1
|
Manna T, Dey S, Karmakar M, Panda AK, Ghosh C. Investigations on genomic, topological and structural properties of diguanylate cyclases involved in Vibrio cholerae biofilm signalling using in silico techniques: Promising drug targets in combating cholera. Curr Res Struct Biol 2025; 9:100166. [PMID: 40330072 PMCID: PMC12051071 DOI: 10.1016/j.crstbi.2025.100166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/19/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
During various stages of its life cycle, Vibrio cholerae initiate biofilm signalling cascade. Intercellular high level of the signalling nucleotide 3'-5' cyclic dimeric guanosine monophosphate (c-di-GMP), synthesized by diguanylate cyclases (DGCs) from its precursor molecule GTP, is crucial for biofilm formation. Present study endeavours to in silico approaches in evaluating genomic, physicochemical, topological and functional properties of six c-di-GMP regulatory DGCs (CdgA, CdgH, CdgK, CdgL, CdgM, VpvC) of V. cholerae. Genomic investigations unveiled that codon preferences were inclined towards AU ending over GC ending codons and overall GC content ranged from 44.6 to 49.5 with codon adaptation index ranging from 0.707 to 0.783. Topological analyses deciphered the presence of transmembrane domains in all proteins. All the DGCs were acidic, hydrophilic and thermostable. Only CdgA, CdgH and VpvC were predicted to be stable during in vitro conditions. Non-polar amino acids with leucine being the most abundant amino acid among these DGCs with α-helix as the predominant secondary structure, responsible for forming the transmembrane regions by secondary structure analysis. Tertiary structures of the proteins were obtained by computation using AlphaFold and trRosetta. Predicted structures by both the servers were compared in various aspects using PROCHECK, ERRAT and Modfold8 servers. Selected 3D structures were refined using GalaxyRefine. InterPro Scan revealed presence of a conserved GGDEF domain in all DGCs and predicted the active site residues in the GGDEF domain. Molecular docking studies using CB-DOCK 2 tool revealed that among the DGCs, VpvC exhibited highest affinity for GTP (-5.6 kcal/mol), which was closely followed by CdgL (-5.5 kcal/mol). MD simulations depicted all DGC-GTP complexes to be stable due to its considerably low eigenvalues. Such studies are considered to provide maiden insights into the genomic and structural properties of V. cholerae DGCs, actively involved in biofilm signalling systems, and it is projected to be beneficial in the discovery of novel DGC inhibitors that can target and downregulate the c-di-GMP regulatory system to develop anti-biofilm strategies against the cholera pathogen.
Collapse
Affiliation(s)
- Tuhin Manna
- Deparment of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Subhamoy Dey
- Deparment of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
- Centre for Life Sciences, Vidyasagar University, Midnapore, West Bengal, India
| | - Monalisha Karmakar
- Deparment of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Amiya Kumar Panda
- Department of Chemistry, Vidyasagar University, Midnapore, West Bengal, India
- Rani Rashmoni Green University, Singur, West Bengal, India
| | - Chandradipa Ghosh
- Deparment of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| |
Collapse
|
2
|
Oladosu VI, Sauer K. FleQ finetunes the expression of a subset of BrlR-activated genes to enable antibiotic tolerance by Pseudomonas aeruginosa biofilms. J Bacteriol 2025; 207:e0050324. [PMID: 40304498 DOI: 10.1128/jb.00503-24] [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: 11/26/2024] [Accepted: 04/03/2025] [Indexed: 05/02/2025] Open
Abstract
The transcriptional regulator FleQ contributes to Pseudomonas aeruginosa biofilm formation by activating the expression and biosynthesis of matrix exopolysaccharides in a manner dependent on c-di-GMP. However, little is known about the role of FleQ in the antibiotic tolerance phenotype of P. aeruginosa biofilms. Inactivation of fleQ impaired biofilm formation and rendered biofilms susceptible to tobramycin and norfloxacin. The phenotypes were similar to biofilms inactivated in sagS encoding the orphan sensor SagS that promotes the switch from planktonic to biofilm growth via BfiSR and antibiotic tolerance via BrlR. While FleQ was found to contribute to biofilm formation independently of SagS and BfiSR, FleQ instead converged with SagS-dependent regulation at the level of BrlR. This was supported by multicopy expression of sagS failing to restore biofilm antibiotic tolerance by ΔfleQ to wild-type levels (and vice versa) and by biofilms formed by the ΔfleQΔsagS double mutant being as susceptible as ΔfleQ and ΔsagS biofilms. Increased antibiotic susceptibility was independent of BrlR abundance or BrlR DNA binding but coincided with significantly reduced transcript abundance of the BrlR-activated mexCD-oprJ and PA1874-77, encoding an ABC transporter previously shown to contribute to the tolerance of biofilms to tobramycin and norfloxacin. FleQ- dependent regulation of gene expression was indirect. Co-immunoprecipitation and BACTH assays indicated FleQ to interact with SagS via its HisKA-Rec domain, likely suggesting FleQ and SagS to likely work in concert to enable biofilm antibiotic tolerance, by finetuning the expression of BrlR activated genes.IMPORTANCEIn P. aeruginosa, FleQ inversely regulates the expression of genes encoding flagella and biofilm matrix components, including exopolysaccharide (Pel, Psl) in a manner dependent on the levels of c-di-GMP. Our findings expand on the role of FleQ from regulating the transition to the biofilm mode of growth to FleQ contributing to the antimicrobial tolerance phenotype of biofilms, by FleQ affecting the expression of PA1874-77, a downstream target of the SagS-dependent transcriptional regulator BrlR. Importantly, our findings suggest FleQ works in concert with SagS, likely via FleQ-SagS protein-protein interactions, to enable the formation of inherently tolerant P. aeruginosa biofilms.
Collapse
Affiliation(s)
- Victoria I Oladosu
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | - Karin Sauer
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
3
|
Sun R, Gangan MS, Wang Q, Boedicker JQ, Armani AM. Magnetically Tunable Hydrogel for Biofilm Control. ACS APPLIED BIO MATERIALS 2025. [PMID: 40383927 DOI: 10.1021/acsabm.5c00409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Bacterial biofilm formation contributes to healthcare and energy challenges, and researchers are actively pursuing a range of strategies to restrict the spread of biofilms in an eco-friendly manner. Commonly used approaches in industry rely on physical removal and chemical techniques, frequently targeting mature biofilms. While effective, these methods often face implementation challenges in remote settings and can have off-target environmental impacts. As a result, an alternative strategy is to focus on controlling or limiting the biofilm formation and growth rates with remote stimuli. It has been shown that the mechanotransduction pathway intrinsic to bacteria responds to changes in the storage modulus of the growth surface, modifying the bacteria's motility and biofilm formation. We developed a material with magnetically tunable mechanical properties by intercalating magnetic nanoparticles into an agar gel matrix and investigated its ability to control Escherichia coli motility and biofilm growth. The initial storage modulus ranges from 0.5 to 2.5 kPa, depending on the material composition. Upon exposure to a 20 mT magnetic field using standard neodymium magnets, the modulus is dynamically and reversibly increased by approximately 30%. As a result of this increase, the expansion rate of the E. coli biofilm is reduced by approximately 40%. The simplicity of the manipulation of its mechanical property not only gives this biomaterial potential to further mechanosensing mechanism research but also proves to be an innovative strategy for remote and eco-conscious restriction of biofilm formation.
Collapse
Affiliation(s)
- Ruojiao Sun
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States
| | - Manasi S Gangan
- Department of Physics and Astronomy, University of Southern California, Los Angeles, California 90089, United States
| | - Qiming Wang
- Sonny Astani Department of Civil and Environmental Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - James Q Boedicker
- Department of Physics and Astronomy, University of Southern California, Los Angeles, California 90089, United States
| | - Andrea M Armani
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States
- Ellison Medical Institute, Los Angeles, California 90064-1016, United States
| |
Collapse
|
4
|
Nolan LM, Webber MA, Filloux A. Throwing a spotlight on genomic dark matter: the power and potential of transposon-insertion sequencing. J Biol Chem 2025:110231. [PMID: 40378959 DOI: 10.1016/j.jbc.2025.110231] [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: 03/11/2025] [Revised: 04/29/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025] Open
Abstract
Linking genotype to phenotype is a central goal in biology. In the microbiological field, transposon mutagenesis is a technique that has been widely used since the 1970's to facilitate this connection. The development of modern 'omics approaches and next-generation sequencing, have allowed high-throughput association between genes and their putative function. In 2009, four different variations of modern transposon-insertion sequencing (TIS) approaches were published, being referred to as transposon-directed insertion-site sequencing (TraDIS), transposon sequencing (Tn-seq), insertion sequencing (INSeq) and high-throughput insertion tracking by deep sequencing (HITS). These approaches exploit a similar concept to allow estimation of the essentiality or contribution to fitness of each gene in any bacterial genome. The main rationale is to perform a comparative analysis of the abundance of specific transposon mutants under one or more selective conditions. The approaches themselves only vary in the transposon used for mutagenesis, and in the methodology used for sequencing library preparation. In this review, we discuss how TIS approaches have been used to facilitate a major shift in our fundamental understanding of bacterial biology in a range of areas. We focus on several aspects including pathogenesis, biofilm development, polymicrobial interactions in various ecosystems, and antimicrobial resistance. These studies have provided new insight into bacterial physiology and revealed predicted functions for hundreds of genes previously representing genomic 'dark matter'. We also discuss how TIS approaches have been used to understand complex bacterial systems and interactions and how future developments of TIS could continue to accelerate and enrich our understanding of bacterial biology.
Collapse
Affiliation(s)
- Laura M Nolan
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore; Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
| | - Mark A Webber
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK; Norwich Medical School, Norwich Research Park, Norwich, UK; Center for Microbial Interactions, Norwich Research Park, Norwich, UK
| | - Alain Filloux
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore; Lee Kon Chian School of Medicine, Nanyang Technological University, Singapore; Imperial, Centre for Bacterial Resistance Biology, London, UK
| |
Collapse
|
5
|
Ben-David Y, Sporny M, Brochin Y, Piscon B, Roth S, Zander I, Nisani M, Shoshani S, Yaron O, Karako-Lampert S, Lebenthal-Loinger I, Danielli A, Opatowsky Y, Banin E. SadB, a mediator of AmrZ proteolysis and biofilm development in Pseudomonas aeruginosa. NPJ Biofilms Microbiomes 2025; 11:77. [PMID: 40360526 PMCID: PMC12075610 DOI: 10.1038/s41522-025-00710-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
The ability of bacteria to commit to surface colonization and biofilm formation is a highly regulated process. In this study, we characterized the activity and structure of SadB, initially identified as a key regulator in the transition from reversible to irreversible surface attachment. Our results show that SadB acts as an adaptor protein that tightly regulates the master regulator AmrZ at the post-translational level. SadB directly binds to the C-terminal domain of AmrZ, leading to its rapid degradation, primarily by the Lon protease. Structural analysis suggests that SadB does not directly interact with small molecules upon signal transduction, differing from previous findings in Pseudomonas fluorescens. Instead, the SadB structure supports its role in mediating protein-protein interactions, establishing it as a major checkpoint for biofilm commitment.
Collapse
Affiliation(s)
- Yossi Ben-David
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Michael Sporny
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Yigal Brochin
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Bar Piscon
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Shira Roth
- The Alexander Kofkin Faculty of Engineering, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Itzhak Zander
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Michal Nisani
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Sivan Shoshani
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Orly Yaron
- The Scientific Equipment Center, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Sarit Karako-Lampert
- The Scientific Equipment Center, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Ilana Lebenthal-Loinger
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Amos Danielli
- The Alexander Kofkin Faculty of Engineering, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Yarden Opatowsky
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel
| | - Ehud Banin
- The Mina & Everard Goodman Faculty of Life Sciences, The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb Street, Ramat Gan, 5290002, Israel.
| |
Collapse
|
6
|
Ramesh R, Rekha ND, Gopal S. Pseudomonas aeruginosa biofilm: treatment strategies to combat infection. Arch Microbiol 2025; 207:141. [PMID: 40348909 DOI: 10.1007/s00203-025-04346-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/25/2025] [Accepted: 04/26/2025] [Indexed: 05/14/2025]
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogenic bacterium that is a common cause of both acute and chronic infections. Multidrug-resistant P. aeruginosa poses a significant challenge to antibiotics and therapeutic approaches due to its pathogenicity, virulence, and biofilm-forming ability mediated by quorum sensing. Understanding the pathogenic mechanisms is essential for developing potential drug targets. In this regard, strategies aimed at combating the targeted inhibition of virulence, quorum sensing pathways, secretion systems, biofilm-associated two-component systems, and signalling system regulators (such as c-di-GMP) associated with biofilm formation are critical. Several new antimicrobial agents have been developed using these strategies, including antimicrobial peptides, bacteriophages, nanoantibiotics, photodynamics, and natural products, which are considered promising therapeutic tools. In this review, we address the concept of biofilms, their regulation, and recent treatment strategies to target P. aeruginosa, a clinically significant pathogen known for biofilm formation.
