical global health challenge, necessitating innovative strategies that also target non-antibiotic pathways. Recent studies highlight bacterial lectins as key players in pathogenesis and promising therapeutic targets, with early clinical success using glycomimetics and vaccines to treat and prevent AMR-related infections. This review covers the current knowledge on bacterial lectins, their classifications, and roles in host recognition and adhesion, biofilm formation, cytotoxicity, and host immune evasion, with examples of well-characterized lectins. It also explores their therapeutic potential and highlights novel lectins with unknown functions, encouraging further research.
Collapse
Affiliation(s)
- Mario Fares
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, D-66123 Saarbrücken, Germany; Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany; Department of Chemistry, PharmaScienceHub (PSH), Saarland University, D-66123 Saarbrücken, Germany
| | - Anne Imberty
- University Grenoble Alpes, CNRS, CERMAV, 601 rue de la chimie, Grenoble 38000, France
| | - Alexander Titz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, D-66123 Saarbrücken, Germany; Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany; Department of Chemistry, PharmaScienceHub (PSH), Saarland University, D-66123 Saarbrücken, Germany.
| |
Collapse
|
4
|
Wang X, Ruan C, Shen C, Liao J, Wang D, Alvarez PJJ, Yu P. Synergistic Treatment of Reverse Osmosis Membrane Biofouling with Quorum Quenching Bacteria and Hitchhiking Phages. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:5647-5660. [PMID: 39945492 DOI: 10.1021/acs.est.4c12852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Biofilm formation, which is facilitated by quorum sensing (QS), significantly impairs the performance of pressure-driven membrane systems in water treatment. Herein, we present a quorum quenching (QQ)-phage phoresy system to control biofouling by disrupting QS-mediated interactions. This system, which is composed of the QQ bacterium Paenarthrobacter nicotinovorans as carriers and hitchhiking lytic phages infecting Pseudomonas aeruginosa with active QS systems, significantly decreased QS signal levels, inhibited the extracellular polymeric substance (EPS), and reduced bacterial abundance in mature biofilms. Transcriptomic analysis revealed that phage treatment upregulated QS and EPS synthesis genes in P. aeruginosa, but the QQ bacteria downregulated QS-related genes, weakening the bacterial EPS secretion and antiviral systems and facilitating phages to infect and lyse the target bacteria. Metabolomic profiling corroborated that the phoresy system disrupted pathways critical to biofilm stability, including the tricarboxylic acid cycle, carbohydrate metabolism, and amino acid metabolism. In off-site membrane cleaning experiments, the phoresy system promoted P. nicotinovorans colonization and replaced the niche of P. aeruginosa on the membrane surface, which restored membrane flux (i.e., 90% recovery in severely biofouling systems). Operation studies showed that the phoresy system reduced fouling rates, extended the membrane lifespan, and maintained salt rejection performance for reverse osmosis (RO) membrane systems. These findings highlight the potential of the QQ bacterium-phage system as a sustainable alternative to conventional chemical treatments that damage polymeric membranes.
Collapse
Affiliation(s)
- Xinjie Wang
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chujin Ruan
- Department of Environmental Microbiology, Swiss Federal Institute of Aquatic Science and Technology (Eawag), Dübendorf 8600, Switzerland
| | - Chaofeng Shen
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China
| | - Jingqiu Liao
- Department of Civil and Environmental Engineering, Virginia Tech, Blacksburg, Virginia 24060, United States
| | - Dongsheng Wang
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China
| | - Pedro J J Alvarez
- Department of Civil and Environmental Engineering and Rice WaTER Institute, Rice University, Houston, Texas 77005, United States
| | - Pingfeng Yu
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China
| |
Collapse
|
5
|
Reichhardt C, Matwichuk ML, Lewerke LT, Jacobs HM, Yan J, Parsek MR. Non-disruptive matrix turnover is a conserved feature of biofilm aggregate growth in paradigm pathogenic species. mBio 2025; 16:e0393524. [PMID: 39982068 PMCID: PMC11898600 DOI: 10.1128/mbio.03935-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/23/2025] [Indexed: 02/22/2025] Open
Abstract
Bacteria form multicellular aggregates called biofilms. A crucial component of these aggregates is a protective matrix that holds the community together. Biofilm matrix composition varies depending upon bacterial species but typically includes exopolysaccharides (EPS), proteins, and extracellular DNA. Pseudomonas aeruginosa is a model organism for the study of biofilms, and in non-mucoid biofilms, it uses the structurally distinct EPS Psl and Pel, the EPS-binding protein CdrA, and eDNA as key matrix components. An interesting phenomenon that we and others have observed is that the periphery of a biofilm aggregate can be EPS-rich and contain very few cells. In this study, we investigated two possible models of assembly and dynamics of this EPS-rich peripheral region: (i) newly synthesized EPS is inserted and incorporated into the existing EPS-rich region at the periphery during biofilm aggregate growth or (ii) EPS is continuously turned over and newly synthesized EPS is deposited at the outermost edge of the aggregate. Our results support the latter model. Specifically, we observed that new EPS is continually deposited at the aggregate periphery, which is necessary for continued aggregate growth but not aggregate stability. We made similar observations in another paradigm biofilm-forming species, Vibrio cholerae. This pattern of deposition raises the question of how EPS is retained. Specifically, for P. aeruginosa biofilms, the matrix adhesin CdrA is thought to retain EPS. However, current thinking is that cell-associated CdrA is responsible for this retention, and it is not clear how CdrA might function in the relatively cell-free aggregate periphery. We observed that CdrA is enzymatically degraded during aggregate growth without negatively impacting biofilm stability and that cell-free CdrA can partially maintain aggregation and Psl retention. Overall, this study shows that the matrix of P. aeruginosa biofilms undergoes both continuous synthesis of matrix material and matrix turnover to accommodate biofilm aggregate growth and that cell-free matrix can at least partially maintain biofilm aggregation and EPS localization. Furthermore, our similar observations for V. cholerae biofilms suggest that our findings may represent basic principles of aggregate assembly in bacteria. IMPORTANCE Here, we show that, to accommodate growing cellular biomass, newly produced Psl is deposited over existing Psl at the periphery of biofilm aggregates. We demonstrated that V. cholerae employs a similar mechanism with its biofilm matrix EPS, VPS. In addition, we found that the protease LasB is present in the biofilm matrix, resulting in degradation of CdrA to lower molecular weight cell-free forms. We then show that the released forms of CdrA are retained in the matrix and remain functional. Together, our findings support that the P. aeruginosa biofilm matrix is dynamic during the course of aggregate growth and that other species may employ similar mechanisms to remodel their matrix.
Collapse
Affiliation(s)
- Courtney Reichhardt
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | | | - Lincoln T. Lewerke
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Holly M. Jacobs
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Jing Yan
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - Matthew R. Parsek
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
6
|
You Z, Yu H, Zhang B, Liu Q, Xiong B, Li C, Qiao C, Dai L, Li J, Li W, Xin G, Liu Z, Li F, Song H. Engineering Exopolysaccharide Biosynthesis of Shewanella oneidensis to Promote Electroactive Biofilm Formation for Liquor Wastewater Treatment. ACS Synth Biol 2025; 14:373-383. [PMID: 39556104 DOI: 10.1021/acssynbio.4c00417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Microbial electrochemical systems (MESs), as a green and sustainable technology, can decompose organics in wastewater to recover bioelectricity. Electroactive biofilms, a microbial community structure encased in a self-produced matrix, play a decisive role in determining the efficiency of MESs. However, as an essential component of the biofilm matrix, the role of exopolysaccharides in electroactive biofilm formation and their influence on extracellular electron transfer (EET) have been rarely studied. Herein, to explore the effects of exopolysaccharides on biofilm formation and EET rate, we first inhibited the key genes responsible for exopolysaccharide biosynthesis (namely, so_3171, so_3172, so_3177, and so_3178) by using antisense RNA in Shewanella oneidensis MR-1. Then, to explore the underlying mechanisms why inhibition of exopolysaccharide synthesis could enhance biofilm formation and promote the EET rate, we characterized cell physiology and electrophysiology. The results showed inhibition of exopolysaccharide biosynthesis not only altered cell surface hydrophobicity and promoted intercellular adhesion and aggregation, but also increased biosynthesis of c-type cytochromes and decreased interfacial resistance, thus promoting electroactive biofilm formation and improving the EET rate of S. oneidensis. Lastly, to evaluate and intensify the capability of exopolysaccharide-reduced strains in harvesting electrical energy from actual liquor wastewater, engineered strain Δ3171-as3177 was further constructed to treat an actual thin stillage. The results showed that the output power density reached 380.98 mW m-2, 11.1-fold higher than that of WT strain, which exhibited excellent capability of harvesting electricity from actual liquor wastewater. This study sheds light on the underlying mechanism of how inhibition of exopolysaccharides impacts electroactive biofilm formation and EET rate, which suggested that regulating exopolysaccharide biosynthesis is a promising avenue for increasing the EET rate.
Collapse
Affiliation(s)
- Zixuan You
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Huan Yu
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Baocai Zhang
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Qijing Liu
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Bo Xiong
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Chao Li
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Chunxiao Qiao
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Longhai Dai
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Jianxun Li
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Wenwei Li
- Chinese Academy of Sciences (CAS) Key Laboratory of Urban Pollutant Conversion, Department of Chemistry, University of Science and Technology of China, Hefei 230026, China
| | - Guosheng Xin
- School of Life and Sciences, Ningxia University, Yinchuan, 750021, China
| | - Zhanying Liu
- Center for Energy Conservation and Emission Reduction in Fermentation Industry in Inner Mongolia, Engineering Research Center of Inner Mongolia for Green Manufacturing in Bio-fermentation Industry, and School of Chemical Engineering, Inner Mongolia University of Technology, Inner Mongolia, Hohhot 010051, China
| | - Feng Li
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Hao Song
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Key Laboratory of Systems Bioengineering, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| |
Collapse
|
7
|
Roque‐Borda CA, Primo LMDG, Medina‐Alarcón KP, Campos IC, Nascimento CDF, Saraiva MMS, Berchieri Junior A, Fusco‐Almeida AM, Mendes‐Giannini MJS, Perdigão J, Pavan FR, Albericio F. Antimicrobial Peptides: A Promising Alternative to Conventional Antimicrobials for Combating Polymicrobial Biofilms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410893. [PMID: 39530703 PMCID: PMC11714181 DOI: 10.1002/advs.202410893] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Polymicrobial biofilms adhere to surfaces and enhance pathogen resistance to conventional treatments, significantly contributing to chronic infections in the respiratory tract, oral cavity, chronic wounds, and on medical devices. This review examines antimicrobial peptides (AMPs) as a promising alternative to traditional antibiotics for treating biofilm-associated infections. AMPs, which can be produced as part of the innate immune response or synthesized therapeutically, have broad-spectrum antimicrobial activity, often disrupting microbial cell membranes and causing cell death. Many specifically target negatively charged bacterial membranes, unlike host cell membranes. Research shows AMPs effectively inhibit and disrupt polymicrobial biofilms and can enhance conventional antibiotics' efficacy. Preclinical and clinical research is advancing, with animal studies and clinical trials showing promise against multidrug-resistant bacteria and fungi. Numerous patents indicate increasing interest in AMPs. However, challenges such as peptide stability, potential cytotoxicity, and high production costs must be addressed. Ongoing research focuses on optimizing AMP structures, enhancing stability, and developing cost-effective production methods. In summary, AMPs offer a novel approach to combating biofilm-associated infections, with their unique mechanisms and synergistic potential with existing antibiotics positioning them as promising candidates for future treatments.
Collapse
Affiliation(s)
- Cesar Augusto Roque‐Borda
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
- Vicerrectorado de InvestigaciónUniversidad Católica de Santa MaríaArequipa04000Peru
| | - Laura Maria Duran Gleriani Primo
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Kaila Petronila Medina‐Alarcón
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Isabella C. Campos
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Camila de Fátima Nascimento
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Mauro M. S. Saraiva
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Angelo Berchieri Junior
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Ana Marisa Fusco‐Almeida
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Maria José Soares Mendes‐Giannini
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - João Perdigão
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
| | - Fernando Rogério Pavan
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Fernando Albericio
- Peptide Science LaboratorySchool of Chemistry and PhysicsUniversity of KwaZulu‐NatalDurban4001South Africa
- CIBER‐BBNNetworking Centre on BioengineeringBiomaterials and Nanomedicineand Department of Organic ChemistryUniversity of BarcelonaBarcelona08028Spain
| |
Collapse
|
8
|
Zahorska E, Denig LM, Lienenklaus S, Kuhaudomlarp S, Tschernig T, Lipp P, Munder A, Gillon E, Minervini S, Verkhova V, Imberty A, Wagner S, Titz A. High-Affinity Lectin Ligands Enable the Detection of Pathogenic Pseudomonas aeruginosa Biofilms: Implications for Diagnostics and Therapy. JACS AU 2024; 4:4715-4728. [PMID: 39735928 PMCID: PMC11672137 DOI: 10.1021/jacsau.4c00670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 12/31/2024]
Abstract
Pseudomonas aeruginosa is a critical priority pathogen and causes life-threatening acute and biofilm-associated chronic infections. The choice of suitable treatment for complicated infections requires lengthy culturing for species identification from swabs or an invasive biopsy. To date, no fast, pathogen-specific diagnostic tools for P. aeruginosa infections are available. Here, we present the noninvasive pathogen-specific detection of P. aeruginosa using novel fluorescent probes that target the bacterial biofilm-associated lectins LecA and LecB. Several glycomimetic probes were developed to target these extracellular lectins and demonstrated to stain P. aeruginosa biofilms in vitro. Importantly, for the targeting of LecA an activity boost to low-nanomolar affinity could be achieved, which is essential for in vivo application. In vitro, the nanomolar divalent LecA-targeted imaging probe accumulated effectively in biofilms under flow conditions, independent of the fluorophore identity. Investigation of these glycomimetic imaging probes in a murine lung infection model and fluorescence imaging revealed accumulation at the infection site. These findings demonstrate the use of LecA- and LecB-targeting probes for the imaging of P. aeruginosa infections and suggest their potential as pathogen-specific diagnostics to accelerate the start of the appropriate treatment.
Collapse
Affiliation(s)
- Eva Zahorska
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| | - Lisa Marie Denig
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| | - Stefan Lienenklaus
- Hannover
Medical School, Institute of Laboratory
Animal Science, Hannover 30625, Germany
| | - Sakonwan Kuhaudomlarp
- Université
Grenoble Alpes, CNRS, CERMAV, Grenoble 38000, France
- Department
of Biochemistry, Faculty of Science, Mahidol
University, Bangkok 10400, Thailand
- Center
for Excellence in Protein and Enzyme Technology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Thomas Tschernig
- Medical
Faculty of Saarland University, Institute
of Anatomy and Cell Biology, Homburg/Saar, D-66421, Germany
| | - Peter Lipp
- Center
for Molecular Signaling (PZMS), Medical
Faculty of Saarland University, Homburg/Saar D-66421, Germany
| | - Antje Munder
- Department
of Pediatric Pneumology, Allergology and
Neonatology, Hannover Medical School, Carl Neuberg-Str. 1, Hannover D-30625, Germany
- Biomedical
Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover D-30625, Germany
| | - Emilie Gillon
- Université
Grenoble Alpes, CNRS, CERMAV, Grenoble 38000, France
| | - Saverio Minervini
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
| | - Varvara Verkhova
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| | - Anne Imberty
- Université
Grenoble Alpes, CNRS, CERMAV, Grenoble 38000, France
| | - Stefanie Wagner
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| | - Alexander Titz
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre
for Infection Research, Saarbrücken D-66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Braunschweig 38124, Germany
- Department
of Chemistry, Saarland University, Saarbrücken D-66123, Germany
- PharmaScienceHub, Saarland University, Saarbrücken D-66123, Germany
| |
Collapse
|
9
|
Smith OER, Bharat TAM. Architectural dissection of adhesive bacterial cell surface appendages from a "molecular machines" viewpoint. J Bacteriol 2024; 206:e0029024. [PMID: 39499080 PMCID: PMC7616799 DOI: 10.1128/jb.00290-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024] Open
Abstract
The ability of bacteria to interact with and respond to their environment is crucial to their lifestyle and survival. Bacterial cells routinely need to engage with extracellular target molecules, in locations spatially separated from their cell surface. Engagement with distant targets allows bacteria to adhere to abiotic surfaces and host cells, sense harmful or friendly molecules in their vicinity, as well as establish symbiotic interactions with neighboring cells in multicellular communities such as biofilms. Binding to extracellular molecules also facilitates transmission of information back to the originating cell, allowing the cell to respond appropriately to external stimuli, which is critical throughout the bacterial life cycle. This requirement of bacteria to bind to spatially separated targets is fulfilled by a myriad of specialized cell surface molecules, which often have an extended, filamentous arrangement. In this review, we compare and contrast such molecules from diverse bacteria, which fulfil a range of binding functions critical for the cell. Our comparison shows that even though these extended molecules have vastly different sequence, biochemical and functional characteristics, they share common architectural principles that underpin bacterial adhesion in a variety of contexts. In this light, we can consider different bacterial adhesins under one umbrella, specifically from the point of view of a modular molecular machine, with each part fulfilling a distinct architectural role. Such a treatise provides an opportunity to discover fundamental molecular principles governing surface sensing, bacterial adhesion, and biofilm formation.
Collapse
Affiliation(s)
- Olivia E. R. Smith
- Structural Studies Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Tanmay A. M. Bharat
- Structural Studies Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
10
|
Carey JN, Lamont S, Wozniak DJ, Dandekar AA, Parsek MR. Quorum sensing regulation of Psl polysaccharide production by Pseudomonas aeruginosa. J Bacteriol 2024; 206:e0031224. [PMID: 39530727 PMCID: PMC11656772 DOI: 10.1128/jb.00312-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Pseudomonas aeruginosa is a common opportunistic pathogen and a model organism for studying bacterial sociality. A social behavior of P. aeruginosa that is critical for its success as a pathogen is its ability to form protective biofilms. Many of P. aeruginosa's social phenotypes are regulated by quorum sensing-a type of cell-cell communication that allows bacteria to respond to population density. Although biofilm formation is known to be affected by quorum sensing, evidence for direct regulation of biofilm production by quorum regulators has remained elusive. In this work, we show that production of the major biofilm matrix polysaccharide Psl in P. aeruginosa PAO1 is regulated by the quorum regulators LasR and RhlR in stationary-phase cultures. Secretion of Psl into the culture medium requires LasR, RhlR, and the quorum signal molecules N-3-oxo-dodecanoyl-homoserine lactone and N-butanoyl homoserine lactone. Psl production in strains unable to synthesize the homoserine lactone signals can be restored by exogenous introduction of the signal molecules. We found that LasR and RhlR perform different roles in the regulation of Psl production: LasR acts at the promoter of the psl operon and activates transcription of the Psl biosynthetic genes, while RhlR activates translation of the psl transcripts. This work contributes to our understanding of the overlapping but distinct functions of the Las and Rhl quorum-sensing systems and implicates both in the direct regulation of biofilm matrix production.IMPORTANCEPseudomonas aeruginosa biofilms are responsible for many treatment-resistant infections in humans. Many cooperative behaviors in P. aeruginosa are controlled by quorum sensing, but evidence for a direct role of quorum sensing in the regulation of biofilm matrix production has been scant. In this work, we show that the Las and Rhl quorum-sensing systems have distinct roles in regulating production of the matrix polysaccharide Psl and that this regulation happens at the level of transcription (Las) and translation (Rhl) of the psl operon. These findings deepen our understanding of overlapping functions of Las and Rhl quorum sensing and the complex regulation of biofilm development in P. aeruginosa.
Collapse
Affiliation(s)
- Jeffrey N. Carey
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Sabrina Lamont
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Daniel J. Wozniak
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Ajai A. Dandekar
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Matthew R. Parsek
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
11
|
Xiang SL, Xu KZ, Yin LJ, Rao Y, Wang B, Jia AQ. Dopamine, an exogenous quorum sensing signaling molecule or a modulating factor in Pseudomonas aeruginosa? Biofilm 2024; 8:100208. [PMID: 39036334 PMCID: PMC11260039 DOI: 10.1016/j.bioflm.2024.100208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 07/23/2024] Open
Abstract
Pseudomonas aeruginosa is recognized globally as an opportunistic pathogen of considerable concern due to its high virulence and pathogenicity, especially in immunocompromised individuals. While research has identified several endogenous quorum sensing (QS) signaling molecules that enhance the virulence and pathogenicity of P. aeruginosa, investigations on exogenous QS signaling molecules or modulating factors remain limited. This study found that dopamine serves as an exogenous QS signaling molecule or modulating factor of P. aeruginosa PAO1, enhancing the production of virulence factors and biofilms. Compared to the control group, treatment with 40 μM dopamine resulted in a 33.1 % increase in biofilm formation, 68.1 % increase in swimming mobility, 63.1 % increase in swarming mobility, 147.2 % increase in the signaling molecule 3-oxo-C12-HSL, and 50.5 %, 28.5 %, 27.0 %, and 33.2 % increases in the virulence factors alginate, rhamnolipids, protease, and pyocyanin, respectively. This study further explored the mechanism of dopamine regulating the biofilm formation and virulence of P. aeruginosa PAO1 through transcriptome and metabolome. Transcriptomic analysis showed that dopamine promoted the expression of virulence genes psl, alg, lasA, rhlABC, rml, and phz in P. aeruginosa PAO1. Metabolomic analysis revealed changes in the concentrations of tryptophan, pyruvate, ethanolamine, glycine, 3-hydroxybutyric acid, and alizarin. Furthermore, KEGG enrichment analysis of altered genes and metabolites indicated that dopamine enhanced phenylalanine, tyrosine, and tryptophan in P. aeruginosa PAO1. The results of this study will contribute to the development of novel exogenous QS signaling molecules or modulating factors and advance our understanding of the interactions between P. aeruginosa and the host environment.
Collapse
Affiliation(s)
- Shi-Liang Xiang
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Kai-Zhong Xu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Lu-Jun Yin
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Yong Rao
- School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Bo Wang
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
| | - Ai-Qun Jia
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
| |
Collapse
|
12
|
Tan X, Ma B, Wang X, Cui F, Li X, Li J. Characterization of Exopolysaccharides from Lactiplantibacillus plantarum PC715 and Their Antibiofilm Activity Against Hafnia alvei. Microorganisms 2024; 12:2229. [PMID: 39597618 PMCID: PMC11596824 DOI: 10.3390/microorganisms12112229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/29/2024] Open
Abstract
Exopolysaccharides (EPSs) secreted by lactic acid bacteria have the potential to enhance human health by showing various biological functions. This study investigated the biological role and antibiofilm properties of EPS715, a new neutral EPS produced by pickled vegetables originating from Lactobacillus plantarum PC715. The results indicate that EPS715 is primarily composed of rhamnose, glucose, and mannose. Its molecular weight (Mw) is 47.87 kDa, containing an α-glucoside linkage and an α-pyranose ring. It showed an amorphous morphology without a triple helix structure. Furthermore, EPS715 showed improved antioxidant activity. Specifically, its scavenging capacity of ABTS+ radicals, DPPH radicals, and the hydroxyl (·OH) reduction capacity at 5 mg/mL was 98.64 ± 2.70%, 97.37 ± 0.79%, and 1.64 ± 0.05%, respectively. Its maximal scavenging capacity was >40%, and the hydroxyl (·OH) radical scavenging ability was dose-dependent. Moreover, the biofilm of various pathogens including S. aureus, B. cereus, S. saprophyticus, Acinetobacter spp., and H. alvei was substantially dispersed and affected by EPS715, with a maximum inhibition rate of 78.17% for H. alvei. The possible mechanism by which EPS715 shows antibiofilm properties against the H. alvei may be attributed to its effects on the auto-aggregation, hydrophilic characteristics, and motility of Hafnia spp. Thus, EPS715 has significant antioxidant and antibiofilm characteristics that may hold substantial potential for applications in food and medicinal products.
Collapse
Affiliation(s)
- Xiqian Tan
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
- College of Food Science and Engineering & Institute of Marine Science and Technology, Bohai University, Jinzhou 121013, China
| | - Bingyu Ma
- College of Food Science and Engineering & Institute of Marine Science and Technology, Bohai University, Jinzhou 121013, China
| | - Xiaoqing Wang
- College of Food Science and Engineering & Institute of Marine Science and Technology, Bohai University, Jinzhou 121013, China
| | - Fangchao Cui
- College of Food Science and Engineering & Institute of Marine Science and Technology, Bohai University, Jinzhou 121013, China
| | - Xuepeng Li
- College of Food Science and Engineering & Institute of Marine Science and Technology, Bohai University, Jinzhou 121013, China
| | - Jianrong Li
- College of Food Science and Engineering & Institute of Marine Science and Technology, Bohai University, Jinzhou 121013, China
| |
Collapse
|
13
|
Mesas Vaz C, Guembe Mülberger A, Torrent Burgas M. The battle within: how Pseudomonas aeruginosa uses host-pathogen interactions to infect the human lung. Crit Rev Microbiol 2024:1-36. [PMID: 39381985 DOI: 10.1080/1040841x.2024.2407378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 08/11/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
Pseudomonas aeruginosa is a versatile Gram-negative pathogen known for its ability to invade the respiratory tract, particularly in cystic fibrosis patients. This review provides a comprehensive analysis of the multifaceted strategies for colonization, virulence, and immune evasion used by P. aeruginosa to infect the host. We explore the extensive protein arsenal of P. aeruginosa, including adhesins, exotoxins, secreted proteases, and type III and VI secretion effectors, detailing their roles in the infective process. We also address the unique challenge of treating diverse lung conditions that provide a natural niche for P. aeruginosa on the airway surface, with a particular focus in cystic fibrosis. The review also discusses the current limitations in treatment options due to antibiotic resistance and highlights promising future approaches that target host-pathogen protein-protein interactions. These approaches include the development of new antimicrobials, anti-attachment therapies, and quorum-sensing inhibition molecules. In summary, this review aims to provide a holistic understanding of the pathogenesis of P. aeruginosa in the respiratory system, offering insights into the underlying molecular mechanisms and potential therapeutic interventions.