Collapse
Affiliation(s)
- Rashmi Ramesh
- Department of Studies in Microbiology, University of Mysore, Manasagangotri, Mysuru, Karnataka, India
| | - N D Rekha
- Department of Biotechnology, JSS College of Arts, Commerce and Science (Autonomous), Mysuru, Karnataka, India
| | - Shubha Gopal
- Department of Studies in Microbiology, University of Mysore, Manasagangotri, Mysuru, Karnataka, India.
| |
Collapse
|
7
|
Yang S, Li D, Fu S, Zheng J, Zhu Y, Li H, Zeng H, Zhang J. Decoding the effect of antibiotics on biofilm formation in biofilters. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 385:125698. [PMID: 40347873 DOI: 10.1016/j.jenvman.2025.125698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/21/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
Biofilms have extensive applications and important roles in biological processes. This study aimed to investigate the effect and mechanism of low-concentration sulfamethoxazole (SMX) on biofilm development in biofilters. The effects of various SMX concentrations (0, 100 ng/L, 1000 ng/L) on microbial development were compared. Compared with the control group without SMX, the start-up period of R2 and R3 filters with SMX added was decreased by 9 % and 21 %, respectively. Under antibiotic stimulation, reactive oxygen species (ROS) and bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP) concentrations increased, aligning with changes in extracellular polymer content and biofilm formation. Microbial community results showed that the presence of SMX promoted the growth of some manganese-oxidizing bacteria (MnOB), such as Massilia, Pedomicrobium, Sphingopyxis, Pseudomonas, and Bacillus. Functional gene analysis further revealed higher expression levels of genes related to c-di-GMP transformation in the presence of SMX. These findings suggest that microbial communities can adapt to their environment by accelerating biofilm formation at lower antibiotic concentrations. The results of this study provide new insights into the impact of low-concentration antibiotics on biofilm development and offer a crucial reference for biofilter design and optimization.
Collapse
Affiliation(s)
- Sen Yang
- Key Laboratory of Water Science and Water Environment Recovery Engineering, Beijing University of Technology, Beijing, 100123, China.
| | - Dong Li
- Key Laboratory of Water Science and Water Environment Recovery Engineering, Beijing University of Technology, Beijing, 100123, China.
| | - Sibo Fu
- Key Laboratory of Water Science and Water Environment Recovery Engineering, Beijing University of Technology, Beijing, 100123, China.
| | - Jifang Zheng
- Key Laboratory of Water Science and Water Environment Recovery Engineering, Beijing University of Technology, Beijing, 100123, China.
| | - Yanjun Zhu
- Key Laboratory of Water Science and Water Environment Recovery Engineering, Beijing University of Technology, Beijing, 100123, China.
| | - Hongpeng Li
- Key Laboratory of Water Science and Water Environment Recovery Engineering, Beijing University of Technology, Beijing, 100123, China.
| | - Huiping Zeng
- Key Laboratory of Water Science and Water Environment Recovery Engineering, Beijing University of Technology, Beijing, 100123, China.
| | - Jie Zhang
- Key Laboratory of Water Science and Water Environment Recovery Engineering, Beijing University of Technology, Beijing, 100123, China; State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150090, China.
| |
Collapse
|
8
|
Lu Z, Zhang J, Li J, Feng L, Wang Y, Zhu J. Biofilm formation of Pseudomonas fluorescens induced by a novel diguanylate cyclase modulated c-di-GMP promotes spoilage of large yellow croaker (Larimichthys crocea). Food Res Int 2025; 208:116231. [PMID: 40263799 DOI: 10.1016/j.foodres.2025.116231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 04/24/2025]
Abstract
Pseudomonas as major agents cause the microbial spoilage in aerobically stored seafoods due to the strong biofilm forming ability, resulting in significant economic losses. C-di-GMP regulates the transition to biofilm states in numerous bacteria, however, its function in biofilm and spoilage of Pseudomonas fluorescens has still been scarce. Here, in a fish spoiler P. fluorescens PF07 strain, 26 proteins of diguanylate cyclase (DGC) containing a GGDEF domain were characterized, and both intracellular c-di-GMP and biofilm formation consistently decreased in the constructed 12 deletion mutants of DGC domain. Compared to wild type (WT) strain, both swimming and swarming in these mutants remarkably enhanced, while the secretion of siderophore, protease activity, and the production of total volatile basic nitrogen (TVB-N) were decreased in several mutants, indicating the different modulating effects among these DGC mutants. Furthermore, correlation analysis of these six phenotypes, PF07_04309 exhibited the most significant alteration, which was identified a novel functional DGC enzyme. Moreover, the GGAAA mutation of PF07_04309 induced the down-regulation of Psl and Alg operons and increased flagellar related gene, resulting in forming the sparser and thinner biofilms. Two mutants of 04309 induced by low c-di-GMP significantly declined the accumulation of TVB-N, thiobarbituric acid, extracellular protease activity and spoilage flavor compounds, especially methylamine and carbon disulfide, in the fillets of large yellow croaker stored at 4 °C. Thus, our results indicated that a novel DGC 04309 modulated the polysaccharide secretion, flagellar, and iron carrier by synthesis of c-di-GMP, positively regulating the spoilage potential of P. fluorescens, which expanded the original insights of DGC and c-di-GMP function on microbial food spoilage.
Collapse
Affiliation(s)
- Zhong Lu
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China
| | - Jun Zhang
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China
| | - Jiashi Li
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China
| | - Lifang Feng
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China
| | - Yanbo Wang
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Junli Zhu
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| |
Collapse
|
9
|
Vennard CS, Oladeji SM, Sintim HO. Inhibitors of Cyclic Dinucleotide Phosphodiesterases and Cyclic Oligonucleotide Ring Nucleases as Potential Drugs for Various Diseases. Cells 2025; 14:663. [PMID: 40358186 PMCID: PMC12072042 DOI: 10.3390/cells14090663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
The phosphodiester linkage is found in DNA, RNA and many signaling molecules, such as cyclic mononucleotide, cyclic dinucleotides (CDNs) and cyclic oligonucleotides (cONs). Enzymes that cleave the phosphodiester linkage (nucleases and phosphodiesterases) play important roles in cell persistence and fitness and have therefore become targets for various diseased states. While various inhibitors have been developed for nucleases and cyclic mononucleotide phosphodiesterases, and some have become clinical successes, there is a paucity of inhibitors of the recently discovered phosphodiesterases or ring nucleases that cleave CDNs and cONs. Inhibitors of bacterial c-di-GMP or c-di-AMP phosphodiesterases have the potential to be used as anti-virulence compounds, while compounds that inhibit the degradation of 3',3'-cGAMP, cA3, cA4, cA6 could serve as antibiotic adjuvants as the accumulation of these second messengers leads to bacterial abortive infection. In humans, 2'3'-cGAMP plays critical roles in antiviral and antitumor responses. ENPP1 (the 2'3'-cGAMP phosphodiesterase) or virally encoded cyclic dinucleotide phosphodiesterases, such as poxin, however, blunt this response. Inhibitors of ENPP1 or poxin-like enzymes have the potential to be used as anticancer and antiviral agents, respectively. This review summarizes efforts made towards the discovery and development of compounds that inhibit CDN phosphodiesterases and cON ring nucleases.
Collapse
Affiliation(s)
- Christopher S. Vennard
- Chemistry Department, Purdue University, West Lafayette, IN 47907, USA; (C.S.V.); (S.M.O.)
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Samson Marvellous Oladeji
- Chemistry Department, Purdue University, West Lafayette, IN 47907, USA; (C.S.V.); (S.M.O.)
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Herman O. Sintim
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
10
|
Czerwinski A, Löwenstrom J, Franzenburg S, Groth EE, Obeng N, Schulenburg H. PelD is required downstream of c-di-GMP for host specialization of Pseudomonas lurida. BMC Microbiol 2025; 25:220. [PMID: 40241006 PMCID: PMC12001729 DOI: 10.1186/s12866-025-03945-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND The bacterial second messenger c-di-GMP is known to influence the formation of biofilms and thereby persistence of pathogenic and beneficial bacteria in hosts. A previous evolution experiment with Pseudomonas lurida MYb11, occasional symbiont of the nematode Caenorhabditis elegans, led to the emergence of host-specialized variants with elevated intracellular c-di-GMP. Thus far, the molecular underpinnings of c-di-GMP-mediated host specialization were unknown in this symbiosis. Therefore, the current study aimed at identifying candidate molecular processes by combining transcriptomic and functional genetic analyses. RESULTS We found that MYb11 host specialists differentially expressed genes related to attachment, motility and biofilm production, including pelD from the pel gene cluster. pelD deletion resulted in reduced intra-host competitive fitness, lower bacterial numbers in C. elegans and loss of biofilm biomass. CONCLUSION Our results identify pelD as a previously unknown key modulator of beneficial symbiont-host associations that acts downstream of c-di-GMP.
Collapse
Affiliation(s)
- Anna Czerwinski
- Department of Evolutionary Ecology and Genetics, University of Kiel, 24118, Kiel, Germany
| | - Julia Löwenstrom
- Department of Evolutionary Ecology and Genetics, University of Kiel, 24118, Kiel, Germany
| | - Sören Franzenburg
- Institute of Clinical Molecular Biology, University of Kiel, 24118, Kiel, Germany
| | - Espen Elias Groth
- Department of Pneumology, LungenClinic Grosshansdorf, 22927, Großhansdorf, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 22927, Großhansdorf, Germany
| | - Nancy Obeng
- Department of Evolutionary Ecology and Genetics, University of Kiel, 24118, Kiel, Germany
- Roche Pharma Research and Early Development, Infectious Disease, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, 4070, Switzerland
| | - Hinrich Schulenburg
- Department of Evolutionary Ecology and Genetics, University of Kiel, 24118, Kiel, Germany.
- Max Planck Institute for Evolutionary Biology, 24306, Plön, Germany.
| |
Collapse
|
11
|
de la Viuda V, Buceta J, Grobas I. Physical communication pathways in bacteria: an extra layer to quorum sensing. Biophys Rev 2025; 17:667-685. [PMID: 40376406 PMCID: PMC12075086 DOI: 10.1007/s12551-025-01290-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 02/13/2025] [Indexed: 05/18/2025] Open
Abstract
Bacterial communication is essential for survival, adaptation, and collective behavior. While chemical signaling, such as quorum sensing, has been extensively studied, physical cues play a significant role in bacterial interactions. This review explores the diverse range of physical stimuli, including mechanical forces, electromagnetic fields, temperature, acoustic vibrations, and light that bacteria may experience with their environment and within a community. By integrating these diverse communication pathways, bacteria can coordinate their activities and adapt to changing environmental conditions. Furthermore, we discuss how these physical stimuli modulate bacterial growth, lifestyle, motility, and biofilm formation. By understanding the underlying mechanisms, we can develop innovative strategies to combat bacterial infections and optimize industrial processes.