Collapse
Affiliation(s)
- Carmen Mesas Vaz
- The Systems Biology of Infection Lab, Department of Biochemistry and Molecular Biology, Biosciences Faculty, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Alba Guembe Mülberger
- The Systems Biology of Infection Lab, Department of Biochemistry and Molecular Biology, Biosciences Faculty, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Marc Torrent Burgas
- The Systems Biology of Infection Lab, Department of Biochemistry and Molecular Biology, Biosciences Faculty, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
14
|
Badger-Emeka L, Emeka P, Thirugnanasambantham K, Alatawi AS. The Role of Pseudomonas aeruginosa in the Pathogenesis of Corneal Ulcer, Its Associated Virulence Factors, and Suggested Novel Treatment Approaches. Pharmaceutics 2024; 16:1074. [PMID: 39204419 PMCID: PMC11360345 DOI: 10.3390/pharmaceutics16081074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Pseudomonas aeruginosa (P. aeruginosa), is a diverse Gram-negative pathogen commonly associated with a wide spectrum of infections. It is indicated to be the most prevalent causative agent in the development of bacterial keratitis linked with the use of contact lens. Corneal infections attributed to P. aeruginosa frequently have poor clinical outcomes necessitating lengthy and costly therapies. Therefore, this review looks at the aetiology of P. aeruginosa bacterial keratitis as well as the bacterial drivers of its virulence and the potential therapeutics on the horizon. METHOD A literature review with the articles used for the review searched for and retrieved from PubMed, Scopus, and Google Scholar (date last accessed 1 April 2024). The keywords used for the search criteria were "Pseudomonas and keratitis, biofilm and cornea as well as P. aeruginosa". RESULTS P. aeruginosa is implicated in the pathogenesis of bacterial keratitis associated with contact lens usage. To reduce the potential seriousness of these infections, a variety of contact lens-cleaning options are available. However, continuous exposure to a range of antibiotics doses, from sub-inhibitory to inhibitory, has been shown to lead to the development of resistance to both antibiotics and disinfectant. Generally, there is a global public health concern regarding the rise of difficult-to-treat infections, particularly in the case of P. aeruginosa virulence in ocular infections. This study of the basic pathogenesis of a prevalent P. aeruginosa strain is therefore implicated in keratitis. To this effect, anti-virulence methods and phage therapy are being researched and developed in response to increasing antibiotic resistance. CONCLUSION This review has shown P. aeruginosa to be a significant cause of bacterial keratitis, particularly among users of contact lens. It also revealed treatment options, their advantages, and their drawbacks, including prospective candidates.
Collapse
Affiliation(s)
- Lorina Badger-Emeka
- Department of Biomedical Science, College of Medicine King Faisal University, Al Ahsa 31982, Saudi Arabia
| | - Promise Emeka
- Department of Pharmaceutical Science, College of Clinical Pharmacy, King Faisal University, Al Ahsa 31982, Saudi Arabia; (P.E.); (A.S.A.)
| | | | - Abdulaziz S. Alatawi
- Department of Pharmaceutical Science, College of Clinical Pharmacy, King Faisal University, Al Ahsa 31982, Saudi Arabia; (P.E.); (A.S.A.)
| |
Collapse
|
15
|
Mudgil U, Khullar L, Chadha J, Prerna, Harjai K. Beyond antibiotics: Emerging antivirulence strategies to combat Pseudomonas aeruginosa in cystic fibrosis. Microb Pathog 2024; 193:106730. [PMID: 38851361 DOI: 10.1016/j.micpath.2024.106730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that poses a significant threat to individuals suffering from cystic fibrosis (CF). The pathogen is highly prevalent in CF individuals and is responsible for chronic infection, resulting in severe tissue damage and poor patient outcome. Prolonged antibiotic administration has led to the emergence of multidrug resistance in P. aeruginosa. In this direction, antivirulence strategies achieving targeted inhibition of bacterial virulence pathways, including quorum sensing, efflux pumps, lectins, and iron chelators, have been explored against CF isolates of P. aeruginosa. Hence, this review article presents a bird's eye view on the pulmonary infections involving P. aeruginosa in CF patients by laying emphasis on factors contributing to bacterial colonization, persistence, and disease progression along with the current line of therapeutics against P. aeruginosa in CF. We further collate scientific literature and discusses various antivirulence strategies that have been tested against P. aeruginosa isolates from CF patients.
Collapse
Affiliation(s)
- Umang Mudgil
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Lavanya Khullar
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Jatin Chadha
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Prerna
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Kusum Harjai
- Department of Microbiology, Panjab University, Chandigarh, India.
| |
Collapse
|
16
|
David A, Tahrioui A, Tareau AS, Forge A, Gonzalez M, Bouffartigues E, Lesouhaitier O, Chevalier S. Pseudomonas aeruginosa Biofilm Lifecycle: Involvement of Mechanical Constraints and Timeline of Matrix Production. Antibiotics (Basel) 2024; 13:688. [PMID: 39199987 PMCID: PMC11350761 DOI: 10.3390/antibiotics13080688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen causing acute and chronic infections, especially in immunocompromised patients. Its remarkable adaptability and resistance to various antimicrobial treatments make it difficult to eradicate. Its persistence is enabled by its ability to form a biofilm. Biofilm is a community of sessile micro-organisms in a self-produced extracellular matrix, which forms a scaffold facilitating cohesion, cell attachment, and micro- and macro-colony formation. This lifestyle provides protection against environmental stresses, the immune system, and antimicrobial treatments, and confers the capacity for colonization and long-term persistence, often characterizing chronic infections. In this review, we retrace the events of the life cycle of P. aeruginosa biofilm, from surface perception/contact to cell spreading. We focus on the importance of extracellular appendages, mechanical constraints, and the kinetics of matrix component production in each step of the biofilm life cycle.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sylvie Chevalier
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, CBSA UR 4312, F-76000 Rouen, France
| |
Collapse
|
17
|
Zhang Q, Soulère L, Queneau Y. Amide bioisosteric replacement in the design and synthesis of quorum sensing modulators. Eur J Med Chem 2024; 273:116525. [PMID: 38801798 DOI: 10.1016/j.ejmech.2024.116525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/08/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
The prevention or control of bacterial infections requires continuous search for novel approaches among which bacterial quorum sensing inhibition is considered as a complementary antibacterial strategy. Quorum sensing, used by many different bacteria, functions through a cell-to-cell communication mechanism relying on chemical signals, referred to as autoinducers, such as N-acyl homoserine lactones (AHLs) which are the most common chemical signals in this system. Designing analogs of these autoinducers is one of the possible ways to interfere with quorum sensing. Since bioisosteres are powerful tools in medicinal chemistry, targeting analogs of AHLs or other signal molecules and mimics of known QS modulators built on amide bond bioisosteres is a relevant strategy in molecular design and synthetic routes. This review highlights the application of amide bond bioisosteric replacement in the design and synthesis of novel quorum sensing inhibitors.
Collapse
Affiliation(s)
- Qiang Zhang
- Hubei Key Laboratory of Purification and Application of Plant Anti-cancer Active Ingredients, Hubei University of Education, 129 Second Gaoxin Road, Wuhan 430205, China
| | - Laurent Soulère
- INSA Lyon, CNRS, Universite Claude Bernard Lyon 1, UMR 5246, ICBMS, Bât. E. Lederer, 1 rue Victor Grignard, F-69622, Villeurbanne, France
| | - Yves Queneau
- INSA Lyon, CNRS, Universite Claude Bernard Lyon 1, UMR 5246, ICBMS, Bât. E. Lederer, 1 rue Victor Grignard, F-69622, Villeurbanne, France.
| |
Collapse
|
18
|
Badillo-Larios NS, Turrubiartes-Martínez EA, Layseca-Espinosa E, González-Amaro R, Pérez-González LF, Niño-Moreno P. Interesting Cytokine Profile Caused by Clinical Strains of Pseudomonas aeruginosa MDR Carrying the exoU Gene. Int J Microbiol 2024; 2024:2748842. [PMID: 38974708 PMCID: PMC11227949 DOI: 10.1155/2024/2748842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 07/09/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen in HAIs with two facets: the most studied is the high rate of antimicrobial resistance, and the less explored is the long list of virulence factors it possesses. This study aimed to characterize the virulence genes carried by strains as well as the profile of cytokines related to inflammation, according to the resistance profile presented. This study aims to identify the virulence factors associated with MDR strains, particularly those resistant to carbapenems, and assess whether there is a cytokine profile that correlates with these characteristics. As methodology species were identified by classical microbiological techniques and confirmed by molecular biology, resistance levels were determined by the minimum inhibitory concentration and identification of MDR strains. Virulence factor genotyping was performed using PCR. In addition, biofilm production was assessed using crystal violet staining. Finally, the strains were cocultured with PBMC, and cell survival and the cytokines IL-1β, IL-6, IL-10, IL-8, and TNF-α were quantified using flow cytometry. Bacteremia and nosocomial pneumonia in adults are the most frequent types of infection. In the toxigenic aspect, genes corresponding to the type III secretion system were present in at least 50% of cases. In addition, PBMC exposed to strains of four different categories according to their resistance and toxicity showed a differential pattern of cytokine expression, a decrease in IL-10, IL-6, and IL-8, and an over-secretion of IL-1b. In conclusion, the virulence genes showed a differentiated appearance for the two most aggressive exotoxins of T3SS (exoU and exoS) in multidrug-resistant strains. Moreover, the cytokine profile displays a low expression of cytokines with anti-inflammatory and proinflammatory effects in strains carrying the exoU gene.
Collapse
Affiliation(s)
- Nallely S. Badillo-Larios
- Center of Research in Health Sciences and BiomedicineFaculty of MedicineAutonomous University of San Luis Potosi, San Luis Potosi, Mexico
| | - Edgar Alejandro Turrubiartes-Martínez
- Center of Research in Health Sciences and BiomedicineFaculty of MedicineAutonomous University of San Luis Potosi, San Luis Potosi, Mexico
- Laboratory of Hematology, Faculty of Chemical SciencesAutonomous University of San Luis Potosi, San Luis Potosi, Mexico
| | - Esther Layseca-Espinosa
- Center of Research in Health Sciences and BiomedicineFaculty of MedicineAutonomous University of San Luis Potosi, San Luis Potosi, Mexico
- Faculty of MedicineAutonomous University of San Luis Potosi, San Luis Potosi, Mexico
| | - Roberto González-Amaro
- Center of Research in Health Sciences and BiomedicineFaculty of MedicineAutonomous University of San Luis Potosi, San Luis Potosi, Mexico
- Faculty of MedicineAutonomous University of San Luis Potosi, San Luis Potosi, Mexico
| | - Luis Fernando Pérez-González
- Faculty of MedicineAutonomous University of San Luis Potosi, San Luis Potosi, Mexico
- Central Hospital Dr. Ignacio Morones Prieto, San Luis Potosi, Mexico
| | - Perla Niño-Moreno
- Center of Research in Health Sciences and BiomedicineFaculty of MedicineAutonomous University of San Luis Potosi, San Luis Potosi, Mexico
- Genetics LaboratoryFaculty of Chemical SciencesAutonomous University of San Luis Potosi, San Luis Potosi, Mexico
| |
Collapse
|
19
|
Omran BA, Tseng BS, Baek KH. Nanocomposites against Pseudomonas aeruginosa biofilms: Recent advances, challenges, and future prospects. Microbiol Res 2024; 282:127656. [PMID: 38432017 DOI: 10.1016/j.micres.2024.127656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/10/2024] [Accepted: 02/17/2024] [Indexed: 03/05/2024]
Abstract
Pseudomonas aeruginosa is an opportunistic bacterial pathogen that causes life-threatening and persistent infections in immunocompromised patients. It is the culprit behind a variety of hospital-acquired infections owing to its multiple tolerance mechanisms against antibiotics and disinfectants. Biofilms are sessile microbial aggregates that are formed as a result of the cooperation and competition between microbial cells encased in a self-produced matrix comprised of extracellular polymeric constituents that trigger surface adhesion and microbial aggregation. Bacteria in biofilms exhibit unique features that are quite different from planktonic bacteria, such as high resistance to antibacterial agents and host immunity. Biofilms of P. aeruginosa are difficult to eradicate due to intrinsic, acquired, and adaptive resistance mechanisms. Consequently, innovative approaches to combat biofilms are the focus of the current research. Nanocomposites, composed of two or more different types of nanoparticles, have diverse therapeutic applications owing to their unique physicochemical properties. They are emerging multifunctional nanoformulations that combine the desired features of the different elements to obtain the highest functionality. This review assesses the recent advances of nanocomposites, including metal-, metal oxide-, polymer-, carbon-, hydrogel/cryogel-, and metal organic framework-based nanocomposites for the eradication of P. aeruginosa biofilms. The characteristics and virulence mechanisms of P. aeruginosa biofilms, as well as their devastating impact and economic burden are discussed. Future research addressing the potential use of nanocomposites as innovative anti-biofilm agents is emphasized. Utilization of nanocomposites safely and effectively should be further strengthened to confirm the safety aspects of their application.
Collapse
Affiliation(s)
- Basma A Omran
- Department of Biotechnology, Yeungnam University, Gyeongbuk, Gyeongsan 38541, Republic of Korea; Department of Processes Design & Development, Egyptian Petroleum Research Institute (EPRI), PO 11727, Nasr City, Cairo, Egypt
| | - Boo Shan Tseng
- School of Life Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA.
| | - Kwang-Hyun Baek
- Department of Biotechnology, Yeungnam University, Gyeongbuk, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
20
|
Chance DL, Wang W, Waters JK, Mawhinney TP. Insights on Pseudomonas aeruginosa Carbohydrate Binding from Profiles of Cystic Fibrosis Isolates Using Multivalent Fluorescent Glycopolymers Bearing Pendant Monosaccharides. Microorganisms 2024; 12:801. [PMID: 38674745 PMCID: PMC11051836 DOI: 10.3390/microorganisms12040801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Pseudomonas aeruginosa contributes to frequent, persistent, and, often, polymicrobial respiratory tract infections for individuals with cystic fibrosis (CF). Chronic CF infections lead to bronchiectasis and a shortened lifespan. P. aeruginosa expresses numerous adhesins, including lectins known to bind the epithelial cell and mucin glycoconjugates. Blocking carbohydrate-mediated host-pathogen and intra-biofilm interactions critical to the initiation and perpetuation of colonization offer promise as anti-infective treatment strategies. To inform anti-adhesion therapies, we profiled the monosaccharide binding of P. aeruginosa from CF and non-CF sources, and assessed whether specific bacterial phenotypic characteristics affected carbohydrate-binding patterns. Focusing at the cellular level, microscopic and spectrofluorometric tools permitted the solution-phase analysis of P. aeruginosa binding to a panel of fluorescent glycopolymers possessing distinct pendant monosaccharides. All P. aeruginosa demonstrated significant binding to glycopolymers specific for α-D-galactose, β-D-N-acetylgalactosamine, and β-D-galactose-3-sulfate. In each culture, a small subpopulation accounted for the binding. The carbohydrate anomeric configuration and sulfate ester presence markedly influenced binding. While this opportunistic pathogen from CF hosts presented with various colony morphologies and physiological activities, no phenotypic, physiological, or structural feature predicted enhanced or diminished monosaccharide binding. Important to anti-adhesive therapeutic strategies, these findings suggest that, regardless of phenotype or clinical source, P. aeruginosa maintain a small subpopulation that may readily associate with specific configurations of specific monosaccharides. This report provides insights into whole-cell P. aeruginosa carbohydrate-binding profiles and into the context within which successful anti-adhesive and/or anti-virulence anti-infective agents for CF must contend.
Collapse
Affiliation(s)
- Deborah L. Chance
- Department of Molecular Microbiology & Immunology, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Department of Pediatrics, University of Missouri School of Medicine, Columbia, MO 65212, USA;
| | - Wei Wang
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA;
| | - James K. Waters
- Experiment Station Chemical Laboratories, University of Missouri, Columbia, MO 65211, USA;
| | - Thomas P. Mawhinney
- Department of Pediatrics, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA;
- Experiment Station Chemical Laboratories, University of Missouri, Columbia, MO 65211, USA;
| |
Collapse
|
21
|
Wang X, Wang D, Lu H, Wang X, Wang X, Su J, Xia G. Strategies to Promote the Journey of Nanoparticles Against Biofilm-Associated Infections. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305988. [PMID: 38178276 DOI: 10.1002/smll.202305988] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/08/2023] [Indexed: 01/06/2024]
Abstract
Biofilm-associated infections are one of the most challenging healthcare threats for humans, accounting for 80% of bacterial infections, leading to persistent and chronic infections. The conventional antibiotics still face their dilemma of poor therapeutic effects due to the high tolerance and resistance led by bacterial biofilm barriers. Nanotechnology-based antimicrobials, nanoparticles (NPs), are paid attention extensively and considered as promising alternative. This review focuses on the whole journey of NPs against biofilm-associated infections, and to clarify it clearly, the journey is divided into four processes in sequence as 1) Targeting biofilms, 2) Penetrating biofilm barrier, 3) Attaching to bacterial cells, and 4) Translocating through bacterial cell envelope. Through outlining the compositions and properties of biofilms and bacteria cells, recent advances and present the strategies of each process are comprehensively discussed to combat biofilm-associated infections, as well as the combined strategies against these infections with drug resistance, aiming to guide the rational design and facilitate wide application of NPs in biofilm-associated infections.
Collapse
Affiliation(s)
- Xiaobo Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| | - Dan Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| | - Hongwei Lu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| | - Xiaowei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| | - Xuelei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| | - Jiayi Su
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| | - Guimin Xia
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| |
Collapse
|
22
|
Böhning J, Tarafder AK, Bharat TA. The role of filamentous matrix molecules in shaping the architecture and emergent properties of bacterial biofilms. Biochem J 2024; 481:245-263. [PMID: 38358118 PMCID: PMC10903470 DOI: 10.1042/bcj20210301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/16/2024]
Abstract
Numerous bacteria naturally occur within spatially organised, multicellular communities called biofilms. Moreover, most bacterial infections proceed with biofilm formation, posing major challenges to human health. Within biofilms, bacterial cells are embedded in a primarily self-produced extracellular matrix, which is a defining feature of all biofilms. The biofilm matrix is a complex, viscous mixture primarily composed of polymeric substances such as polysaccharides, filamentous protein fibres, and extracellular DNA. The structured arrangement of the matrix bestows bacteria with beneficial emergent properties that are not displayed by planktonic cells, conferring protection against physical and chemical stresses, including antibiotic treatment. However, a lack of multi-scale information at the molecular level has prevented a better understanding of this matrix and its properties. Here, we review recent progress on the molecular characterisation of filamentous biofilm matrix components and their three-dimensional spatial organisation within biofilms.
Collapse
Affiliation(s)
- Jan Böhning
- Structural Studies Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, U.K
| | - Abul K. Tarafder
- Structural Studies Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, U.K
| | - Tanmay A.M. Bharat
- Structural Studies Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, U.K
| |
Collapse
|
23
|
Potapova A, Garvey W, Dahl P, Guo S, Chang Y, Schwechheimer C, Trebino MA, Floyd KA, Phinney BS, Liu J, Malvankar NS, Yildiz FH. Outer membrane vesicles and the outer membrane protein OmpU govern Vibrio cholerae biofilm matrix assembly. mBio 2024; 15:e0330423. [PMID: 38206049 PMCID: PMC10865864 DOI: 10.1128/mbio.03304-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Biofilms are matrix-encased microbial communities that increase the environmental fitness and infectivity of many human pathogens including Vibrio cholerae. Biofilm matrix assembly is essential for biofilm formation and function. Known components of the V. cholerae biofilm matrix are the polysaccharide Vibrio polysaccharide (VPS), matrix proteins RbmA, RbmC, Bap1, and extracellular DNA, but the majority of the protein composition is uncharacterized. This study comprehensively analyzed the biofilm matrix proteome and revealed the presence of outer membrane proteins (OMPs). Outer membrane vesicles (OMVs) were also present in the V. cholerae biofilm matrix and were associated with OMPs and many biofilm matrix proteins suggesting that they participate in biofilm matrix assembly. Consistent with this, OMVs had the capability to alter biofilm structural properties depending on their composition. OmpU was the most prevalent OMP in the matrix, and its absence altered biofilm architecture by increasing VPS production. Single-cell force spectroscopy revealed that proteins critical for biofilm formation, OmpU, the matrix proteins RbmA, RbmC, Bap1, and VPS contribute to cell-surface adhesion forces at differing efficiency, with VPS showing the highest efficiency whereas Bap1 showing the lowest efficiency. Our findings provide new insights into the molecular mechanisms underlying biofilm matrix assembly in V. cholerae, which may provide new opportunities to develop inhibitors that specifically alter biofilm matrix properties and, thus, affect either the environmental survival or pathogenesis of V. cholerae.IMPORTANCECholera remains a major public health concern. Vibrio cholerae, the causative agent of cholera, forms biofilms, which are critical for its transmission, infectivity, and environmental persistence. While we know that the V. cholerae biofilm matrix contains exopolysaccharide, matrix proteins, and extracellular DNA, we do not have a comprehensive understanding of the majority of biofilm matrix components. Here, we discover outer membrane vesicles (OMVs) within the biofilm matrix of V. cholerae. Proteomic analysis of the matrix and matrix-associated OMVs showed that OMVs carry key matrix proteins and Vibrio polysaccharide (VPS) to help build biofilms. We also characterize the role of the highly abundant outer membrane protein OmpU in biofilm formation and show that it impacts biofilm architecture in a VPS-dependent manner. Understanding V. cholerae biofilm formation is important for developing a better prevention and treatment strategy framework.
Collapse
Affiliation(s)
- Anna Potapova
- Department of Microbiology and Environmental Toxicology, University of California-Santa Cruz, Santa Cruz, California, USA
| | - William Garvey
- Department of Microbiology and Environmental Toxicology, University of California-Santa Cruz, Santa Cruz, California, USA
| | - Peter Dahl
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
- Microbial Sciences Institute, Yale University, West Haven, Connecticut, USA
| | - Shuaiqi Guo
- Microbial Sciences Institute, Yale University, West Haven, Connecticut, USA
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
| | - Yunjie Chang
- Microbial Sciences Institute, Yale University, West Haven, Connecticut, USA
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
| | - Carmen Schwechheimer
- Department of Microbiology and Environmental Toxicology, University of California-Santa Cruz, Santa Cruz, California, USA
| | - Michael A. Trebino
- Department of Microbiology and Environmental Toxicology, University of California-Santa Cruz, Santa Cruz, California, USA
| | - Kyle A. Floyd
- Department of Microbiology and Environmental Toxicology, University of California-Santa Cruz, Santa Cruz, California, USA
| | - Brett S. Phinney
- Proteomics Core Facility, UC Davis Genome Center, University of California-Davis, Davis, California, USA
| | - Jun Liu
- Microbial Sciences Institute, Yale University, West Haven, Connecticut, USA
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nikhil S. Malvankar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
- Microbial Sciences Institute, Yale University, West Haven, Connecticut, USA
| | - Fitnat H. Yildiz
- Department of Microbiology and Environmental Toxicology, University of California-Santa Cruz, Santa Cruz, California, USA
| |
Collapse
|
24
|
Würstle S, Lee A, Kortright KE, Winzig F, An W, Stanley GL, Rajagopalan G, Harris Z, Sun Y, Hu B, Blazanin M, Hajfathalian M, Bollyky PL, Turner PE, Koff JL, Chan BK. Optimized preparation pipeline for emergency phage therapy against Pseudomonas aeruginosa at Yale University. Sci Rep 2024; 14:2657. [PMID: 38302552 PMCID: PMC10834462 DOI: 10.1038/s41598-024-52192-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
Bacteriophage therapy is one potential strategy to treat antimicrobial resistant or persistent bacterial infections, and the year 2021 marked the centennial of Felix d'Hérelle's first publication on the clinical applications of phages. At the Center for Phage Biology & Therapy at Yale University, a preparatory modular approach has been established to offer safe and potent phages for single-patient investigational new drug applications while recognizing the time constraints imposed by infection(s). This study provides a practical walkthrough of the pipeline with an Autographiviridae phage targeting Pseudomonas aeruginosa (phage vB_PaeA_SB, abbreviated to ΦSB). Notably, a thorough phage characterization and the evolutionary selection pressure exerted on bacteria by phages, analogous to antibiotics, are incorporated into the pipeline.
Collapse
Affiliation(s)
- Silvia Würstle
- Yale Center for Phage Biology and Therapy, Yale University, 165 Prospect Street, New Haven, CT, 06520, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, 06520, USA
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06519, USA
- Technical University of Munich, 81675, Munich, Germany
| | - Alina Lee
- Yale Center for Phage Biology and Therapy, Yale University, 165 Prospect Street, New Haven, CT, 06520, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, 06520, USA
| | - Kaitlyn E Kortright
- Yale Center for Phage Biology and Therapy, Yale University, 165 Prospect Street, New Haven, CT, 06520, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, 06520, USA
| | - Franziska Winzig
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, 06520, USA
- Technical University of Munich, 81675, Munich, Germany
| | - William An
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, 06520, USA
| | - Gail L Stanley
- Yale Center for Phage Biology and Therapy, Yale University, 165 Prospect Street, New Haven, CT, 06520, USA
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Govindarajan Rajagopalan
- Yale Center for Phage Biology and Therapy, Yale University, 165 Prospect Street, New Haven, CT, 06520, USA
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Zach Harris
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Ying Sun
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Buqu Hu
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Michael Blazanin
- Yale Center for Phage Biology and Therapy, Yale University, 165 Prospect Street, New Haven, CT, 06520, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, 06520, USA
| | - Maryam Hajfathalian
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Paul E Turner
- Yale Center for Phage Biology and Therapy, Yale University, 165 Prospect Street, New Haven, CT, 06520, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, 06520, USA
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06519, USA
- Program in Microbiology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Jonathan L Koff
- Yale Center for Phage Biology and Therapy, Yale University, 165 Prospect Street, New Haven, CT, 06520, USA.
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, 06519, USA.
| | - Benjamin K Chan
- Yale Center for Phage Biology and Therapy, Yale University, 165 Prospect Street, New Haven, CT, 06520, USA.
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
25
|
Vadakkan K, Ngangbam AK, Sathishkumar K, Rumjit NP, Cheruvathur MK. A review of chemical signaling pathways in the quorum sensing circuit of Pseudomonas aeruginosa. Int J Biol Macromol 2024; 254:127861. [PMID: 37939761 DOI: 10.1016/j.ijbiomac.2023.127861] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/10/2023]
Abstract
Pseudomonas aeruginosa, an increasingly common competitive and biofilm organism in healthcare infection with sophisticated, interlinked and hierarchic quorum systems (Las, Rhl, PQS, and IQS), creates the greatest threats to the medical industry and has rendered prevailing chemotherapy medications ineffective. The rise of multidrug resistance has evolved into a concerning and potentially fatal occurrence for human life. P. aeruginosa biofilm development is assisted by exopolysaccharides, extracellular DNA, proteins, macromolecules, cellular signaling and interaction. Quorum sensing is a communication process between cells that involves autonomous inducers and regulators. Quorum-induced infectious agent biofilms and the synthesis of virulence factors have increased disease transmission, medication resistance, infection episodes, hospitalizations and mortality. Hence, quorum sensing may be a potential therapeutical target for bacterial illness, and developing quorum inhibitors as an anti-virulent tool could be a promising treatment strategy for existing antibiotics. Quorum quenching is a prevalent technique for treating infections caused by microbes because it diminishes microbial pathogenesis and increases microbe biofilm sensitivity to antibiotics, making it a potential candidate for drug development. This paper examines P. aeruginosa quorum sensing, the hierarchy of quorum sensing mechanism, quorum sensing inhibition and quorum sensing inhibitory agents as a drug development strategy to supplement traditional antibiotic strategies.