Collapse
Affiliation(s)
- Virgilio de la Viuda
- Theoretical and Computational Systems Biology Program, Institute for Integrative Systems Biology (I2sysbio), CSIC-UV, Catedrático Agustín Escardino Benlloch 9, 46980 Paterna, Spain
| | - Javier Buceta
- Theoretical and Computational Systems Biology Program, Institute for Integrative Systems Biology (I2sysbio), CSIC-UV, Catedrático Agustín Escardino Benlloch 9, 46980 Paterna, Spain
| | - Iago Grobas
- Theoretical and Computational Systems Biology Program, Institute for Integrative Systems Biology (I2sysbio), CSIC-UV, Catedrático Agustín Escardino Benlloch 9, 46980 Paterna, Spain
| |
Collapse
|
12
|
Martino RA, Volke DC, Tenaglia AH, Tribelli PM, Nikel PI, Smania AM. Genetic Dissection of Cyclic di-GMP Signalling in Pseudomonas aeruginosa via Systematic Diguanylate Cyclase Disruption. Microb Biotechnol 2025; 18:e70137. [PMID: 40172309 PMCID: PMC11963287 DOI: 10.1111/1751-7915.70137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 04/04/2025] Open
Abstract
The second messenger bis-(3' → 5')-cyclic dimeric guanosine monophosphate (c-di-GMP) governs adaptive responses in the opportunistic pathogen Pseudomonas aeruginosa, including biofilm formation and the transition from acute to chronic infections. Understanding the intricate c-di-GMP signalling network remains challenging due to the overlapping activities of numerous diguanylate cyclases (DGCs). In this study, we employed a CRISPR-based multiplex genome-editing tool to disrupt all 32 GGDEF domain-containing proteins (GCPs) implicated in c-di-GMP signalling in P. aeruginosa PA14. Phenotypic and physiological analyses revealed that the resulting mutant was unable to form biofilms and had attenuated virulence. Residual c-di-GMP levels were still detected despite the extensive GCP disruption, underscoring the robustness of this regulatory network. Taken together, these findings provide insights into the complex c-di-GMP metabolism and showcase the importance of functional overlapping in bacterial signalling. Moreover, our approach overcomes the native redundancy in c-di-GMP synthesis, providing a framework to dissect individual DGC functions and paving the way for targeted strategies to address bacterial adaptation and pathogenesis.
Collapse
Affiliation(s)
- Román A. Martino
- Universidad Nacional de CórdobaFacultad de Ciencias Químicas, Departamento de Química Biológica Ranwel CaputtoCórdobaArgentina
- CONICET, Universidad Nacional de CórdobaCentro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC)CórdobaArgentina
| | - Daniel C. Volke
- The Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkKongens LyngbyDenmark
| | - Albano H. Tenaglia
- Universidad Nacional de CórdobaFacultad de Ciencias Químicas, Departamento de Química Biológica Ranwel CaputtoCórdobaArgentina
- CONICET, Universidad Nacional de CórdobaCentro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC)CórdobaArgentina
| | - Paula M. Tribelli
- Universidad de Buenos AiresFacultad de Ciencias Exactas y Naturales, Departamento de Química BiológicaBuenos AiresArgentina
- CONICET, Universidad de Buenos AiresInstituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)Buenos AiresArgentina
| | - Pablo I. Nikel
- The Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkKongens LyngbyDenmark
| | - Andrea M. Smania
- Universidad Nacional de CórdobaFacultad de Ciencias Químicas, Departamento de Química Biológica Ranwel CaputtoCórdobaArgentina
- CONICET, Universidad Nacional de CórdobaCentro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC)CórdobaArgentina
| |
Collapse
|
13
|
Nguyen E, Agbavor C, Steenhaut A, Pratyush MR, Hiller NL, Cahoon LA, Mikheyeva IV, Ng WL, Bridges AA. A small periplasmic protein governs broad physiological adaptations in Vibrio cholerae via regulation of the DbfRS two-component system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.645060. [PMID: 40196685 PMCID: PMC11974885 DOI: 10.1101/2025.03.24.645060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Two-component signaling pathways allow bacteria to sense and respond to environmental changes, yet the sensory mechanisms of many remain poorly understood. In the pathogen Vibrio cholerae, the DbfRS two-component system controls the biofilm lifecycle, a critical process for environmental persistence and host colonization. Here, we identified DbfQ, a small periplasmic protein encoded adjacent to dbfRS, as a direct modulator of pathway activity. DbfQ directly binds the sensory domain of the histidine kinase DbfS, shifting it toward phosphatase activity and promoting biofilm dispersal. In contrast, outer membrane perturbations, caused by mutations in lipopolysaccharide biosynthesis genes or membrane-damaging antimicrobials, activate phosphorylation of the response regulator DbfR. Transcriptomic analyses reveal that DbfR phosphorylation leads to broad transcriptional changes spanning genes involved in biofilm formation, central metabolism, peptidoglycan synthesis, and cellular stress responses. Constitutive DbfR phosphorylation imposes severe fitness costs in an infection model, highlighting this pathway as a potential target for anti-infective therapeutics. We find that dbfQRS-like genetic modules are widely present across bacterial phyla, underscoring their broad relevance in bacterial physiology. Collectively, these findings establish DbfQ as a new class of periplasmic regulator that influences two-component signaling and bacterial adaptation.
Collapse
Affiliation(s)
- Emmy Nguyen
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Charles Agbavor
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Anjali Steenhaut
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - M R Pratyush
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - N. Luisa Hiller
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Laty A. Cahoon
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Irina V. Mikheyeva
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Wai-Leung Ng
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Andrew A. Bridges
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
14
|
Tuytschaever T, Raes K, Sampers I. Biofilm detection in the food industry: Challenges in identifying biofilm eps markers and analytical techniques with insights for Listeria monocytogenes. Int J Food Microbiol 2025; 432:111091. [PMID: 39923351 DOI: 10.1016/j.ijfoodmicro.2025.111091] [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: 10/17/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Extracellular polymeric substances (EPS) in biofilms are promising targets for eradicating biofilms and monitoring their presence, especially in the food industry. For this understanding, the composition of the EPS matrix is crucial. Ideally, a biofilm marker is found serving both purposes, but such a compound has not yet been discovered. This review aims to identify general biofilm EPS markers distinct from planktonic cells, focusing on macromolecules in the EPS matrix. It also evaluates the feasibility of this goal across different bacterial groups and environmental conditions and discusses EPS analysis methods. This review digs deeper into the EPS matrix starting with an introduction to the EPS matrix itself and describing some of its influencing factors. Next, a brief description of cell-to-cell communication within biofilms is provided, as these interactions significantly influence the EPS matrix. The main part of this review describes the macromolecules inside the EPS matrix and attempts to find biofilm EPS markers applied to bacteria in general and specifically to Listeria monocytogenes as biofilms are a major contributor to its persistence. The last part of the review focuses on the analytical techniques available to characterize the EPS matrix. The review revealed that although multiple candidates showed great potential as biofilm markers, none were unique but ubiquitous in all bacteria tested. To achieve easy biofilm detection with current techniques, it's necessary to identify markers specific to the environmental conditions and common bacterial groups within each food category, sector, or facility, due to the lack of standardization in these techniques. This tailored approach ensures more accurate and effective biofilm monitoring. Moreover, the lack of standardized analytical techniques, including quantification techniques, complexifies studying the EPS matrix and developing monitoring and intervention strategies. Optimizing analytical techniques is crucial for this tailored approach, as it requires refined methods for detection, characterization, and quantification. This ensures the accurate identification of biofilm markers specific to environmental conditions and bacterial groups within each food sector.
Collapse
Affiliation(s)
- Tessa Tuytschaever
- Research Unit VEG-i-TEC, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Campus Kortrijk, Sint-Martens-Latemlaan 2B, 8500 Kortrijk, Belgium.
| | - Katleen Raes
- Research Unit VEG-i-TEC, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Campus Kortrijk, Sint-Martens-Latemlaan 2B, 8500 Kortrijk, Belgium.
| | - Imca Sampers
- Research Unit VEG-i-TEC, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Campus Kortrijk, Sint-Martens-Latemlaan 2B, 8500 Kortrijk, Belgium.
| |
Collapse
|
15
|
Feng Q, Dai X, Wu Q, Zhang L, Yang L, Fu Y. c-di-GMP phosphodiesterase ProE interacts with quorum sensing protein PqsE to promote pyocyanin production in Pseudomonas aeruginosa. mSphere 2025; 10:e0102624. [PMID: 39873511 PMCID: PMC11852716 DOI: 10.1128/msphere.01026-24] [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: 12/07/2024] [Accepted: 01/02/2025] [Indexed: 01/30/2025] Open
Abstract
The universal bacterial second messenger bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP) plays critical roles in regulating a variety of bacterial functions such as biofilm formation and virulence. The metabolism of c-di-GMP is inversely controlled by diguanylate cyclases (DGCs) and phosphodiesterases (PDEs). Recently, increasing studies suggested that the protein-protein interactions between DGCs/PDEs and their partners appear to be a common way to achieve specific regulation. In this work, we showed that the PDE ProE can interact with PQS quorum sensing protein PqsE to regulate pyocyanin production in Pseudomonas aeruginosa. Our bacterial two-hybrid assay demonstrated that ProE directly interacts with PqsE, and isothermal titration calorimetry and surface plasmon resonance assay further confirmed that the binding affinity of ProE with PqsE is at micromolar level. Both ProE and PqsE negatively regulate intracellular c-di-GMP levels. Furthermore, our transcriptomic study showed that co-expression of ProE and PqsE significantly changes the gene expression profiles in P. aeruginosa, especially with increased expression of pyocyanin genes, and the qPCR and phenotypic results confirmed the transcriptome data. Taken together, our study suggested that the interaction between ProE and PqsE plays a critical role in regulation of pyocyanin production and highlights the importance of protein-protein interaction mediated c-di-GMP signaling in P. aeruginosa.IMPORTANCEc-di-GMP is pivotal in orchestrating various bacterial functions. In Pseudomonas aeruginosa, the nuanced balance of intracellular c-di-GMP is maintained by approximately 41 diguanylate cyclases (DGCs) and phosphodiesterases (PDEs). Emerging studies indicate that the c-di-GMP metabolic DGCs and PDEs may be involved in the signal transduction process by directly binding to the target protein, thus influencing downstream function. Despite their known importance, the precise functions of these proteins, especially their interacting partners, remain unclear. In this study, we identified that PQS quorum sensing system protein PqsE is a binding partner of c-di-GMP phosphodiesterase ProE; further analysis suggested that the ProE specifically interacts with PqsE to promote pyocyanin production. Our study extended the regulatory mechanism of the c-di-GMP signal transduction and quorum sensing in governing bacterial physiology.
Collapse
Affiliation(s)
- Qishun Feng
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, China
| | - Xin Dai
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Qiulan Wu
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Lianhui Zhang
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China
| | - Liang Yang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yang Fu
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
16
|
Snega Priya P, Meenatchi R, Pasupuleti M, Namasivayam SKR, Arockiaraj J. Harnessing Cyclic di-GMP Signaling: A Strategic Approach to Combat Bacterial Biofilm-Associated Chronic Infections. Curr Microbiol 2025; 82:118. [PMID: 39909925 DOI: 10.1007/s00284-025-04091-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/11/2025] [Indexed: 02/07/2025]
Abstract
Cyclic dimeric guanosine monophosphate (c-di-GMP) plays a vital role within the nucleotide signaling network of bacteria, participating in various biological processes such as biofilm formation and toxin production, among others. Substantial evidence demonstrates its critical involvement in the progression of chronic infections. Treating chronic infections seems critical, and there is a worldwide quest for drugs that target pathogens' unique and complex virulence-associated signaling networks. c-di-GMP is a promising therapeutic target by serving as a distinct virulence factor, solving problems associated with drug resistance, biofilm dispersion, and its related septicemia complications. c-di-GMP levels act as checkpoints for several biofilm-associated molecular pathways, viz., Gac/Rsm, BrlR, and SagS signaling systems. C-di-GMP is also engaged in the Wsp chemosensory pathway responsible for rugose small colony variants observed in cystic fibrosis-related lung infections. Considering all factors, c-di-GMP serves as a pivotal hub in the intricate cascade of biofilm regulation. By overseeing QS systems, exopolysaccharide synthesis, and antibiotic resistance pathways in chronic infections, it emerges as a linchpin for effective drug development strategies against biofilm-related ailments. This underscores the significance of understanding the multifaceted signaling networks. c-di-GMP's role is highlighted in this review as a concealed virulence component in various bacterial pathogens, suggesting that medications targeting it could hold promise in treating chronic disorders associated with biofilms.
Collapse
Affiliation(s)
- P Snega Priya
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulatur, Tamil Nadu, 603203, India
| | - Ramu Meenatchi
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulatur, Tamil Nadu, 603203, India
| | - Mukesh Pasupuleti
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute (CDRI), Sitapur Road, Sector 10, Janakipuram Extension, Lucknow, 226031, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - S Karthick Raja Namasivayam
- Centre for Applied Research, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, 602105, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulatur, Tamil Nadu, 603203, India
| |
Collapse
|
17
|
Sowmeya VG, Sathiavelu M. Biofilm dynamics in space and their potential for sustainable space exploration - A comprehensive review. LIFE SCIENCES IN SPACE RESEARCH 2025; 44:108-121. [PMID: 39864903 DOI: 10.1016/j.lssr.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 08/23/2024] [Indexed: 01/28/2025]
Abstract
Microbial biofilms are universal. The intricate tapestry of biofilms has remarkable implications for the environment, health, and industrial processes. The field of space microbiology is actively investigating the effects of microgravity on microbes, and discoveries are constantly being made. Recent evidence suggests that extraterrestrial environments also fuel the biofilm formation. Understanding the biofilm mechanics under microgravitational conditions is crucial at this stage and could have an astounding impact on inter-planetary missions. This review systematically examines the existing understanding of biofilm development in space and provides insight into how molecules, physiology, or environmental factors influence biofilm formation during microgravitational conditions. In addition, biocontrol strategies targeting the formation and dispersal of biofilms in space environments are explored. In particular, the article highlights the potential benefits of using microbial biofilms in space for bioremediation, life support systems, and biomass production applications.
Collapse
Affiliation(s)
- V G Sowmeya
- School of Biosciences and Technology, VIT, Vellore 632014, India
| | | |
Collapse
|
18
|
Fogg PCM. Gene transfer agents: The ambiguous role of selfless viruses in genetic exchange and bacterial evolution. Mol Microbiol 2025; 123:124-131. [PMID: 38511257 PMCID: PMC11841831 DOI: 10.1111/mmi.15251] [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: 12/08/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024]
Abstract
Gene transfer agents (GTAs) are genetic elements derived from ancestral bacteriophages that have become domesticated by the host. GTAs are present in diverse prokaryotic organisms, where they can facilitate horizontal gene transfer under certain conditions. Unlike typical bacteriophages, GTAs do not exhibit any preference for the replication or transfer of the genes encoding them; instead, they exhibit a remarkable capacity to package chromosomal, and sometimes extrachromosomal, DNA into virus-like capsids and disseminate it to neighboring cells. Because GTAs resemble defective prophages, identification of novel GTAs is not trivial. The detection of candidates relies on the genetic similarity to known GTAs, which has been fruitful in α-proteobacterial lineages but challenging in more distant bacteria. Here we consider several fundamental questions: What is the true prevalence of GTAs in prokaryote genomes? Given there are high costs for GTA production, what advantage do GTAs provide to the bacterial host to justify their maintenance? How is the bacterial chromosome recognized and processed for inclusion in GTA particles? This article highlights the challenges in comprehensively understanding GTAs' prevalence, function and DNA packaging method. Going forward, broad study of atypical GTAs and use of ecologically relevant conditions are required to uncover their true impact on bacterial chromosome evolution.
Collapse
|
19
|
Bagchi S, Sharma AK, Mal S, Kundu M, Basu J. Crosstalk between cyclic-di-guanosine monophosphate and the sensor kinase MtrB regulates MtrA-dependent genes, bacterial growth, biofilm formation and lysosomal trafficking of Mycobacterium tuberculosis. MICROBIOLOGY (READING, ENGLAND) 2025; 171. [PMID: 39918987 DOI: 10.1099/mic.0.001532] [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: 02/09/2025]
Abstract
Cyclic-di-guanosine monophosphate (c-di-GMP) plays an important role in bacterial signalling networks. C-di-GMP exerts a regulatory function through binding to diverse molecules that include transcription factors, riboswitches and sensor kinases (SKs), thereby regulating diverse processes. Here, we demonstrate the crosstalk between c-di-GMP and the SK MtrB of Mycobacterium tuberculosis. MtrB phosphorylates and regulates its cognate response regulator MtrA. C-di-GMP binds directly to the cytosolic domain of MtrB to inhibit its autophosphorylation. C-di-GMP levels in M. tuberculosis were manipulated by overexpressing a c-di-GMP synthesizing enzyme ydeH and a degrading enzyme rv1357c. We demonstrate that overexpression of ydeH lowers growth of the bacterium both in vitro and in M. tuberculosis grown in macrophages. This is in conformity with lowered expression of mtrA and selected genes of the mtrA regulon involved in cell wall turnover in the ydeH-overexpressing strain compared to the parent strain. We also demonstrate that overexpression of ydeH in M. tuberculosis hinders biofilm formation, whereas overexpression of rv1357c has the opposite effect. Neither of the two genes could rescue the biofilm defective phenotype of the MtrB knock out mutant (ΔmtrB), suggesting that c-di-GMP exerts its role on biofilm formation through MtrB. Finally, we show by fluorescence microscopy that the trafficking of M. tuberculosis overexpressing ydeH is significantly higher than that of the parent strain and that this is linked to reduced expression of the MtrB-dependent genes esxG and esxH, which play a role in subversion of lysosomal trafficking of M. tuberculosis. These results provide important new insight into the crosstalk between c-di-GMP and MtrB in M. tuberculosis.
Collapse
Affiliation(s)
- Shreya Bagchi
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| | - Arun Kumar Sharma
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| | - Soumya Mal
- Department of Biological Sciences, Bose Institute, Unified Academic Campus, EN 80, Sector V, Bidhan Nagar, Kolkata 700091, India
| | - Manikuntala Kundu
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| | - Joyoti Basu
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| |
Collapse
|
20
|
Xue Y, Kang X. Time-resolved compositional and dynamics analysis of biofilm maturation and dispersal via solid-state NMR spectroscopy. NPJ Biofilms Microbiomes 2025; 11:21. [PMID: 39880834 PMCID: PMC11779841 DOI: 10.1038/s41522-025-00655-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
Dispersal plays a crucial role in the development and ecology of biofilms. While extensive studies focused on elucidating the molecular mechanisms governing this process, few have characterized the associated temporal changes in composition and structure. Here, we employed solid-state nuclear magnetic resonance (NMR) techniques to achieve time-resolved characterization of Bacillus subtilis biofilms over a 5-day period. The mature biofilm, established within 48 h, undergoes significant degradation in following 72 h. The steepest decline of proteins precedes that of exopolysaccharides, likely reflecting their distinct spatial distribution. Exopolysaccharide sugar units display clustered temporal patterns, suggesting the presence of distinct polysaccharide types. A sharp rise in aliphatic carbon signals on day 4 probably corresponds to a surge in biosurfactant production. Different dynamic regimes respond differently to dispersal: the mobile domain exhibits increased rigidity, while the rigid domain remains stable. These findings provide novel insights and perspectives on the complex process of biofilm dispersal.
Collapse
Affiliation(s)
- Yi Xue
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Xue Kang
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, Zhejiang, China.
| |
Collapse
|
21
|
Carey CJ, Duggan N, Drabinska J, McClean S. Harnessing hypoxia: bacterial adaptation and chronic infection in cystic fibrosis. FEMS Microbiol Rev 2025; 49:fuaf018. [PMID: 40312783 PMCID: PMC12071387 DOI: 10.1093/femsre/fuaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 04/04/2025] [Accepted: 04/29/2025] [Indexed: 05/03/2025] Open
Abstract
The exquisite ability of bacteria to adapt to their environment is essential for their capacity to colonize hostile niches. In the cystic fibrosis (CF) lung, hypoxia is among several environmental stresses that opportunistic pathogens must overcome to persist and chronically colonize. Although the role of hypoxia in the host has been widely reviewed, the impact of hypoxia on bacterial pathogens has not yet been studied extensively. This review considers the bacterial oxygen-sensing mechanisms in three species that effectively colonize the lungs of people with CF, namely Pseudomonas aeruginosa, Burkholderia cepacia complex, and Mycobacterium abscessus and draws parallels between their three proposed oxygen-sensing two-component systems: BfiSR, FixLJ, and DosRS, respectively. Moreover, each species expresses regulons that respond to hypoxia: Anr, Lxa, and DosR, and encode multiple proteins that share similar homologies and function. Many adaptations that these pathogens undergo during chronic infection, including antibiotic resistance, protease expression, or changes in motility, have parallels in the responses of the respective species to hypoxia. It is likely that exposure to hypoxia in their environmental habitats predispose these pathogens to colonization of hypoxic niches, arming them with mechanisms than enable their evasion of the immune system and establish chronic infections. Overcoming hypoxia presents a new target for therapeutic options against chronic lung infections.
Collapse
Affiliation(s)
- Ciarán J Carey
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Niamh Duggan
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Joanna Drabinska
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Siobhán McClean
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
22
|
Hanot M, Lohou E, Sonnet P. Anti-Biofilm Agents to Overcome Pseudomonas aeruginosa Antibiotic Resistance. Pharmaceuticals (Basel) 2025; 18:92. [PMID: 39861155 PMCID: PMC11768670 DOI: 10.3390/ph18010092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/06/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Pseudomonas aeruginosa is one of world's most threatening bacteria. In addition to the emerging prevalence of multi-drug resistant (MDR) strains, the bacterium also possesses a wide variety of virulence traits that worsen the course of the infections. Particularly, its ability to form biofilms that protect colonies from antimicrobial agents is a major cause of chronic and hard-to-treat infections in immune-compromised patients. This protective barrier also ensures cell growth on abiotic surfaces and thus enables bacterial survival on medical devices. Hence, as the WHO alerted to the need to develop new treatments, the use of anti-biofilm agents (ABAs) appeared as a promising approach. Given the selection pressure imposed by conventional antibiotics, a new therapeutic strategy has emerged that aims at reducing bacterial virulence without inhibiting cell growth. So-called anti-virulence agents (AVAs) would then restore the efficacy of conventional antibiotics (ATBs) or potentiate the effectiveness of the immune system. The last decade has seen the development of ABAs as AVAs against P. aeruginosa. This review aims to highlight the design strategy and critical features of these molecules to pave the way for further discoveries of highly potent compounds.
Collapse
Affiliation(s)
| | | | - Pascal Sonnet
- AGIR, UR 4294, Faculté de Pharmacie, Université de Picardie Jules Verne, 1 Rue des Louvels, 80000 Amiens, France; (M.H.); (E.L.)
| |
Collapse
|
23
|
Azeem K, Fatima S, Ali A, Ubaid A, Husain FM, Abid M. Biochemistry of Bacterial Biofilm: Insights into Antibiotic Resistance Mechanisms and Therapeutic Intervention. Life (Basel) 2025; 15:49. [PMID: 39859989 PMCID: PMC11767195 DOI: 10.3390/life15010049] [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: 12/01/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Biofilms, composed of structured communities of bacteria embedded in a self-produced extracellular matrix, pose a significant challenge due to their heightened resistance to antibiotics and immune responses. This review highlights the mechanisms underpinning antibiotic resistance within bacterial biofilms, elucidating the adaptive strategies employed by microorganisms to withstand conventional antimicrobial agents. This encompasses the role of the extracellular matrix, altered gene expression, and the formation of persister cells, contributing to the recalcitrance of biofilms to eradication. A comprehensive understanding of these resistance mechanisms provides a for exploring innovative therapeutic interventions. This study explores promising avenues for future research, emphasizing the necessity of uncovering the specific genetic and phenotypic adaptations occurring within biofilms. The identification of vulnerabilities in biofilm architecture and the elucidation of key biofilm-specific targets emerge as crucial focal points for the development of targeted therapeutic strategies. In addressing the limitations of traditional antibiotics, this review discusses innovative therapeutic approaches. Nanomaterials with inherent antimicrobial properties, quorum-sensing inhibitors disrupting bacterial communication, and bacteriophages as biofilm-specific viral agents are highlighted as potential alternatives. The exploration of combination therapies, involving antimicrobial agents, biofilm-disrupting enzymes, and immunomodulators, is emphasized to enhance the efficacy of existing treatments and overcome biofilm resilience.