Collapse
Affiliation(s)
- Kayeen Vadakkan
- Department of Biology, St. Mary's College, Thrissur, Kerala 680020, India; Manipur International University, Imphal, Manipur 795140, India.
| | | | - Kuppusamy Sathishkumar
- Rhizosphere Biology Laboratory, Department of Microbiology, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India; Department of Computational Biology, Institute of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai 602 105, Tamil Nadu, India
| | | | | |
Collapse
|
26
|
Zhou Y, Huang J, Wang G, Zhai Z, Ahmed MU, Xia X, Liu C, Jin Y, Pan X, Huang Y, Wu C, Zhang X. Polymyxin B sulfate inhalable microparticles with high-lectin-affinity sugar carriers for efficient treatment of biofilm-associated pulmonary infections. Sci Bull (Beijing) 2023; 68:3225-3239. [PMID: 37973467 DOI: 10.1016/j.scib.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/29/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
Pulmonary infections caused by multidrug-resistant bacteria have become a significant threat to human health. Bacterial biofilms exacerbate the persistence and recurrence of pulmonary infections, hindering the accessibility and effectiveness of antibiotics. In this study, a dry powder inhalation (DPI) consisting of polymyxin B sulfate (PMBS) inhalable microparticles and high-lectin-affinity (HLA) sugar (i.e., raffinose) carriers was developed for treating pulmonary infections and targeting bacterial lectins essential for biofilm growth. The formulated PMBS-HLA DPIs exhibited particle sizes of approximately 3 μm, and surface roughness varied according to the drug-to-carrier ratio. Formulation F5 (PMBS: raffinose = 10:90) demonstrated the highest fine particle fraction (FPF) value (64.86%), signifying its substantially enhanced aerosol performance, potentially attributable to moderate roughness and smallest mass median aerodynamic particle size. The efficacy of PMBS-HLA DPIs in inhibiting biofilm formation and eradicating mature biofilms was significantly improved with the addition of raffinose, suggesting the effectiveness of lectin-binding strategy for combating bacterial biofilm-associated infections. In rat models with acute and chronic pulmonary infections, F5 demonstrated superior bacterial killing and amelioration of inflammatory responses compared to spray-dried PMBS (F0). In conclusion, our HLA carrier-based formulation presents considerable potential for the efficient treatment of multidrug-resistant bacterial biofilm-associated pulmonary infections.
Collapse
Affiliation(s)
- Yue Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510006, China; College of Pharmacy, Jinan University, Guangzhou 510006, China
| | - Jiayuan Huang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Guanlin Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zizhao Zhai
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510006, China; College of Pharmacy, Jinan University, Guangzhou 510006, China
| | - Maizbha Uddin Ahmed
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette IN 47907, USA
| | - Xiao Xia
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510006, China; College of Pharmacy, Jinan University, Guangzhou 510006, China
| | - Cenfeng Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510006, China; College of Pharmacy, Jinan University, Guangzhou 510006, China
| | - Yuzhen Jin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510006, China; College of Pharmacy, Jinan University, Guangzhou 510006, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Ying Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510006, China; College of Pharmacy, Jinan University, Guangzhou 510006, China.
| | - Chuanbin Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510006, China; College of Pharmacy, Jinan University, Guangzhou 510006, China
| | - Xuejuan Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510006, China; College of Pharmacy, Jinan University, Guangzhou 510006, China.
| |
Collapse
|
27
|
Heredia-Ponce Z, Secchi E, Toyofuku M, Marinova G, Savorana G, Eberl L. Genotoxic stress stimulates eDNA release via explosive cell lysis and thereby promotes streamer formation of Burkholderia cenocepacia H111 cultured in a microfluidic device. NPJ Biofilms Microbiomes 2023; 9:96. [PMID: 38071361 PMCID: PMC10710452 DOI: 10.1038/s41522-023-00464-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
DNA is a component of biofilms, but the triggers of DNA release during biofilm formation and how DNA contributes to biofilm development are poorly investigated. One key mechanism involved in DNA release is explosive cell lysis, which is a consequence of prophage induction. In this article, the role of explosive cell lysis in biofilm formation was investigated in the opportunistic human pathogen Burkholderia cenocepacia H111 (H111). Biofilm streamers, flow-suspended biofilm filaments, were used as a biofilm model in this study, as DNA is an essential component of their matrix. H111 contains three prophages on chromosome 1 of its genome, and the involvement of each prophage in causing explosive cell lysis of the host and subsequent DNA and membrane vesicle (MV) release, as well as their contribution to streamer formation, were studied in the presence and absence of genotoxic stress. The results show that two of the three prophages of H111 encode functional lytic prophages that can be induced by genotoxic stress and their activation causes DNA and MVs release by explosive cell lysis. Furthermore, it is shown that the released DNA enables the strain to develop biofilm streamers, and streamer formation can be enhanced by genotoxic stress. Overall, this study demonstrates the involvement of prophages in streamer formation and uncovers an often-overlooked problem with the use of antibiotics that trigger the bacterial SOS response for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Zaira Heredia-Ponce
- Department of Plant and Microbial Biology, University of Zürich, 8008, Zürich, Switzerland
| | - Eleonora Secchi
- Institute of Environmental Engineering, Department of Civil, Environmental and Geomatic Engineering, ETH Zürich, 8093, Zürich, Switzerland
| | - Masanori Toyofuku
- Faculty of Life and Environmental Sciences, Microbiology Research Center for Sustainability (MiCS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, Japan
| | - Gabriela Marinova
- Department of Plant and Microbial Biology, University of Zürich, 8008, Zürich, Switzerland
| | - Giovanni Savorana
- Institute of Environmental Engineering, Department of Civil, Environmental and Geomatic Engineering, ETH Zürich, 8093, Zürich, Switzerland
| | - Leo Eberl
- Department of Plant and Microbial Biology, University of Zürich, 8008, Zürich, Switzerland.
| |
Collapse
|
28
|
Yu Y, Han F, Yang M, Zhang X, Chen Y, Yu M, Wang Y. Pseudomonas composti isolate from oyster digestive tissue specifically binds with norovirus GII.6 via Psl extracellular polysaccharide. Int J Food Microbiol 2023; 406:110369. [PMID: 37666026 DOI: 10.1016/j.ijfoodmicro.2023.110369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 09/06/2023]
Abstract
Oysters are recognized as important vectors for human norovirus transmission in the environment. Whether norovirus binds to bacteria in oyster digestive tissues (ODTs) remains unknown. To shed light on this concern, ODT-54 and ODT-32, positive for histo-blood group antigen (HBGA) -like substances, were isolated from ODTs and identified as Pseudomonas composti and Enterobacter cloacae, respectively. The binding of noroviruses (GII.4 and GII.6 P domains) to bacterial cells (ODT-32 and ODT-54; in situ assay) as well as extracted extracellular polysaccharides (EPSs; in vitro assay) was analyzed by flow cytometry, confocal laser scanning microscopy, ELISA, and gene knock-out mutants. ODT-32 bound to neither GII.4 nor GII.6 P domains, while ODT-54 specifically binds with GII.6 P domain through Psl, an exopolysaccharide encoded by the polysaccharide synthesis locus (psl), identified based on gene annotation, gene transcription, Psl specific staining, and ELISAs. These findings attest that ODT bacteria specifically bind with certain norovirus genotypes in a strain-dependent manner, contributing to a better understanding of the transmission and enrichment of noroviruses in the environment.
Collapse
Affiliation(s)
- Yongxin Yu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, China
| | - Feng Han
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; Ministry of Agriculture and Rural Affairs, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Mingshu Yang
- College of Food Science and Engineering, Hainan Tropical Ocean University, Sanya, China
| | - Xiaoya Zhang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Yunfei Chen
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Mingxia Yu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Yongjie Wang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, China.
| |
Collapse
|
29
|
Guo Q, Cheng Y, Fan Z, Wu W, Wu Z, Zhang X. Zwitterion‐conjugated Topological Glycomimics for Dual‐Blocking Effects to Eradicate Biofilm Infection. ADVANCED THERAPEUTICS 2023; 6. [DOI: 10.1002/adtp.202300217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Indexed: 01/06/2025]
Abstract
AbstractP. aeruginosa, a leading nosocomial pathogen, commonly causes chronic biofilm infections in tissues and biomedical devices, including wound infections, osteomyelitis, and infective endocarditis, heavily threatening life. The dynamic lifecycle of these biofilms leads to persistent generation, making it challenging to prevent and disperse these biofilms effectively. Herein, a topological eight‐arm zwitterion‐conjugated glycomimetics (PCBAA‐b‐PLAMA)8 to address this challenge by exerting a dual‐blocking effect on P. aeruginosa biofilms is introduced. Initially, carboxybetaine acrylate (CBAA) and 2‐lactobionamidoethyl methacrylate (LAMA) are introduced to the topological bromine‐based initiator to prepare (PCBAA‐b‐PLAMA)8. This copolymer demonstrats remarkable efficiency in dispersing P. aeruginosa biofilms, approximately up to 99%. This high efficacy can be attributed to the multivalent and triaxial interactions between LAMA and CBAA groups, which enable the capture of P. aeruginosa cells and the biofilm matrix. Furthermore, (PCBAA‐b‐PLAMA)8 efficiently inhibit the expression of resistance genes related to biofilm formation and antibiotic efflux, including cdrA, lasB, mexE, and mexH, regaining bacterial cell sensitivity to antibiotics and further facilitating the natural dispersal of biofilms. This study provides a generic dual‐blocking strategy for the efficient eliminating of biofilms from biomedical devices.
Collapse
Affiliation(s)
- Qianqian Guo
- Key Laboratory of Functional Polymer Materials of Ministry of Education Institute of Polymer Chemistry College of Chemistry Nankai University Tianjin 300071 China
- The State Key Laboratory of Functions and Applications of Medicinal Plants School of Pharmaceutical Sciences Guizhou Medical University University Town Guian New District Guizhou 550025 China
| | - Yijie Cheng
- Key Laboratory of Functional Polymer Materials of Ministry of Education Institute of Polymer Chemistry College of Chemistry Nankai University Tianjin 300071 China
| | - Zheng Fan
- State Key Laboratory of Medicinal Chemical Biology Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education Department of Microbiology College of Life Sciences Nankai University Tianjin 300071 China
| | - Weihui Wu
- State Key Laboratory of Medicinal Chemical Biology Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education Department of Microbiology College of Life Sciences Nankai University Tianjin 300071 China
| | - Zhongming Wu
- Department of Endocrinology Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong 250021 China
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education Institute of Polymer Chemistry College of Chemistry Nankai University Tianjin 300071 China
| |
Collapse
|
30
|
Keenan T, Hatton NE, Porter J, Vendeville JB, Wheatley DE, Ghirardello M, Wahart AJC, Ahmadipour S, Walton J, Galan MC, Linclau B, Miller GJ, Fascione MA. Reverse thiophosphorylase activity of a glycoside phosphorylase in the synthesis of an unnatural Manβ1,4GlcNAc library. Chem Sci 2023; 14:11638-11646. [PMID: 37920340 PMCID: PMC10619541 DOI: 10.1039/d3sc04169g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/28/2023] [Indexed: 11/04/2023] Open
Abstract
β-Mannosides are ubiquitous in nature, with diverse roles in many biological processes. Notably, Manβ1,4GlcNAc a constituent of the core N-glycan in eukaryotes was recently identified as an immune activator, highlighting its potential for use in immunotherapy. Despite their biological significance, the synthesis of β-mannosidic linkages remains one of the major challenges in glycoscience. Here we present a chemoenzymatic strategy that affords a series of novel unnatural Manβ1,4GlcNAc analogues using the β-1,4-d-mannosyl-N-acetyl-d-glucosamine phosphorylase, BT1033. We show that the presence of fluorine in the GlcNAc acceptor facilitates the formation of longer β-mannan-like glycans. We also pioneer a "reverse thiophosphorylase" enzymatic activity, favouring the synthesis of longer glycans by catalysing the formation of a phosphorolysis-stable thioglycoside linkage, an approach that may be generally applicable to other phosphorylases.
Collapse
Affiliation(s)
- Tessa Keenan
- Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - Natasha E Hatton
- Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - Jack Porter
- School of Chemical and Physical Sciences and Centre for Glycosciences, Keele University Keele, Staffordshire ST5 5BG UK
| | | | - David E Wheatley
- School of Chemistry, University of Southampton Highfield Southampton SO17 1BJ UK
| | - Mattia Ghirardello
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Alice J C Wahart
- School of Chemical and Physical Sciences and Centre for Glycosciences, Keele University Keele, Staffordshire ST5 5BG UK
| | - Sanaz Ahmadipour
- School of Chemical and Physical Sciences and Centre for Glycosciences, Keele University Keele, Staffordshire ST5 5BG UK
| | - Julia Walton
- Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - M Carmen Galan
- School of Chemistry, University of Bristol Cantock's Close Bristol BS8 1TS UK
| | - Bruno Linclau
- School of Chemistry, University of Southampton Highfield Southampton SO17 1BJ UK
- Department of Organic and Macromolecular Chemistry, Ghent University Campus Sterre, Krijgslaan 281-S4 Ghent 9000 Belgium
| | - Gavin J Miller
- School of Chemical and Physical Sciences and Centre for Glycosciences, Keele University Keele, Staffordshire ST5 5BG UK
| | - Martin A Fascione
- Department of Chemistry, University of York Heslington York YO10 5DD UK
| |
Collapse
|
31
|
Bonincontro G, Scuderi SA, Marino A, Simonetti G. Synergistic Effect of Plant Compounds in Combination with Conventional Antimicrobials against Biofilm of Staphylococcus aureus, Pseudomonas aeruginosa, and Candida spp. Pharmaceuticals (Basel) 2023; 16:1531. [PMID: 38004397 PMCID: PMC10675371 DOI: 10.3390/ph16111531] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Bacterial and fungal biofilm has increased antibiotic resistance and plays an essential role in many persistent diseases. Biofilm-associated chronic infections are difficult to treat and reduce the efficacy of medical devices. This global problem has prompted extensive research to find alternative strategies to fight microbial chronic infections. Plant bioactive metabolites with antibiofilm activity are known to be potential resources to alleviate this problem. The phytochemical screening of some medicinal plants showed different active groups, such as stilbenes, tannins, alkaloids, terpenes, polyphenolics, flavonoids, lignans, quinones, and coumarins. Synergistic effects can be observed in the interaction between plant compounds and conventional drugs. This review analyses and summarises the current knowledge on the synergistic effects of plant metabolites in combination with conventional antimicrobials against biofilms of Staphylococcus aureus, Pseudomonas aeruginosa, and Candida albicans. The synergism of conventional antimicrobials with plant compounds can modify and inhibit the mechanisms of acquired resistance, reduce undesirable effects, and obtain an appropriate therapeutic effect at lower doses. A deeper knowledge of these combinations and of their possible antibiofilm targets is needed to develop next-generation novel antimicrobials and/or improve current antimicrobials to fight drug-resistant infections attributed to biofilm.