Collapse
Affiliation(s)
- Kashish Azeem
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Sadaf Fatima
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Asghar Ali
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
- Clinical Biochemistry Laboratory, Department of Biochemistry, School of Chemical and Life Science, Jamia Hamdard, New Delhi 110062, India
| | - Ayesha Ubaid
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Abid
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| |
Collapse
|
24
|
Agarwal H, Gurnani B, Pippal B, Jain N. Capturing the micro-communities: Insights into biogenesis and architecture of bacterial biofilms. BBA ADVANCES 2024; 7:100133. [PMID: 39839441 PMCID: PMC11750278 DOI: 10.1016/j.bbadva.2024.100133] [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] [Indexed: 01/23/2025] Open
Abstract
Biofilm is an assemblage of microorganisms embedded within the extracellular matrix that provides mechanical stability, nutrient absorption, antimicrobial resistance, cell-cell interactions, and defence against host immune system. Various biomolecules such as lipids, carbohydrates, protein polymers (amyloid), and eDNA are present in the matrix playing significant role in determining the distinctive properties of biofilm. The formation of biofilms contributes to resistance against antimicrobial therapy in most of the human infections and exacerbates existing diseases. Therefore, this field requires several state-of-the-art techniques to fully understand the 3-D organization of biofilms, their cell behaviour and responses to pharmaceutical treatments. Here, we explore the assembly and regulation of biofilm biogenesis in the context of matrix components and highlight the significance of high-resolution imaging and analysing techniques for monitoring complex biofilm architecture. Our review also emphasizes the novelty and advancements in techniques to visualise biofilm structure and composition, providing valuable insights to understand biofilm-related infections.
Collapse
Affiliation(s)
- Harshita Agarwal
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Nagaur Road, Karwar, Rajasthan 342037, India
| | - Bharat Gurnani
- Centre of Excellence-AyurTech, Indian Institute of Technology Jodhpur, NH 65, Nagaur Road, Karwar, Rajasthan 342037, India
| | - Bhumika Pippal
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Nagaur Road, Karwar, Rajasthan 342037, India
| | - Neha Jain
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Nagaur Road, Karwar, Rajasthan 342037, India
| |
Collapse
|
25
|
Kalia M, Sauer K. Distinct transcriptome and traits of freshly dispersed Pseudomonas aeruginosa cells. mSphere 2024; 9:e0088424. [PMID: 39601567 DOI: 10.1128/msphere.00884-24] [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: 10/17/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Bacteria assume two distinct lifestyles: the planktonic and biofilm modes of growth. Additionally, dispersion has emerged as a third phenotype, accompanied by the distinct phenotypes and the unique expression of >600 genes. Here, we asked whether the distinct phenotype of dispersed cells is already apparent within minutes of egressing from the biofilm. We used RNA-seq to show that the physiology of freshly dispersed cells from Pseudomonas aeruginosa biofilms is highly different from those of planktonic and biofilm cells, apparent by dispersed cells uniquely expressing 194 genes. Unique and differentially expressed genes relative to planktonic or biofilm cells include genes associated with type IV pili, pyoverdine, type III and type VI secretion systems, and antibiotic resistance that are downregulated in dispersed cells, whereas the transcript abundance of genes involved in swimming motility, Hxc type II secretion system and various other virulence factors, and metabolic and energy-generating pathways are increased, indicative of dispersion coinciding with an awakening and re-energizing of dispersed cells, and a switch in virulence, further apparent by freshly dispersed cells significantly subverting engulfment by macrophages. The findings suggest that dispersed cells display a distinct phenotype within minutes of egressing from the biofilm, with freshly dispersed cells already capable of efficiently evading phagocytosis. IMPORTANCE Dispersion is considered a transitionary phenotype, enabling bacteria to switch between the communal, biofilm lifestyle, where cells share resources and are protected from harmful conditions to the planktonic state. Here, we demonstrate that within minutes of leaving the biofilm, dispersed cells express genes and display phenotypic traits that are distinct from biofilms and planktonic cells. Our findings suggest that dispersed cells quickly adapt to a less structured and protected but more nutrient-rich environment, with this trade-off in environment coinciding with an awakening and a switch in virulence, specifically a switch from directly intoxicating host cells and potential competitors toward more broadly active virulence factors and strategies of evasion. To our knowledge, this is the first report of dispersed cells' distinct (trade-off) phenotype and their enhanced resilience so soon after egressing from the biofilm.
Collapse
Affiliation(s)
- Manmohit Kalia
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | - Karin Sauer
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
26
|
Roberts JM, Milo S, Metcalf DG. Harnessing the Power of Our Immune System: The Antimicrobial and Antibiofilm Properties of Nitric Oxide. Microorganisms 2024; 12:2543. [PMID: 39770746 PMCID: PMC11677572 DOI: 10.3390/microorganisms12122543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Nitric oxide (NO) is a free radical of the human innate immune response to invading pathogens. NO, produced by nitric oxide synthases (NOSs), is used by the immune system to kill microorganisms encapsulated within phagosomes via protein and DNA disruption. Owing to its ability to disperse biofilm-bound microorganisms, penetrate the biofilm matrix, and act as a signal molecule, NO may also be effective as an antibiofilm agent. NO can be considered an underappreciated antimicrobial that could be levied against infected, at-risk, and hard-to-heal wounds due to the inherent lack of bacterial resistance, and tolerance by human tissues. NO produced within a wound dressing may be an effective method of disrupting biofilms and killing microorganisms in hard-to-heal wounds such as diabetic foot ulcers, venous leg ulcers, and pressure injuries. We have conducted a narrative review of the evidence underlying the key antimicrobial and antibiofilm mechanisms of action of NO for it to serve as an exogenously-produced antimicrobial agent in dressings used in the treatment of hard-to-heal wounds.
Collapse
Affiliation(s)
| | | | - Daniel Gary Metcalf
- Advanced Wound Care Research & Development, Convatec, Deeside Industrial Park, Deeside CH5 2NU, UK; (J.M.R.); (S.M.)
| |
Collapse
|
27
|
Luo X, Zhang M, Zhang Y, Li X, Lu R. Phenotypic changes and gene expression profiles of Vibrio parahaemolyticus in response to low concentrations of ampicillin. J Antibiot (Tokyo) 2024; 77:823-836. [PMID: 39322835 DOI: 10.1038/s41429-024-00772-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/28/2024] [Accepted: 09/01/2024] [Indexed: 09/27/2024]
Abstract
Vibrio parahaemolyticus is a leading cause of seafood-associated gastroenteritis and possesses intrinsic resistance to ampicillin. While ampicillin can trigger transcriptional responses of global genes, the behavioral and molecular changes that occur in V. parahaemolyticus when exposed to ampicillin are not fully understood. In this work, we investigated the effects of low concentrations of ampicillin on the physiology and gene expression of V. parahaemolyticus by combining phenotypic assays and RNA sequencing (RNA-seq) analysis. Our results showed that the growth of V. parahaemolyticus were notably delayed, and both motility and c-di-GMP production were significantly inhibited in the response to low concentrations of ampicillin stress. In contrast, biofilm formation by V. parahaemolyticus was enhanced by exposure to low concentrations of ampicillin. However, low concentrations of ampicillin had no effect on the cytotoxicity or adherence activity of V. parahaemolyticus. The RNA-seq data revealed that a low concentration of ampicillin significantly affected the expression levels of 676 genes, including those involved in antibiotic resistance, virulence, biofilm formation, and regulation. This work contributes to our understanding of how V. parahaemolyticus alters its behavior and gene expression in response to ampicillin exposure.
Collapse
Affiliation(s)
- Xi Luo
- Department of Clinical Laboratory, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Miaomiao Zhang
- Department of Clinical Laboratory, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Yiquan Zhang
- Department of Clinical Laboratory, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China.
| | - Xue Li
- Department of Clinical Laboratory, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Renfei Lu
- Department of Clinical Laboratory, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
28
|
Wang Z, Wu Y, Liu M, Chen L, Xiao K, Huang Z, Zhao Y, Wang H, Ding Y, Lin X, Zeng J, Peng F, Zhang J, Wang J, Wu Q. The Gene Cluster Cj0423- Cj0425 Negatively Regulates Biofilm Formation in Campylobacter jejuni. Int J Mol Sci 2024; 25:12116. [PMID: 39596184 PMCID: PMC11595200 DOI: 10.3390/ijms252212116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/02/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
Campylobacter jejuni (C. jejuni) is a zoonotic foodborne pathogen that is widely distributed worldwide. Its optimal growth environment is microaerophilic conditions (5% O2, 10% CO2), but it can spread widely in the atmospheric environment. Biofilms are thought to play an important role in this process. However, there are currently relatively few research works on the regulatory mechanisms of C. jejuni biofilm formation. In this study, a pan-genome analysis, combined with the analysis of biofilm phenotypic information, revealed that the gene cluster Cj0423-Cj0425 is associated with the negative regulation of biofilm formation in C. jejuni. Through gene knockout experiments, it was observed that the Cj0423-Cj0425 mutant strain significantly increased biofilm formation and enhanced flagella formation. Furthermore, pull-down assay revealed that Cj0424 interacts with 93 proteins involved in pathways such as fatty acid synthesis and amino acid metabolism, and it also contains the quorum sensing-related gene luxS. This suggests that Cj0423-Cj0425 affects fatty acid synthesis and amino acid metabolism, influencing quorum sensing and strain motility, ultimately inhibiting biofilm formation.
Collapse
Affiliation(s)
- Zhi Wang
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510641, China; (Z.W.); (M.L.)
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (Y.W.); (L.C.); (K.X.); (Y.Z.); (H.W.); (X.L.); (J.Z.); (F.P.); (J.Z.)
| | - Yuwei Wu
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (Y.W.); (L.C.); (K.X.); (Y.Z.); (H.W.); (X.L.); (J.Z.); (F.P.); (J.Z.)
| | - Ming Liu
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510641, China; (Z.W.); (M.L.)
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (Y.W.); (L.C.); (K.X.); (Y.Z.); (H.W.); (X.L.); (J.Z.); (F.P.); (J.Z.)
| | - Ling Chen
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (Y.W.); (L.C.); (K.X.); (Y.Z.); (H.W.); (X.L.); (J.Z.); (F.P.); (J.Z.)
| | - Kaishan Xiao
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (Y.W.); (L.C.); (K.X.); (Y.Z.); (H.W.); (X.L.); (J.Z.); (F.P.); (J.Z.)
- College of Food Science, South China Agricultural University, Guangzhou 510432, China
| | - Zhenying Huang
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (Y.W.); (L.C.); (K.X.); (Y.Z.); (H.W.); (X.L.); (J.Z.); (F.P.); (J.Z.)
- College of Food Science, South China Agricultural University, Guangzhou 510432, China
| | - Yibing Zhao
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (Y.W.); (L.C.); (K.X.); (Y.Z.); (H.W.); (X.L.); (J.Z.); (F.P.); (J.Z.)
- College of Food Science, South China Agricultural University, Guangzhou 510432, China
| | - Huixian Wang
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (Y.W.); (L.C.); (K.X.); (Y.Z.); (H.W.); (X.L.); (J.Z.); (F.P.); (J.Z.)
| | - Yu Ding
- Department of Food Science and Technology, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China;
| | - Xiuhua Lin
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (Y.W.); (L.C.); (K.X.); (Y.Z.); (H.W.); (X.L.); (J.Z.); (F.P.); (J.Z.)
| | - Jiahui Zeng
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (Y.W.); (L.C.); (K.X.); (Y.Z.); (H.W.); (X.L.); (J.Z.); (F.P.); (J.Z.)
| | - Feiting Peng
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (Y.W.); (L.C.); (K.X.); (Y.Z.); (H.W.); (X.L.); (J.Z.); (F.P.); (J.Z.)
| | - Jumei Zhang
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (Y.W.); (L.C.); (K.X.); (Y.Z.); (H.W.); (X.L.); (J.Z.); (F.P.); (J.Z.)
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou 510432, China
| | - Qingping Wu
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510641, China; (Z.W.); (M.L.)
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (Y.W.); (L.C.); (K.X.); (Y.Z.); (H.W.); (X.L.); (J.Z.); (F.P.); (J.Z.)
| |
Collapse
|
29
|
Yu L, Wang H, Zhang X, Xue T. Oxidative stress response in avian pathogenic Escherichia coli. Res Vet Sci 2024; 180:105426. [PMID: 39342922 DOI: 10.1016/j.rvsc.2024.105426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/09/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Avian pathogenic Escherichia coli (APEC) leads to significant economic losses in the poultry industry worldwide and restricts the development of the poultry industry. Oxidative stress, through the production of reactive oxygen species (ROS), damage iron‑sulfur (FeS) clusters, cysteine and methionine protein residues, and DNA, and then result in bacterial cells death. APEC has evolved a series of regulation systems to sense and quickly and appropriately respond to oxidative stress. Quorum sensing (QS), second messenger (SM), transcription factors (TFs), small regulatory RNAs (sRNAs), and two-component system (TCS) are important regulation systems ubiquitous in bacteria. It is of great significance to control APEC infection through investigating the molecular regulation mechanism on APEC adapting to oxidative stress. However, how the cross-talk among these regulation systems co-regulates transcription of oxidative stress-response genes in APEC has not been reported. This review suggests exploring connector proteins that co-regulate these regulation systems that co-activate transcription of oxidative stress-response genes to disrupt bacterial antioxidative defense mechanism in APEC, and then using these connector proteins as drug targets to control APEC infection. This review might contribute to illustrating the functional mechanism of APEC adapting to oxidative stress and exploring potential drug targets for the prevention and treatment of APEC infection.
Collapse
Affiliation(s)
- Lumin Yu
- Institute of Microbe and Host Health, Linyi University, Linyi, Shandong 276005, China.
| | - Hui Wang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xinglin Zhang
- Institute of Microbe and Host Health, Linyi University, Linyi, Shandong 276005, China
| | - Ting Xue
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China.
| |
Collapse
|
30
|
Sun X, Xiao F, Su Y, Li Z, Yu X, Parales RE, Li L. Cyclic di-GMP incorporates the transcriptional factor FleQ03 in Pseudomonas syringae MB03 to elicit biofilm-dependent resistance in response to Caenorhabditis elegans predation. J Invertebr Pathol 2024; 207:108189. [PMID: 39251105 DOI: 10.1016/j.jip.2024.108189] [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: 03/23/2024] [Revised: 08/24/2024] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
Bacteria usually form biofilms as a defense mechanism against predation by bacterivorous nematodes. In this context, the second messenger c-di-GMP from the wild-type Pseudomonas syringae MB03 actuates the transcriptional factor FleQ03 to elicit biofilm-dependent nematicidal activity against Caenorhabditis elegans N2. P. syringae MB03 cells exhibited nematicidal activity and c-di-GMP content in P. syringae MB03 cells was increased after feeding to nematodes. Expression of a diguanylate cyclase (DGC) gene in P. syringae MB03 resulted in an increased c-di-GMP content, biofilm yield and nematicidal activity, whereas converse effects were obtained when expressing a phosphodiesterase (PDE) gene. Molecular docking and isothermal titration calorimetry assays verified the affinity activity between c-di-GMP and the FleQ03 protein. The disruption of the fleQ03 gene in P. syringae MB03, while increasing c-di-GMP content, significantly diminished both biofilm formation and nematicidal activity. Interestingly, P. syringae MB03 formed a full-body biofilm around the worms against predation, probably extending from the tail to the head, whereas it was not observed in the fleQ03 gene disrupted cells. Thus, we hypothesized that c-di-GMP incorporated FleQ03 to reinforce bacterial biofilm and biofilm-dependent pathogenicity in response to C. elegans predation, providing insights into a possible means of resisting bacterivorous nematodes by bacteria in natural ecosystems.
Collapse
Affiliation(s)
- Xiaowen Sun
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; School of Life Sciences, Jianghan University, Wuhan 430056, China
| | - Fan Xiao
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yuwei Su
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhi Li
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xun Yu
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Rebecca E Parales
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, USA.
| | - Lin Li
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
31
|
Manna T, Chandra Guchhait K, Jana D, Dey S, Karmakar M, Hazra S, Manna M, Jana P, Panda AK, Ghosh C. Wastewater-based surveillance of Vibrio cholerae: Molecular insights on biofilm regulatory diguanylate cyclases, virulence factors and antibiotic resistance patterns. Microb Pathog 2024; 196:106995. [PMID: 39368563 DOI: 10.1016/j.micpath.2024.106995] [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: 07/30/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
Vibrio cholerae is an inherent inhabitant of aquatic ecosystems. The Indian state of West Bengal, especially the Gangetic delta region is the highest cholera affected region and is considered as the hub of Asiatic cholera. V. cholerae were isolated from publicly accessible wastewater of Midnapore, West Bengal, India. Serotyping determined all isolates to be of non-O1/non-O139 serogroups. Moderate biofilm-forming abilities were noticed in most of the isolates (74.7 %) while, high biofilm formation was recorded for only 6.3 % isolates and 19 % of isolates exhibited low/non-biofilm-forming abilities. PCR-based screening of crucial diguanylate cyclases (DGCs) involved in cyclic-di-GMP-mediated biofilm signaling was performed. cdgH and cdgM were the most abundant DGCs among 93.7 % and 91.5 % of isolates, respectively. Other important DGCs, i.e., cdgK, cdgA, cdgL, and vpvC were present in 84 %, 75.5 %, 72 % and 68 % of isolates, respectively. Besides, the non-O1/non-O139 isolates were screened for the occurrence of virulence factor encoding genes. Moreover, among these non-O1/non-O139 isolates, two strains (3.17 %) harbored both ctxA and ctxB genes, which encode the cholera toxin associated with epidemic cholera. ompU was the most prevalent virulence factor, present in 24.8 % of isolates. Other virulence factors like, zot and st were found in 4.7 % and 9.5 % of isolates. Genes encoding tcp and ace were found to be PCR-negative for the isolates. Additionally, crucial virulence factor regulators, toxT, toxR and hapR were found to be PCR-positive in all the isolates. Antibiotic resistance patterns displayed further vulnerabilities with decreased sensitivity towards commonly used antibiotics with multiple antibiotic resistance index ranging between 0.37 and 0.62. The presence of cholera toxin-encoding multi-drug resistant (MDR) V. cholerae strains in environmental settings is alarming. High occurrence of DGCs are considered to encourage further investigations to use them as alternative therapeutic targets against MDR cholera pathogen due to their unique presence in bacterial systems.
Collapse
Affiliation(s)
- Tuhin Manna
- Deparment of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | | | - Debarati Jana
- Deparment of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Subhamoy Dey
- Deparment of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India; Centre for Life Sciences, Vidyasagar University, Midnapore, West Bengal, India
| | - Monalisha Karmakar
- Deparment of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Subrata Hazra
- Deparment of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Mousumi Manna
- Deparment of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Pradip Jana
- Deparment of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Amiya Kumar Panda
- Department of Chemistry, Vidyasagar University, Midnapore, West Bengal, India
| | - Chandradipa Ghosh
- Deparment of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India.
| |
Collapse
|
32
|
Di Nezio F, Ong ILH, Riedel R, Goshal A, Dhar J, Roman S, Storelli N, Sengupta A. Synergistic phenotypic adaptations of motile purple sulphur bacteria Chromatium okenii during lake-to-laboratory domestication. PLoS One 2024; 19:e0310265. [PMID: 39436933 PMCID: PMC11495639 DOI: 10.1371/journal.pone.0310265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/05/2024] [Indexed: 10/25/2024] Open
Abstract
Isolating microorganisms from natural environments for cultivation under optimized laboratory settings has markedly improved our understanding of microbial ecology. Artificial growth conditions often diverge from those in natural ecosystems, forcing wild isolates into distinct selective pressures, resulting in diverse eco-physiological adaptations mediated by modification of key phenotypic traits. For motile microorganisms we still lack a biophysical understanding of the relevant traits emerging during domestication and their mechanistic interplay driving short-to-long-term microbial adaptation under laboratory conditions. Using microfluidics, atomic force microscopy, quantitative imaging, and mathematical modeling, we study phenotypic adaptation of Chromatium okenii, a motile phototrophic purple sulfur bacterium from meromictic Lake Cadagno, grown under laboratory conditions over multiple generations. Our results indicate that naturally planktonic C. okenii leverage shifts in cell-surface adhesive interactions, synergistically with changes in cell morphology, mass density, and distribution of intracellular sulfur globules, to suppress their swimming traits, ultimately switching to a sessile lifeform. A computational model of cell mechanics confirms the role of such phenotypic shifts in suppressing the planktonic lifeform. By investigating key phenotypic traits across different physiological stages of lab-grown C. okenii, we uncover a progressive loss of motility during the early stages of domestication, followed by concomitant deflagellation and enhanced surface attachment, ultimately driving the transition of motile sulfur bacteria to a sessile state. Our results establish a mechanistic link between suppression of motility and surface attachment via phenotypic changes, underscoring the emergence of adaptive fitness under laboratory conditions at the expense of traits tailored for natural environments.
Collapse
Affiliation(s)
- Francesco Di Nezio
- Department of Environment, Institute of Microbiology, Constructions and Design, University of Applied Sciences and Arts of Southern Switzerland (SUPSI), Mendrisio, Switzerland
- Microbiology Unit, Department of Plant Sciences, University of Geneva, Geneva, Switzerland
| | - Irvine Lian Hao Ong
- Physics of Living Matter Group, Department of Physics and Materials Science, University of Luxembourg, Luxembourg City, Luxembourg
| | - René Riedel
- Physics of Living Matter Group, Department of Physics and Materials Science, University of Luxembourg, Luxembourg City, Luxembourg
| | - Arkajyoti Goshal
- Physics of Living Matter Group, Department of Physics and Materials Science, University of Luxembourg, Luxembourg City, Luxembourg
| | - Jayabrata Dhar
- Department of Mechanical Engineering, National Institute of Technology, Durgapur, India
| | - Samuele Roman
- Department of Environment, Institute of Microbiology, Constructions and Design, University of Applied Sciences and Arts of Southern Switzerland (SUPSI), Mendrisio, Switzerland
- Alpine Biology Center Foundation, Bellinzona, Switzerland
| | - Nicola Storelli
- Department of Environment, Institute of Microbiology, Constructions and Design, University of Applied Sciences and Arts of Southern Switzerland (SUPSI), Mendrisio, Switzerland
- Microbiology Unit, Department of Plant Sciences, University of Geneva, Geneva, Switzerland
| | - Anupam Sengupta
- Physics of Living Matter Group, Department of Physics and Materials Science, University of Luxembourg, Luxembourg City, Luxembourg
- Institute for Advanced Studies, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
33
|
Liao H, Yan X, Wang C, Huang C, Zhang W, Xiao L, Jiang J, Bao Y, Huang T, Zhang H, Guo C, Zhang Y, Pu Y. Cyclic di-GMP as an antitoxin regulates bacterial genome stability and antibiotic persistence in biofilms. eLife 2024; 13:RP99194. [PMID: 39365286 PMCID: PMC11452175 DOI: 10.7554/elife.99194] [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: 10/05/2024] Open
Abstract
Biofilms are complex bacterial communities characterized by a high persister prevalence, which contributes to chronic and relapsing infections. Historically, persister formation in biofilms has been linked to constraints imposed by their dense structures. However, we observed an elevated persister frequency accompanying the stage of cell adhesion, marking the onset of biofilm development. Subsequent mechanistic studies uncovered a comparable type of toxin-antitoxin (TA) module (TA-like system) triggered by cell adhesion, which is responsible for this elevation. In this module, the toxin HipH acts as a genotoxic deoxyribonuclease, inducing DNA double strand breaks and genome instability. While the second messenger c-di-GMP functions as the antitoxin, exerting control over HipH expression and activity. The dynamic interplay between c-di-GMP and HipH levels emerges as a crucial determinant governing genome stability and persister generation within biofilms. These findings unveil a unique TA system, where small molecules act as the antitoxin, outlining a biofilm-specific molecular mechanism influencing genome stability and antibiotic persistence, with potential implications for treating biofilm infections.