Collapse
Affiliation(s)
- Graziana Bonincontro
- Department of Environmental Biology, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Roma, Italy;
| | - Sarah Adriana Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98100 Messina, Italy;
| | - Andreana Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98100 Messina, Italy;
| | - Giovanna Simonetti
- Department of Environmental Biology, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Roma, Italy;
| |
Collapse
|
32
|
Esfahani MB, Khodavandi A, Alizadeh F, Bahador N. Antibacterial and Anti-Biofilm Activities of Microbial Synthesized Silver and Magnetic Iron Oxide Nanoparticles Against Pseudomonas aeruginosa. IEEE Trans Nanobioscience 2023; 22:956-966. [PMID: 37071524 DOI: 10.1109/tnb.2023.3268138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
Pseudomonas aeruginosa is a human bacterial pathogen causing devastating diseases and equipped with various virulence factors like biofilm formation. Common antibiotic treatment has limited efficacy for the P. aeruginosa present in biofilms because of the increased resistance. In this study, we focused our attention on the antibacterial and anti-biofilm activities of various microbial synthesized silver (nano-Ag) and magnetic iron oxide (nano-Fe3O4) nanoparticles against clinical isolates of P. aeruginosa that displayed ceftazidime resistance. The nano-Ag and nano-Fe3O4 represented great antibacterial properties. Nano-Ag and nano-Fe3O4 exhibited a reduction in the biofilm formation by P. aeruginosa reference strain as determined by crystal violet and XTT assays and light microscopy method. Among all, nano-Ag-2 and 7 owing to inherent attributes and mechanisms of resistance in the bacterial biofilm, exhibited anti-biofilm efficacy against ceftazidime resistance clinical isolate of P. aeruginosa. Moreover, nano-Ag and nano-Fe3O4 changed the relative expression of biofilm-associated genes, PELA and PSLA in a concentration dependent manner by P. aeruginosa reference strain. As revealed by qRT-PCR, the expression levels of biofilm-associated genes were downregulated in P. aeruginosa biofilms treated with nano-Ag, while selected biofilm-associated genes were low expressed under treated with nano-Fe3O4. Results of the study demonstrate the potential of microbial synthesized nano-Ag-2 and 7 to act as anti-biofilm agents against ceftazidime resistance clinical isolate of P. aeruginosa. Molecular targeting of biofilm-associated genes by nano-Ag and nano-Fe3O4 may be candidate for new therapeutics against P. aeruginosa diseases.
Collapse
|
33
|
Xu T, Xiao Y, Wang H, Zhu J, Lu W, Chen W. Multiomics reveals the mechanism of B. longum in promoting the formation of mixed-species biofilms. Food Funct 2023; 14:8276-8290. [PMID: 37602484 DOI: 10.1039/d3fo01751f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
It has been found previously that Bifidobacterium longum, Bacteroides ovatus, Enterococcus faecalis, and Lactobacillus gasseri can form a biofilm better when co-cultured in vitro and B. longum is the core biofilm-formation-promoting strain in this community. B. longum is part of the core microbiota in the gut and is widely recognized as a probiotic. Therefore, it is necessary to explore its role in mixed-species biofilms through transcriptomics and metabolomics. Metabolomics showed that the increase in amino acid and purine content could promote biofilm formation. In transcriptomic analysis, many genes related to carbohydrate metabolism, amino acid metabolism, and environmental tolerance of B. longum were up-regulated. Combined with the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and Gene Ontology (GO) analysis, the differentially expressed genes (DEGs) of B. longum in mixed-species biofilms were mainly correlated to "quorum sensing (QS)", "ABC transporters", "biosynthesis of amino acids", "microbial metabolism in different environments", "carbohydrate metabolism" and "two-component system". In addition, the rpl and rps gene families, which function in the metabolism of organic substances and the biosynthesis of amino acids, were the core DEGs according to the analysis of the protein-protein interaction (PPI) network. Finally, by combining metabolomics and quorum sensing mechanisms, it was found that the metabolism of autoinducer peptides (proliylglycine and glycylleucine), N-acyl homoserine lactone (N-(3-oxo hydroxy) homoserine lactone), and AI-2 can promote the formation of biofilms, both mono- and mixed-species biofilms composed of B. longum. Our research enabled us to understand the critical role of B. longum in mixed-species biofilms and the interactions between biofilm metabolism and gut health. In addition, the generated knowledge will be of great significance for us to develop biofilm products with beneficial functions in future.
Collapse
Affiliation(s)
- Tao Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yue Xiao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hongchao Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jinlin Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
34
|
Abstract
Ralstonia solanacearum species complex (RSSC) strains are devastating plant pathogens distributed worldwide. The primary cell density-dependent gene expression system in RSSC strains is phc quorum sensing (QS). It regulates the expression of about 30% of all genes, including those related to cellular activity, primary and secondary metabolism, pathogenicity, and more. The phc regulatory elements encoded by the phcBSRQ operon and phcA gene play vital roles. RSSC strains use methyl 3-hydroxymyristate (3-OH MAME) or methyl 3-hydroxypalmitate (3-OH PAME) as the QS signal. Each type of RSSC strain has specificity in generating and receiving its QS signal, but their signaling pathways might not differ significantly. In this review, I describe the genetic and biochemical factors involved in QS signal input and the regulatory network and summarize control of the phc QS system, new cell-cell communications, and QS-dependent interactions with soil fungi.
Collapse
Affiliation(s)
- Kenji Kai
- Graduate School of Agriculture, Osaka Metropolitan University, Osaka, Japan;
| |
Collapse
|
35
|
Ghosh M, Raghav S, Ghosh P, Maity S, Mohela K, Jain D. Structural analysis of novel drug targets for mitigation of Pseudomonas aeruginosa biofilms. FEMS Microbiol Rev 2023; 47:fuad054. [PMID: 37771093 DOI: 10.1093/femsre/fuad054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 09/30/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogen responsible for acute and chronic, hard to treat infections. Persistence of P. aeruginosa is due to its ability to develop into biofilms, which are sessile bacterial communities adhered to substratum and encapsulated in layers of self-produced exopolysaccharides. These biofilms provide enhanced protection from the host immune system and resilience towards antibiotics, which poses a challenge for treatment. Various strategies have been expended for combating biofilms, which involve inhibiting biofilm formation or promoting their dispersal. The current remediation approaches offer some hope for clinical usage, however, treatment and eradication of preformed biofilms is still a challenge. Thus, identifying novel targets and understanding the detailed mechanism of biofilm regulation becomes imperative. Structure-based drug discovery (SBDD) provides a powerful tool that exploits the knowledge of atomic resolution details of the targets to search for high affinity ligands. This review describes the available structural information on the putative target protein structures that can be utilized for high throughput in silico drug discovery against P. aeruginosa biofilms. Integrating available structural information on the target proteins in readily accessible format will accelerate the process of drug discovery.
Collapse
Affiliation(s)
- Moumita Ghosh
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Shikha Raghav
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Puja Ghosh
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Swagatam Maity
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Kavery Mohela
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| | - Deepti Jain
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, Haryana-121001, India
| |
Collapse
|
36
|
Vo LH, Hong S, Stepler KE, Liyanaarachchi SM, Yang J, Nemes P, Poulin MB. Mapping protein-exopolysaccharide binding interaction in Staphylococcus epidermidis biofilms by live cell proximity labeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.29.555326. [PMID: 37693546 PMCID: PMC10491226 DOI: 10.1101/2023.08.29.555326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Bacterial biofilms consist of cells encased in an extracellular polymeric substance (EPS) composed of exopolysaccharides, extracellular DNA, and proteins that are critical for cell-cell adhesion and protect the cells from environmental stress, antibiotic treatments, and the host immune response. Degrading EPS components or blocking their production have emerged as promising strategies for prevention or dispersal of bacterial biofilms, but we still have little information about the specific biomolecular interactions that occur between cells and EPS components and how those interactions contribute to biofilm production. Staphylococcus epidermidis is a leading cause of nosocomial infections as a result of producing biofilms that use the exopolysaccharide poly-(1→6)-β-N-acetylglucosamine (PNAG) as a major structural component. In this study, we have developed a live cell proximity labeling approach combined with quantitative mass spectrometry-based proteomics to map the PNAG interactome of live S. epidermidis biofilms. Through these measurements we discovered elastin-binding protein (EbpS) as a major PNAG-interacting protein. Using live cell binding measurements, we found that the lysin motif (LysM) domain of EbpS specifically binds to PNAG present in S. epidermidis biofilms. Our work provides a novel method for the rapid identification of exopolysaccharide-binding proteins in live biofilms that will help to extend our understanding of the biomolecular interactions that are required for bacterial biofilm formation.
Collapse
Affiliation(s)
- Luan H. Vo
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Steven Hong
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Kaitlyn E. Stepler
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Sureshee M. Liyanaarachchi
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Jack Yang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Peter Nemes
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Myles B. Poulin
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
37
|
Tsumori C, Matsuo S, Murai Y, Kai K. Quorum Sensing-Dependent Invasion of Ralstonia solanacearum into Fusarium oxysporum Chlamydospores. Microbiol Spectr 2023; 11:e0003623. [PMID: 37367297 PMCID: PMC10433826 DOI: 10.1128/spectrum.00036-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Strains of the Ralstonia solanacearum species complex (RSSC), although known as the causative agent of bacterial wilt disease in plants, induce the chlamydospores of many fungal species and invade them through the spores. The lipopeptide ralstonins are the chlamydospore inducers produced by RSSC and are essential for this invasion. However, no mechanistic investigation of this interaction has been conducted. In this study, we report that quorum sensing (QS), which is a bacterial cell-cell communication, is important for RSSC to invade the fungus Fusarium oxysporum (Fo). ΔphcB, a deletion mutant of QS signal synthase, lost the ability to both produce ralstonins and invade Fo chlamydospores. The QS signal methyl 3-hydroxymyristate rescued these disabilities. In contrast, exogenous ralstonin A, while inducing Fo chlamydospores, failed to rescue the invasive ability. Gene-deletion and -complementation experiments revealed that the QS-dependent production of extracellular polysaccharide I (EPS I) is essential for this invasion. The RSSC cells adhered to Fo hyphae and formed biofilms there before inducing chlamydospores. This biofilm formation was not observed in the EPS I- or ralstonin-deficient mutant. Microscopic analysis showed that RSSC infection resulted in the death of Fo chlamydospores. Altogether, we report that the RSSC QS system is important for this lethal endoparasitism. Among the factors regulated by the QS system, ralstonins, EPS I, and biofilm are important parasitic factors. IMPORTANCE Ralstonia solanacearum species complex (RSSC) strains infect both plants and fungi. The phc quorum-sensing (QS) system of RSSC is important for parasitism on plants, because it allows them to invade and proliferate within the hosts by causing appropriate activation of the system at each infection step. In this study, we confirm that ralstonin A is important not only for Fusarium oxysporum (Fo) chlamydospore induction but also for RSSC biofilm formation on Fo hyphae. Extracellular polysaccharide I (EPS I) is also essential for biofilm formation, while the phc QS system controls these factors in terms of production. The present results advocate a new QS-dependent mechanism for the process by which a bacterium invades a fungus.
Collapse
Affiliation(s)
- Chiaki Tsumori
- Graduate School of Agriculture, Osaka Metropolitan University, Osaka, Japan
| | - Shoma Matsuo
- Graduate School of Agriculture, Osaka Metropolitan University, Osaka, Japan
| | - Yuta Murai
- Graduate School of Agriculture, Osaka Metropolitan University, Osaka, Japan
| | - Kenji Kai
- Graduate School of Agriculture, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
38
|
Reichhardt C. The Pseudomonas aeruginosa Biofilm Matrix Protein CdrA Has Similarities to Other Fibrillar Adhesin Proteins. J Bacteriol 2023; 205:e0001923. [PMID: 37098957 PMCID: PMC10210978 DOI: 10.1128/jb.00019-23] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023] Open
Abstract
The ability of bacteria to adhere to each other and both biotic and abiotic surfaces is key to biofilm formation, and one way that bacteria adhere is using fibrillar adhesins. Fibrillar adhesins share several key characteristics, including (i) they are extracellular, surface-associated proteins, (ii) they contain an adhesive domain as well as a repetitive stalk domain, and (iii) they are either a monomer or homotrimer (i.e., identical, coiled-coil) of a high molecular weight protein. Pseudomonas aeruginosa uses the fibrillar adhesin called CdrA to promote bacterial aggregation and biofilm formation. Here, the current literature on CdrA is reviewed, including its transcriptional and posttranslational regulation by the second messenger c-di-GMP as well as what is known about its structure and ability to interact with other molecules. I highlight its similarities to other fibrillar adhesins and discuss open questions that remain to be answered toward a better understanding of CdrA.
Collapse
Affiliation(s)
- Courtney Reichhardt
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
39
|
Zammuto V, Spanò A, Agostino E, Macrì A, De Pasquale C, Ferlazzo G, Rizzo MG, Nicolò MS, Guglielmino S, Gugliandolo C. Anti-Bacterial Adhesion on Abiotic and Biotic Surfaces of the Exopolysaccharide from the Marine Bacillus licheniformis B3-15. Mar Drugs 2023; 21:md21050313. [PMID: 37233507 DOI: 10.3390/md21050313] [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: 03/28/2023] [Revised: 05/05/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023] Open
Abstract
The eradication of bacterial biofilm represents a crucial strategy to prevent a clinical problem associated with microbial persistent infection. In this study we evaluated the ability of the exopolysaccharide (EPS) B3-15, produced by the marine Bacillus licheniformis B3-15, to prevent the adhesion and biofilm formation of Pseudomonas aeruginosa ATCC 27853 and Staphylococcus aureus ATCC 29213 on polystyrene and polyvinyl chloride surfaces. The EPS was added at different times (0, 2, 4 and 8 h), corresponding to the initial, reversible and irreversible attachment, and after the biofilm development (24 or 48 h). The EPS (300 µg/mL) impaired the initial phase, preventing bacterial adhesion even when added after 2 h of incubation, but had no effects on mature biofilms. Without exerting any antibiotic activity, the antibiofilm mechanisms of the EPS were related to the modification of the (i) abiotic surface properties, (ii) cell-surface charges and hydrophobicity, and iii) cell-to-cell aggregation. The addition of EPS downregulated the expression of genes (lecA and pslA of P. aeruginosa and clfA of S. aureus) involved in the bacterial adhesion. Moreover, the EPS reduced the adhesion of P. aeruginosa (five logs-scale) and S. aureus (one log) on human nasal epithelial cells. The EPS could represent a promising tool for the prevention of biofilm-related infections.
Collapse
Affiliation(s)
- Vincenzo Zammuto
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
- Research Centre for Extreme Environments and Extremophiles, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
| | - Antonio Spanò
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
- Research Centre for Extreme Environments and Extremophiles, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
| | - Eleonora Agostino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
| | - Angela Macrì
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
| | - Claudia De Pasquale
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Via Consolare Valeria, 1, 98124 Messina, Italy
| | - Guido Ferlazzo
- Department of Experimental Medicine (DIMES), University of Genoa and IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Maria Giovanna Rizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
- Research Centre for Extreme Environments and Extremophiles, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
| | - Marco Sebastiano Nicolò
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
- Research Centre for Extreme Environments and Extremophiles, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
| | - Salvatore Guglielmino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
- Research Centre for Extreme Environments and Extremophiles, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
| | - Concetta Gugliandolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
- Research Centre for Extreme Environments and Extremophiles, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
40
|
Islam OK, Islam I, Saha O, Rahaman MM, Sultana M, Bockmühl DP, Hossain MA. Genomic variability correlates with biofilm phenotypes in multidrug resistant clinical isolates of Pseudomonas aeruginosa. Sci Rep 2023; 13:7867. [PMID: 37188866 DOI: 10.1038/s41598-023-35056-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 05/11/2023] [Indexed: 05/17/2023] Open
Abstract
The multifactorial nature of Pseudomonas aeruginosa biofilm development and genomic variabilities implicates its resistance to conventional antimicrobials and virulence. Therefore, genetic determinants need to be extensively studied to block the early steps of biofilm or already formed biofilms. In this study, a total of 20 multidrug resistant (MDR) clinical P. aeruginosa isolates were evaluated for their biofilm forming abilities and related genes. Of the isolates tested, all of them showed surface attachment tendencies in nutrient limiting conditions, and classified as strong (SBF = 45%), moderate (MBF = 30%) and weak (WBF = 25%) biofilm formers. Complete genome sequencing of representative strong (DMC-27b), moderate (DMC-20c) and weak biofilm former (DMC-30b) isolates was performed. Analysis of biofilm related genes in the sequenced genomes revealed that, 80 of the 88 biofilm related genes possess 98-100% sequence identity to the reference PAO1 strain. Complete and partial sequence data of LecB proteins from tested isolates indicate that isolates containing PA14-like LecB sequences produced strong biofilms. All of the 7 pel operon protein coding genes in weak biofilm former isolate 30b showed significant nucleotide sequence variation with other tested isolates, and their corresponding proteins are 99% identical with the pel operon proteins of PA7. Bioinformatics analyses identified divergent sequence and structural features that separate PA7 like pel operon proteins from reference PAO1-like pel operon. Congo red and pellicle forming assays revealed that the sequence and structure variations may have interfered with the Pel production pathway and resulted in impaired Pel production in isolate 30b that has a PA7 like pel operon. Expression analysis also showed that both pelB and lecB genes were about 5 to 6 folds upregulated after 24 h in SBF 27b in comparison with WBF 30b. Our findings indicate significant genomic divergence in biofilm related genes of P. aeruginosa strains that affect their biofilm phenotypes.
Collapse
Affiliation(s)
- Ovinu Kibria Islam
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
- Department of Microbiology, Jashore University of Science & Technology, Jashore, Bangladesh
| | - Israt Islam
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
- Department of Microbiology, Noakhali University of Science & Technology, Noakhali, Bangladesh
| | - Otun Saha
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
- Department of Microbiology, Noakhali University of Science & Technology, Noakhali, Bangladesh
| | | | - Munawar Sultana
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
| | - Dirk P Bockmühl
- Faculty of Life Science, Rhine-Waal University of Applied Science, Kleve, Germany
| | - M Anwar Hossain
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh.
- Department of Microbiology, Jashore University of Science & Technology, Jashore, Bangladesh.
| |
Collapse
|
41
|
Carneiro J, Magalhães RP, de la Oliva Roque VM, Simões M, Pratas D, Sousa SF. TargIDe: a machine-learning workflow for target identification of molecules with antibiofilm activity against Pseudomonas aeruginosa. J Comput Aided Mol Des 2023; 37:265-278. [PMID: 37085636 DOI: 10.1007/s10822-023-00505-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/12/2023] [Indexed: 04/23/2023]
Abstract
Bacterial biofilms are a source of infectious human diseases and are heavily linked to antibiotic resistance. Pseudomonas aeruginosa is a multidrug-resistant bacterium widely present and implicated in several hospital-acquired infections. Over the last years, the development of new drugs able to inhibit Pseudomonas aeruginosa by interfering with its ability to form biofilms has become a promising strategy in drug discovery. Identifying molecules able to interfere with biofilm formation is difficult, but further developing these molecules by rationally improving their activity is particularly challenging, as it requires knowledge of the specific protein target that is inhibited. This work describes the development of a machine learning multitechnique consensus workflow to predict the protein targets of molecules with confirmed inhibitory activity against biofilm formation by Pseudomonas aeruginosa. It uses a specialized database containing all the known targets implicated in biofilm formation by Pseudomonas aeruginosa. The experimentally confirmed inhibitors available on ChEMBL, together with chemical descriptors, were used as the input features for a combination of nine different classification models, yielding a consensus method to predict the most likely target of a ligand. The implemented algorithm is freely available at https://github.com/BioSIM-Research-Group/TargIDe under licence GNU General Public Licence (GPL) version 3 and can easily be improved as more data become available.
Collapse
Affiliation(s)
- João Carneiro
- Interdisciplinary Centre of Marine and Environmental Research, CIIMAR, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos, s/n, Porto, 4450-208, Portugal.
| | - Rita P Magalhães
- Faculty of Medicine, Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4200-319, Porto, Portugal
- Department of Biomedicine, Faculty of Medicine, UCIBIO-Applied Molecular Biosciences Unit, University of Porto, BioSIM, Porto, 4200-319, Portugal
| | - Victor M de la Oliva Roque
- Faculty of Medicine, Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4200-319, Porto, Portugal
- Department of Biomedicine, Faculty of Medicine, UCIBIO-Applied Molecular Biosciences Unit, University of Porto, BioSIM, Porto, 4200-319, Portugal
| | - Manuel Simões
- Faculty of Engineering, LEPABE Laboratory for Process Engineering, Environment, Biotechnology and Energy, University of Porto, Rua Dr. Roberto Frias, s/n, Porto, 4200-465, Portugal
- Faculty of Engineering, ALiCE-Associate Laboratory in Chemical Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
| | - Diogo Pratas
- Institute of Electronics and Informatics Engineering of Aveiro, IEETA, University of Aveiro, Aveiro, Portugal
- Department of Electronics, Telecommunications and Informatics, DETI, University of Aveiro, Aveiro, Portugal
- Department of Virology, DoV, University of Helsinki, Helsinki, Finland
| | - Sérgio F Sousa
- Faculty of Medicine, Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4200-319, Porto, Portugal
- Department of Biomedicine, Faculty of Medicine, UCIBIO-Applied Molecular Biosciences Unit, University of Porto, BioSIM, Porto, 4200-319, Portugal
| |
Collapse
|
42
|
Wang T, Shen P, He Y, Zhang Y, Liu J. Spatial transcriptome uncovers rich coordination of metabolism in E. coli K12 biofilm. Nat Chem Biol 2023:10.1038/s41589-023-01282-w. [PMID: 37055614 DOI: 10.1038/s41589-023-01282-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 02/02/2023] [Indexed: 04/15/2023]
Abstract
Microbial communities often display region-specific properties, which give rise to complex interactions and emergent behaviors that are critical to the homeostasis and stress response of the communities. However, systems-level understanding of these properties still remains elusive. In this study, we established RAINBOW-seq and profiled the transcriptome of Escherichia coli biofilm communities with high spatial resolution and high gene coverage. We uncovered three modes of community-level coordination, including cross-regional resource allocation, local cycling and feedback signaling, which were mediated by strengthened transmembrane transport and spatially specific activation of metabolism. As a consequence of such coordination, the nutrient-limited region of the community maintained an unexpectedly high level of metabolism, enabling it to express many signaling genes and functionally unknown genes with potential sociality functions. Our work provides an extended understanding of the metabolic interplay in biofilms and presents a new approach of investigating complex interactions in bacterial communities on the systems level.
Collapse
Affiliation(s)
- Tianmin Wang
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Ping Shen
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Yihui He
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Yuzhen Zhang
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Jintao Liu
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| |
Collapse
|
43
|
Zhang Y, Cai Y, Chen Z. Community-specific diffusion characteristics determine resistance of biofilms to oxidative stress. SCIENCE ADVANCES 2023; 9:eade2610. [PMID: 36961890 DOI: 10.1126/sciadv.ade2610] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 02/17/2023] [Indexed: 06/18/2023]
Abstract
Biofilms are multicellular communities with a spatial structure. Different from single-cell scale diffusion in planktonic systems, the diffusion distance becomes the dimension of multicellular clusters in a biofilm. Such differences in diffusion behavior affect the tolerance and response to exogenous stress. Here, we found that at the same doses of exogenous hydrogen peroxide (H2O2), planktonic Escherichia coli were completely killed within two hours, whereas the biofilm resumed growth in six hours by building a catalase barrier to block H2O2 penetration, despite the growth burden. Unexpectedly, when we changed the carbon source from glucose to glycerol, H2O2 instantly counterintuitively boosted biofilm growth due to supplemental oxygen, which was the growth-limiting factor. We further demonstrated that the energy metabolism modes determined the growth-limiting factor, which then determined the two patterns of biofilms resistances to H2O2.
Collapse
Affiliation(s)
- Yuzhen Zhang
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Yumin Cai
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Zhaoyuan Chen
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo 315020, China
| |
Collapse
|
44
|
Schuster M, Li C, Smith P, Kuttler C. Parameters, architecture and emergent properties of the Pseudomonas aeruginosa LasI/LasR quorum-sensing circuit. J R Soc Interface 2023; 20:20220825. [PMID: 36919437 PMCID: PMC10015328 DOI: 10.1098/rsif.2022.0825] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/22/2023] [Indexed: 03/16/2023] Open
Abstract
Quorum sensing is a widespread process in bacteria that controls collective behaviours in response to cell density. Populations of cells coordinate gene expression through the perception of self-produced chemical signals. Although this process is well-characterized genetically and biochemically, quantitative information about network properties, including induction dynamics and steady-state behaviour, is scarce. Here we integrate experiments with mathematical modelling to quantitatively analyse the LasI/LasR quorum sensing pathway in the opportunistic pathogen Pseudomonas aeruginosa. We determine key kinetic parameters of the pathway and, using the parametrized model, show that quorum sensing behaves as a bistable hysteretic switch, with stable on and off states. We investigate the significance of feedback architecture and find that positive feedback on signal production is critical for induction dynamics and bistability, whereas positive feedback on receptor expression and negative feedback on signal production play a minor role. Taken together, our data-based modelling approach reveals fundamental and emergent properties of a bacterial quorum sensing circuit, and provides evidence that native quorum sensing can indeed function as the gene expression switch it is commonly perceived to be.