Collapse
Affiliation(s)
- Hebin Liao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
- Translational Medicine Research Center, North Sichuan Medical CollegeNanchongChina
| | - Xiaodan Yan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
| | - Chenyi Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
| | - Chun Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
| | - Wei Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
| | - Leyi Xiao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
| | - Jun Jiang
- Center for Life Sciences, School of Life Sciences, Yunnan UniversityKunmingChina
| | - Yongjia Bao
- Center for Life Sciences, School of Life Sciences, Yunnan UniversityKunmingChina
| | - Tao Huang
- Center for Life Sciences, School of Life Sciences, Yunnan UniversityKunmingChina
| | - Hanbo Zhang
- Center for Life Sciences, School of Life Sciences, Yunnan UniversityKunmingChina
| | - Chunming Guo
- Center for Life Sciences, School of Life Sciences, Yunnan UniversityKunmingChina
| | - Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
- Taikang Center for Life and Medical Sciences, Wuhan UniversityWuhanChina
| | - Yingying Pu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Wuhan UniversityWuhanChina
- Department of Immunology, Hubei Province Key Laboratory of Allergy and Immunology, State Key Laboratory of Virology and Medical Research Institute, Wuhan University School of Basic Medical SciencesWuhanChina
| |
Collapse
|
34
|
Yeung YWS, Ma Y, Deng Y, Khoo BL, Chua SL. Bacterial Iron Siderophore Drives Tumor Survival and Ferroptosis Resistance in a Biofilm-Tumor Spheroid Coculture Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404467. [PMID: 39135304 PMCID: PMC11496991 DOI: 10.1002/advs.202404467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Indexed: 10/25/2024]
Abstract
Interactions between tumoral cells and tumor-associated bacteria within the tumor microenvironment play a significant role in tumor survival and progression, potentially impacting cancer treatment outcomes. In lung cancer patients, the Gram-negative pathogen Pseudomonas aeruginosa raises questions about its role in tumor survival. Here, a microfluidic-based 3D-human lung tumor spheroid-P. aeruginosa model is developed to study the bacteria's impact on tumor survival. P. aeruginosa forms a tumor-associated biofilm by producing Psl exopolysaccharide and secreting iron-scavenging pyoverdine, which is critical for establishing a bacterial community in tumors. Consequently, pyoverdine promotes cancer progression by reducing susceptibility to iron-induced death (ferroptosis), enhancing cell viability, and facilitating several cancer hallmarks, including epithelial-mesenchymal transition and metastasis. A promising combinatorial therapy approach using antimicrobial tobramycin, ferroptosis-inducing thiostrepton, and anti-cancer doxorubicin could eradicate biofilms and tumors. This work unveils a novel phenomenon of cross-kingdom cooperation, where bacteria protect tumors from death, and it paves the way for future research in developing antibiofilm cancer therapies. Understanding these interactions offers potential new strategies for combatting cancer and enhancing treatment efficacy.
Collapse
Affiliation(s)
- Yoyo Wing Suet Yeung
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityKowloonHong Kong SAR999077China
| | - Yeping Ma
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityKowloonHong Kong SAR999077China
| | - Yanlin Deng
- Department of Biomedical EngineeringCity University of Hong KongHong Kong SAR999077China
| | - Bee Luan Khoo
- Department of Biomedical EngineeringCity University of Hong KongHong Kong SAR999077China
- Hong Kong Center for Cerebro‐Cardiovascular Health Engineering (COCHE)Hong Kong SAR999077China
- City University of Hong Kong‐Shenzhen Futian Research InstituteShenzhen518000China
| | - Song Lin Chua
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityKowloonHong Kong SAR999077China
- State Key Laboratory of Chemical Biology and Drug DiscoveryThe Hong Kong Polytechnic UniversityKowloonHong Kong SAR999077China
- Research Centre of Deep Space Explorations (RCDSE)The Hong Kong Polytechnic UniversityKowloonHong Kong SAR999077China
| |
Collapse
|
35
|
Zheng X, Gomez-Rivas EJ, Lamont SI, Daneshjoo K, Shieh A, Wozniak DJ, Parsek MR. The surface interface and swimming motility influence surface-sensing responses in Pseudomonas aeruginosa. Proc Natl Acad Sci U S A 2024; 121:e2411981121. [PMID: 39284057 PMCID: PMC11441478 DOI: 10.1073/pnas.2411981121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/01/2024] [Indexed: 10/02/2024] Open
Abstract
Bacterial biofilms have been implicated in several chronic infections. After initial attachment, a critical first step in biofilm formation is a cell inducing a surface-sensing response. In the Gram-negative opportunistic pathogen Pseudomonas aeruginosa, two second messengers, cyclic diguanylate monophosphate (c-di-GMP) and cyclic adenosine monophosphate (cAMP), are produced by different surface-sensing mechanisms. However, given the disparate cellular behaviors regulated by these second messengers, how newly attached cells coordinate these pathways remains unclear. Some of the uncertainty relates to studies using different strains, experimental systems, and usually focusing on a single second messenger. In this study, we developed a tricolor reporter system to simultaneously gauge c-di-GMP and cAMP levels in single cells. Using PAO1, we show that c-di-GMP and cAMP are selectively activated in two commonly used experimental systems to study surface sensing. By further examining the conditions that differentiate a c-di-GMP or cAMP response, we demonstrate that an agarose-air interface activates cAMP signaling through type IV pili and the Pil-Chp system. However, a liquid-agarose interface favors the activation of c-di-GMP signaling. This response is dependent on flagellar motility and correlated with higher swimming speed. Collectively, this work indicates that c-di-GMP and cAMP signaling responses are dependent on the surface context.
Collapse
Affiliation(s)
- Xuhui Zheng
- Department of Microbiology, University of Washington, Seattle, WA
| | | | - Sabrina I. Lamont
- Departments of Microbial Infection and Immunity, Microbiology, The Ohio State University, Columbus, OH
| | | | - Angeli Shieh
- Department of Microbiology, University of Washington, Seattle, WA
| | - Daniel J. Wozniak
- Departments of Microbial Infection and Immunity, Microbiology, The Ohio State University, Columbus, OH
| | | |
Collapse
|
36
|
Cui Y, Wang D, Zhang L, Qu X. Research progress on the regulatory mechanism of biofilm formation in probiotic lactic acid bacteria. Crit Rev Food Sci Nutr 2024:1-15. [PMID: 39244761 DOI: 10.1080/10408398.2024.2400593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
Probiotic lactic acid bacteria (LAB) must undergo three key stages of testing, including food processing, storage, and gastrointestinal tract environment, their beneficial effects could exert. The biofilm formation of probiotic LAB is helpful for improving their stress resistances, survival rates, and colonization abilities under adverse environmental conditions, laying an important foundation for their probiotic effects. In this review, the formation process, the composition and function of basic components of probiotic LAB biofilm have been summarized. This review focuses on the regulatory mechanism of probiotic LAB biofilm formation. In addition, the characteristics and related mechanisms of probiotics in biofilm state have been analyzed to guide the application of probiotic LAB biofilms in the field of health and food. The biofilm formation of LAB is an extremely complex process involving multiple regulatory factors. Besides quorum sensing (QS), other regulatory factors are not yet fully understood. The probiotic LAB in biofilm state exhibit superior survival rate, adhesion performance, and immunomodulation ability, attribute to various metabolic processes, including stress response, exopolysaccharide (EPS) metabolism, amino acid and protein metabolisms, etc. The understanding about regulatory mechanism of biofilm formation of different probiotic species and strains will accelerate the development and application of probiotics products.
Collapse
Affiliation(s)
- Yanhua Cui
- Department of Food Nutrition and Health, School of Medicine and Health, Harbin Institute of Technology, Harbin, China
| | - Dongqi Wang
- Department of Food Nutrition and Health, School of Medicine and Health, Harbin Institute of Technology, Harbin, China
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Xiaojun Qu
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin, China
| |
Collapse
|
37
|
Liu YS, Zhang C, Khoo BL, Hao P, Chua SL. Dual-species proteomics and targeted intervention of animal-pathogen interactions. J Adv Res 2024:S2090-1232(24)00383-7. [PMID: 39233003 DOI: 10.1016/j.jare.2024.08.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024] Open
Abstract
INTRODUCTION Host-microbe interactions are important to human health and ecosystems globally, so elucidating the complex host-microbe interactions and associated protein expressions drives the need to develop sensitive and accurate biochemical techniques. Current proteomics techniques reveal information from the point of view of either the host or microbe, but do not provide data on the corresponding partner. Moreover, it remains challenging to simultaneously study host-microbe proteomes that reflect the direct competition between host and microbe. This raises the need to develop a dual-species proteomics method for host-microbe interactions. OBJECTIVES We aim to establish a forward + reverse Stable Isotope Labeling with Amino acids in Cell culture (SILAC) proteomics approach to simultaneously label and quantify newly-expressed proteins of host and microbe without physical isolation, for investigating mechanisms in direct host-microbe interactions. METHODS Using Caenorhabditis elegans-Pseudomonas aeruginosa infection model as proof-of-concept, we employed SILAC proteomics and molecular pathway analysis to characterize the differentially-expressed microbial and host proteins. We then used molecular docking and chemical characterization to identify chemical inhibitors that intercept host-microbe interactions and eliminate microbial infection. RESULTS Based on our proteomics results, we studied the iron competition between pathogen iron scavenger and host iron uptake protein, where P. aeruginosa upregulated pyoverdine synthesis protein (PvdA) (fold-change of 5.2313) and secreted pyoverdine, and C. elegans expressed ferritin (FTN-2) (fold-change of 3.4057). Targeted intervention of iron competition was achieved using Galangin, a ginger-derived phytochemical that inhibited pyoverdine production and biofilm formation in P. aeruginosa. The Galangin-ciprofloxacin combinatorial therapy could eliminate P. aeruginosa biofilms in a fish wound infection model, and enabled animal survival. CONCLUSION Our work provides a novel SILAC-based proteomics method that can simultaneously evaluate host and microbe proteomes, with future applications in higher host organisms and other microbial species. It also provides insights into the mechanisms dictating host-microbe interactions, offering novel strategies for anti-infective therapy.
Collapse
Affiliation(s)
- Yang Sylvia Liu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region
| | - Chengqian Zhang
- School of Life Science and Technology, ShanghaiTech University, China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region; Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong Special Administrative Region; City University of Hong Kong-Shenzhen Futian Research Institute, Shenzhen, China
| | - Piliang Hao
- School of Life Science and Technology, ShanghaiTech University, China.
| | - Song Lin Chua
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region; State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region; Research Centre for Deep Space Explorations (RCDSE), The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region.
| |
Collapse
|
38
|
Mnisi TJ, Matotoka MM, Masoko P. Antioxidant, antibacterial, and anti-biofilm activities of selected indigenous plant species against nosocomial bacterial pathogens. Lett Appl Microbiol 2024; 77:ovae080. [PMID: 39198017 DOI: 10.1093/lambio/ovae080] [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: 12/19/2023] [Revised: 07/26/2024] [Accepted: 08/27/2024] [Indexed: 09/01/2024]
Abstract
Biofilms are responsible for over 60% of nosocomial infections. The focus of this study was to investigate the antioxidant, antibacterial, antibiofilm, and anti-motility activities of Gardenia volkensii, Carissa bispinosa, Peltophorum africanum, and Senna petersiana. Antioxidant activity was evaluated using free radical (DPPH) scavenging and ferric reducing power assays. Antibacterial and antibiofilm activities were evaluated using the broth micro-dilution and the crystal violet assays, respectively. Anti-motility was evaluated using anti-swarming activities, and the brine shrimp lethality assay was used for cytotoxicity. Gardenia volkensii and C. bispinosa acetone extracts had low EC50 values of 9.59 and 9.99 μg ml-1on the free-radical scavenging activity, respectively. All the plant extracts demonstrated broad-spectrum antibacterial activity against Klebsiella pneumoniae, Pseudomonasa aeruginosa, Escherichia coli, Enterococcus faecalis, and Staphylococcus aureus [minimum inhibitory concentration (MIC) < 0.63 mg ml-1]. The initial cell adherence stage of P. aeruginosa and E. coli was the most susceptible stage where sub-MICs resulted in inhibitions >50%. Peltophorum africanum had the least cytotoxic effects. All extracts had anti-motility activity against P. aeruginosa and E. coli. This study showed that not only do the plants have strong antibacterial activity but had noteworthy inhibition (>50%) of initial cell adherence and may be suitable candidates for the treatment of nosocomial pathogens.