Collapse
Affiliation(s)
- Martin Schuster
- Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA
| | - Christina Li
- Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA
| | - Parker Smith
- Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA
| | - Christina Kuttler
- Department of Mathematics, Technische Universität München, 85748 Garching, Germany
| |
Collapse
|
45
|
Wang L, Wang H, Zhang H, Wu H. Formation of a biofilm matrix network shapes polymicrobial interactions. THE ISME JOURNAL 2023; 17:467-477. [PMID: 36639539 PMCID: PMC9938193 DOI: 10.1038/s41396-023-01362-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
Staphylococcus aureus colonizes the same ecological niche as many commensals. However, little is known about how such commensals modulate staphylococcal fitness and persistence. Here we report a new mechanism that mediates dynamic interactions between a commensal streptococcus and S. aureus. Commensal Streptococcus parasanguinis significantly increased the staphylococcal biofilm formation in vitro and enhanced its colonization in vivo. A streptococcal biofilm-associated protein BapA1, not fimbriae-associated protein Fap1, is essential for dual-species biofilm formation. On the other side, three staphylococcal virulence determinants responsible for the BapA1-dependent dual-species biofilm formation were identified by screening a staphylococcal transposon mutant library. The corresponding staphylococcal mutants lacked binding to recombinant BapA1 (rBapA1) due to lower amounts of eDNA in their culture supernatants and were defective in biofilm formation with streptococcus. The rBapA1 selectively colocalized with eDNA within the dual-species biofilm and bound to eDNA in vitro, highlighting the contributions of the biofilm matrix formed between streptococcal BapA1 and staphylococcal eDNA to dual-species biofilm formation. These findings have revealed an additional new mechanism through which an interspecies biofilm matrix network mediates polymicrobial interactions.
Collapse
Affiliation(s)
- Lijun Wang
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA
- Department of Laboratory Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, 102218, Beijing, China
| | - Hongxia Wang
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA
| | - Hua Zhang
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA
- Department of Integrative Biomedical and Diagnostic Sciences, Oregon Health and Science University School of Dentistry, Portland, OR, 97239, USA
| | - Hui Wu
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA.
- Department of Integrative Biomedical and Diagnostic Sciences, Oregon Health and Science University School of Dentistry, Portland, OR, 97239, USA.
| |
Collapse
|
46
|
Gerling-Driessen UIM, Hoffmann M, Schmidt S, Snyder NL, Hartmann L. Glycopolymers against pathogen infection. Chem Soc Rev 2023; 52:2617-2642. [PMID: 36820794 DOI: 10.1039/d2cs00912a] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Pathogens including viruses, bacteria, fungi, and parasites continue to shape our lives in profound ways every day. As we have learned to live in parallel with pathogens, we have gained a better understanding of the rules of engagement for how they bind, adhere, and invade host cells. One such mechanism involves the exploitation of host cell surface glycans for attachment/adhesion, one of the first steps of infection. This knowledge has led to the development of glycan-based diagnostics and therapeutics for the treatment and prevention of infection. One class of compounds that has become increasingly important are the glycopolymers. Glycopolymers are macromolecules composed of a synthetic scaffold presenting carbohydrates as side chain motifs. Glycopolymers are particularly attractive because their properties can be tuned by careful choice of the scaffold, carbohydrate/glycan, and overall presentation. In this review, we highlight studies over the past ten years that have examined the role of glycopolymers in pathogen adhesion and host cell infection, biofilm formation and removal, and drug delivery with the aim of examining the direct effects of these macromolecules on pathogen engagement. In addition, we also examine the role of glycopolymers as diagnostics for the detection and monitoring of pathogens.
Collapse
Affiliation(s)
- Ulla I M Gerling-Driessen
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| | - Miriam Hoffmann
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| | - Stephan Schmidt
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany. .,Institute for Macromolecular Chemistry, University of Freiburg, Stefan-Meier-Str. 31, 79104 Freiburg, Germany
| | - Nicole L Snyder
- Department of Chemistry, Davidson College, Davidson, North Carolina 28035, USA
| | - Laura Hartmann
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| |
Collapse
|
47
|
Arif M, Ahmad R, Sharaf M, Muhammad J, Abdalla M, Eltayb WA, Liu CG. Antibacterial and antibiofilm activity of mannose-modified chitosan/PMLA nanoparticles against multidrug-resistant Helicobacter pylori. Int J Biol Macromol 2022; 223:418-432. [PMID: 36356866 DOI: 10.1016/j.ijbiomac.2022.10.265] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/26/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
Abstract
Because of the apparent stasis in antibiotic discoveries and the growth of multidrug resistance, Helicobacter pylori-associated gastric infections are difficult to eradicate. In the search for alternative therapy, the reductive amination of chitosan with mannose, followed by ionic gelation, produced mannose functionalized chitosan nanoparticles. Then, molecular docking and molecular dynamics (MD) simulations were conducted with H. pylori lectin (HPLectin) as a target protein involved in bacterium adherence to host cells, biofilm formation, and cytotoxicity. Changes in zeta potential and FTIR spectroscopy revealed that chitosan was functionalized with mannose. Time-kill, polystyrene adherence, and antibiofilm studies were utilized to assess nanoparticles as an alternative antibacterial treatment against a resistant gastric pathogen. Man-CS-Nps were discovered to have effective anti-adherence and biofilm disruption characteristics in suppressing the development of resistant H. pylori. In addition, bioimaging studies with CLSM, TEM, and SEM illustrated that Man-CS-Nps interacted with bacterial cells and induced membrane disruption by creating holes in the outer membranes of the bacterial cells, resulting in the leakage of amino acids. Importantly, molecular docking and 20 ns MD simulations revealed that Man-CS-Nps inhibited the target protein through slow-binding inhibition and hydrogen bond interactions with active site residues. As a consequence of the findings of this study, the Man-CS-Nps is an excellent candidate for developing alternative therapies for the increasing incidences of resistant gastric infections.
Collapse
Affiliation(s)
- Muhammad Arif
- College of Marine Life Science, Ocean University of China, No.5 Yushan Road, Qingdao 266003, PR China
| | - Rafiq Ahmad
- Department of Microbiology, The University of Haripur, Haripur 22610, Pakistan
| | - Mohamed Sharaf
- Department of Biochemistry, Faculty of Agriculture, Al-Azhar University, Nasr City, Cairo 11751, Egypt
| | - Javed Muhammad
- Department of Microbiology, The University of Haripur, Haripur 22610, Pakistan
| | - Mohnad Abdalla
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Shandong Province, 250012, PR China.
| | - Wafa Ali Eltayb
- Biotechnology Department, Faculty of Science and Technology, Shendi University, Shendi, Nher Anile, Sudan
| | - Chen-Guang Liu
- College of Marine Life Science, Ocean University of China, No.5 Yushan Road, Qingdao 266003, PR China.
| |
Collapse
|
48
|
Tripathi S, Purchase D, Govarthanan M, Chandra R, Yadav S. Regulatory and innovative mechanisms of bacterial quorum sensing-mediated pathogenicity: a review. ENVIRONMENTAL MONITORING AND ASSESSMENT 2022; 195:75. [PMID: 36334179 DOI: 10.1007/s10661-022-10564-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/29/2022] [Indexed: 06/16/2023]
Abstract
Quorum sensing (QS) is a system of bacteria in which cells communicate with each other; it is linked to cell density in the microbiome. The high-density colony population can provide enough small molecular signals to enable a range of cellular activities, gene expression, pathogenicity, and antibiotic resistance that cause damage to the hosts. QS is the basis of chronic illnesses in human due to microbial sporulation, expression of virulence factors, biofilm formation, secretion of enzymes, or production of membrane vesicles. The transfer of antimicrobial resistance gene (ARG) among antibiotic resistance bacteria is a major public health concern. QS-mediated biofilm is a hub for ARG horizontal gene transfer. To develop innovative approach to prevent microbial pathogenesis, it is essential to understand the role of QS especially in response to environmental stressors such as exposure to antibiotics. This review provides the latest knowledge on the relationship of QS and pathogenicity and explore the novel approach to control QS via quorum quenching (QQ) using QS inhibitors (QSIs) and QQ enzymes. The state-of-the art knowledge on the role of QS and the potential of using QQ will help to overcome the threats of rapidly emerging bacterial pathogenesis.
Collapse
Affiliation(s)
- Sonam Tripathi
- Department of Environmental Microbiology, School for Environmental Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow, 226025, UP, India
| | - Diane Purchase
- Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, The Burroughs, Hendon, London, NW4 4BT, UK
| | - Muthusamy Govarthanan
- Department of Environmental Engineering, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 41566, South Korea
| | - Ram Chandra
- Department of Environmental Microbiology, School for Environmental Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow, 226025, UP, India.
| | - Sangeeta Yadav
- Department of Environmental Microbiology, School for Environmental Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow, 226025, UP, India.
- Department of Botany, Vaishno Devi Prashikshan Mahavidyalaya, Gondahi, Kunda, Pratapgarh, India.
| |
Collapse
|
49
|
Rath S, Palit K, Das S. Variable pH and subsequent change in pCO 2 modulates the biofilm formation, synthesis of extracellular polymeric substances, and survivability of a marine bacterium Bacillus stercoris GST-03. ENVIRONMENTAL RESEARCH 2022; 214:114128. [PMID: 36007573 DOI: 10.1016/j.envres.2022.114128] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/03/2022] [Accepted: 08/14/2022] [Indexed: 06/15/2023]
Abstract
Biofilm-forming bacteria adhere to the substrates and engage in the nutrient cycling process. However, environmental conditions may interrupt the biofilm formation ability, which ultimately may affect various biogeochemical cycles. The present study reports the effect of varying pH and subsequent change in pCO2 on the survivability, biofilm formation, and synthesis of extracellular polymeric substances (EPS) of a biofilm-forming marine bacterium Bacillus stercoris GST-03 isolated from the Bhitarkanika mangrove ecosystem, Odisha, India. Understanding the pH-dependent alteration in EPS constituents, and associated functional groups of a marine bacterium will provide better insight into the adaptability of the bacteria in future ocean acidification scenarios. The strain was found to tolerate and form biofilm up to pH 4, with the maximum biofilm formation at pH 6. EPS yield and the synthesis of the key components of the EPS, including carbohydrate, protein, and lipid, were found maximum at pH 6. Changes in biofilm formation patterns and various topological parameters at varying pH/pCO2 conditions were observed. A cellular chaining pattern was observed at pH 4, and maximum biofilm formation was obtained at pH 6 with biomass of 5.28582 ± 0.5372 μm3/μm2 and thickness of 9.982 ± 1.5288 μm. Structural characterization of EPS showed changes in various functional groups of constituent macromolecules with varying pH. The amorphous nature of the EPS and the changes in linkages and associated functional groups (-R2CHOR, -CH3, and -CH2) with pH variation was confirmed. EPS showed a two-step degradation with a maximum weight loss of 59.147% and thermal stability up to 480 °C at pH 6. The present work efficiently demonstrates the role of EPS in providing structural and functional stability to the biofilm in varying pH conditions. The findings will provide a better understanding of the adaptability of marine bacteria in the future effect of ocean acidification.
Collapse
Affiliation(s)
- Sonalin Rath
- Laboratory of Environmental Microbiology and Ecology (LEnME), Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Krishna Palit
- Laboratory of Environmental Microbiology and Ecology (LEnME), Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Surajit Das
- Laboratory of Environmental Microbiology and Ecology (LEnME), Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| |
Collapse
|
50
|
Mała P, Siebs E, Meiers J, Rox K, Varrot A, Imberty A, Titz A. Discovery of N-β-l-Fucosyl Amides as High-Affinity Ligands for the Pseudomonas aeruginosa Lectin LecB. J Med Chem 2022; 65:14180-14200. [PMID: 36256875 PMCID: PMC9620277 DOI: 10.1021/acs.jmedchem.2c01373] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Indexed: 11/30/2022]
Abstract
The Gram-negative pathogen Pseudomonas aeruginosa causes severe infections mainly in immunocompromised or cystic fibrosis patients and is able to resist antimicrobial treatments. The extracellular lectin LecB plays a key role in bacterial adhesion to the host and biofilm formation. For the inhibition of LecB, we designed and synthesized a set of fucosyl amides, sulfonamides, and thiourea derivatives. Then, we analyzed their binding to LecB in competitive and direct binding assays. We identified β-fucosyl amides as unprecedented high-affinity ligands in the two-digit nanomolar range. X-ray crystallography of one α- and one β-anomer of N-fucosyl amides in complex with LecB revealed the interactions responsible for the high affinity of the β-anomer at atomic level. Further, the molecules showed good stability in murine and human blood plasma and hepatic metabolism, providing a basis for future development into antibacterial drugs.
Collapse
Affiliation(s)
- Patrycja Mała
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical
Research Saarland (HIPS), Helmholtz Centre
for Infection Research, 66123Saarbrücken, Germany
- Faculty
of Chemistry, Adam Mickiewicz University, 61-614Poznań, Poland
| | - Eike Siebs
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical
Research Saarland (HIPS), Helmholtz Centre
for Infection Research, 66123Saarbrücken, Germany
- Department
of Chemistry, Saarland University, 66123Saarbrücken, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, 38124Braunschweig, Germany
| | - Joscha Meiers
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical
Research Saarland (HIPS), Helmholtz Centre
for Infection Research, 66123Saarbrücken, Germany
- Department
of Chemistry, Saarland University, 66123Saarbrücken, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, 38124Braunschweig, Germany
| | - Katharina Rox
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, 38124Braunschweig, Germany
- Chemical
Biology (CBIO), Helmholtz Centre for Infection
Research, 38124Braunschweig, Germany
| | | | - Anne Imberty
- Univ.
Grenoble
Alpes, CNRS, CERMAV, 38000Grenoble, France
| | - Alexander Titz
- Chemical
Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical
Research Saarland (HIPS), Helmholtz Centre
for Infection Research, 66123Saarbrücken, Germany
- Department
of Chemistry, Saarland University, 66123Saarbrücken, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, 38124Braunschweig, Germany
| |
Collapse
|