Collapse
Affiliation(s)
- Talita J Mnisi
- Faculty of Science and Agriculture, Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X1106, Sovenga 0727, South Africa
| | - Mashilo M Matotoka
- Faculty of Science and Agriculture, Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X1106, Sovenga 0727, South Africa
| | - Peter Masoko
- Faculty of Science and Agriculture, Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X1106, Sovenga 0727, South Africa
| |
Collapse
|
39
|
Pham TT, Nguyen TD, Nguyen TT, Pham MN, Nguyen PT, Nguyen TUT, Huynh TTN, Nguyen HT. Rhizosphere bacterial exopolysaccharides: composition, biosynthesis, and their potential applications. Arch Microbiol 2024; 206:388. [PMID: 39196410 DOI: 10.1007/s00203-024-04113-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 08/29/2024]
Abstract
Bacterial exopolysaccharides (EPS) are biopolymers of carbohydrates, often released from cells into the extracellular environment. Due to their distinctive physicochemical properties, biocompatibility, biodegradability, and non-toxicity, EPS finds applications in various industrial sectors. However, the need for alternative EPS has grown over the past few decades as lactic acid bacteria's (LAB) low-yield EPS is unable to meet the demand. In this case, rhizosphere bacteria with the diverse communities in soil leading to variations in composition and structure, are recognized as a potential source of EPS applicable in various industries. In addition, media components and cultivation conditions have an impact on EPS production, which ultimately affects the quantity, structure, and biological functions of the EPS. Therefore, scientists are currently working on manipulating bacterial EPS by developing cultures and applying abiotic and biotic stresses, so that better production of exopolysaccharides can be attained. This review highlights the composition, biosynthesis, and effects of environmental factors on EPS production along with the potential applications in different fields of industry. Ultimately, an overview of potential future paths and tactics for improving EPS implementation and commercialization is pointed out.
Collapse
Affiliation(s)
| | | | - Thi-Tho Nguyen
- Hutech Institute of Applied Science, HUTECH University, Ho Chi Minh City, Vietnam.
| | - Minh-Nhut Pham
- Hutech Institute of Applied Science, HUTECH University, Ho Chi Minh City, Vietnam
| | - Phu-Tho Nguyen
- An Giang University, An Giang, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - To-Uyen Thi Nguyen
- Graduate University of Sciences and Technology, Vietnam Academy of Science and Technology, Ha Noi, Vietnam
| | | | - Huu-Thanh Nguyen
- An Giang University, An Giang, Vietnam.
- Vietnam National University, Ho Chi Minh City, Vietnam.
| |
Collapse
|
40
|
Leinweber A, Laffont C, Lardi M, Eberl L, Pessi G, Kümmerli R. RNA-Seq reveals that Pseudomonas aeruginosa mounts growth medium-dependent competitive responses when sensing diffusible cues from Burkholderia cenocepacia. Commun Biol 2024; 7:995. [PMID: 39143311 PMCID: PMC11324955 DOI: 10.1038/s42003-024-06618-3] [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: 05/05/2023] [Accepted: 07/23/2024] [Indexed: 08/16/2024] Open
Abstract
Most habitats host diverse bacterial communities, offering opportunities for inter-species interactions. While competition might often dominate such interactions, little is known about whether bacteria can sense competitors and mount adequate responses. The competition sensing hypothesis proposes that bacteria can use cues such as nutrient stress and cell damage to prepare for battle. Here, we tested this hypothesis by measuring transcriptome changes in Pseudomonas aeruginosa exposed to the supernatant of its competitor Burkholderia cenocepacia. We found that P. aeruginosa exhibited significant growth-medium-dependent transcriptome changes in response to competition. In an iron-rich medium, P. aeruginosa upregulated genes encoding the type-VI secretion system and the siderophore pyoverdine, whereas genes encoding phenazine toxins and hydrogen cyanide were upregulated under iron-limited conditions. Moreover, general stress response and quorum sensing regulators were upregulated upon supernatant exposure. Altogether, our results reveal nuanced competitive responses of P. aeruginosa when confronted with B. cenocepacia supernatant, integrating both environmental and social cues.
Collapse
Affiliation(s)
- Anne Leinweber
- Department of Plant and Microbial Biology, University of Zurich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Clémentine Laffont
- Department of Quantitative Biomedicine, University of Zurich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.
| | - Martina Lardi
- Department of Plant and Microbial Biology, University of Zurich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Leo Eberl
- Department of Plant and Microbial Biology, University of Zurich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Gabriella Pessi
- Department of Plant and Microbial Biology, University of Zurich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Rolf Kümmerli
- Department of Plant and Microbial Biology, University of Zurich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.
- Department of Quantitative Biomedicine, University of Zurich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.
| |
Collapse
|
41
|
Ronish LA, Biswas B, Bauer RM, Jacob ME, Piepenbrink KH. The role of extracellular structures in Clostridioides difficile biofilm formation. Anaerobe 2024; 88:102873. [PMID: 38844261 DOI: 10.1016/j.anaerobe.2024.102873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/27/2024] [Accepted: 06/03/2024] [Indexed: 07/08/2024]
Abstract
C. difficile infection (CDI) is a costly and increasing burden on the healthcare systems of many developed countries due to the high rates of nosocomial infections. Despite the availability of several antibiotics with high response rates, effective treatment is hampered by recurrent infections. One potential mechanism for recurrence is the existence of C. difficile biofilms in the gut which persist through the course of antibiotics. In this review, we describe current developments in understanding the molecular mechanisms by which C. difficile biofilms form and are stabilized through extracellular biomolecular interactions.
Collapse
Affiliation(s)
- Leslie A Ronish
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Baishakhi Biswas
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Robert M Bauer
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Mallory E Jacob
- Biochemistry Department, University of Geneva, Geneva, Switzerland
| | - Kurt H Piepenbrink
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA; Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA; Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA; Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA; Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA.
| |
Collapse
|
42
|
Ojha R, Krug S, Jones P, Koestler BJ. Intact and mutated Shigella diguanylate cyclases increase c-di-GMP. J Biol Chem 2024; 300:107525. [PMID: 38960033 PMCID: PMC11327459 DOI: 10.1016/j.jbc.2024.107525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/31/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
The intracellular human pathogen Shigella invades the colonic epithelium to cause disease. Prior to invasion, this bacterium navigates through different environments within the human body, including the stomach and the small intestine. To adapt to changing environments, Shigella uses the bacterial second messenger cyclic di-GMP (c di-GMP) signaling system, synthesized by diguanylate cyclases (DGCs) encoding GGDEF domains. Shigella flexneri encodes a total of 9 GGDEF or GGDEF-EAL domain enzymes in its genome, but five of these genes have acquired mutations that presumably inactivated the c-di-GMP synthesis activity of these enzymes. In this study, we examined individual S. flexneri DGCs for their role in c-di-GMP synthesis and pathogenesis. We individually expressed each of the four intact DGCs in a S. flexneri strain, where these four DGCs had been deleted (Δ4DGC). We found that the 4 S. flexneri intact DGCs synthesize c-di-GMP at different levels in vitro and during infection of tissue-cultured cells. We also found that dgcF and dgcI expression significantly reduces invasion and plaque formation, and dgcF expression increases acid sensitivity, and that these phenotypes did not correspond with measured c-di-GMP levels. However, deletion of these four DGCs did not eliminate S. flexneri c-di-GMP, and we found that dgcE, dgcQ, and dgcN, which all have nonsense mutations prior to the GGDEF domain, still produce c-di-GMP. These S. flexneri degenerate DGC pseudogenes are expressed as multiple proteins, consistent with multiple start codons within the gene. We propose that both intact and degenerate DGCs contribute to S. flexneri c-di-GMP signaling.
Collapse
Affiliation(s)
- Ruchi Ojha
- Department of Biological Sciences, Western Michigan University, Kalamazoo, Michigan, USA
| | - Stefanie Krug
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Prentiss Jones
- Department of Pathology, Western Michigan University Homer Stryker, M.D. School of Medicine, Kalamazoo, Michigan, USA
| | - Benjamin J Koestler
- Department of Biological Sciences, Western Michigan University, Kalamazoo, Michigan, USA.
| |
Collapse
|
43
|
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
|
44
|
Schneier A, Melaugh G, Sadler JC. Engineered plastic-associated bacteria for biodegradation and bioremediation. BIOTECHNOLOGY FOR THE ENVIRONMENT 2024; 1:7. [PMID: 39026535 PMCID: PMC11256910 DOI: 10.1186/s44314-024-00007-0] [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: 02/26/2024] [Accepted: 04/29/2024] [Indexed: 07/20/2024]
Abstract
The global plastic waste crisis has triggered the development of novel methods for removal of recalcitrant polymers from the environment. Biotechnological approaches have received particular attention due to their potential for enabling sustainable, low-intensity bioprocesses which could also be interfaced with microbial upcycling pathways to support the emerging circular bioeconomy. However, low biodegradation efficiency of solid plastic materials remains a bottleneck, especially at mesophilic conditions required for one-pot degradation and upcycling. A promising strategy used in nature to address this is localisation of plastic-degrading microbes to the plastic surface via biofilm-mediated surface association. This review highlights progress and opportunities in leveraging these naturally occurring mechanisms of biofilm formation and other cell-surface adhesion biotechnologies to co-localise engineered cells to plastic surfaces. We further discuss examples of combining these approaches with extracellular expression of plastic-degrading enzymes to accelerate plastic degradation. Additionally, we review this topic in the context of nano- and microplastics bioremediation and their removal from wastewater and finally propose future research directions for this nascent field.
Collapse
Affiliation(s)
- Arianna Schneier
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Roger Land Building, Alexander Crum Brown Road, King’s Buildings, Edinburgh, EH9 3FF UK
| | - Gavin Melaugh
- School of Physics and Astronomy, University of Edinburgh, Edinburgh, EH9 3FD UK
- School of Engineering, University of Edinburgh, Edinburgh, EH9 3JL UK
| | - Joanna C. Sadler
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Roger Land Building, Alexander Crum Brown Road, King’s Buildings, Edinburgh, EH9 3FF UK
| |
Collapse
|
45
|
Zhang Y, Zhao X, Wang J, Liao L, Qin H, Zhang R, Li C, He Y, Huang S. VmsR, a LuxR-Type Regulator, Contributes to Virulence, Cell Motility, Extracellular Polysaccharide Production and Biofilm Formation in Xanthomonas oryzae pv. oryzicola. Int J Mol Sci 2024; 25:7595. [PMID: 39062838 PMCID: PMC11277528 DOI: 10.3390/ijms25147595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
LuxR-type regulators play pivotal roles in regulating numerous bacterial processes, including bacterial motility and virulence, thereby exerting a significant influence on bacterial behavior and pathogenicity. Xanthomonas oryzae pv. oryzicola, a rice pathogen, causes bacterial leaf streak. Our research has identified VmsR, which is a response regulator of the two-component system (TCS) that belongs to the LuxR family. These findings of the experiment reveal that VmsR plays a crucial role in regulating pathogenicity, motility, biofilm formation, and the production of extracellular polysaccharides (EPSs) in Xoc GX01. Notably, our study shows that the vmsR mutant exhibits a reduced swimming motility but an enhanced swarming motility. Furthermore, this mutant displays decreased virulence while significantly increasing EPS production and biofilm formation. We have uncovered that VmsR directly interacts with the promoter regions of fliC and fliS, promoting their expression. In contrast, VmsR specifically binds to the promoter of gumB, resulting in its downregulation. These findings indicate that the knockout of vmsR has profound effects on virulence, motility, biofilm formation, and EPS production in Xoc GX01, providing insights into the intricate regulatory network of Xoc.
Collapse
Affiliation(s)
- Yaqi Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Xiyao Zhao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Jiuxiang Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Lindong Liao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Huajun Qin
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Rongbo Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Changyu Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Yongqiang He
- College of Agronomy, Guangxi University, Nanning 530004, China
| | - Sheng Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| |
Collapse
|
46
|
Elbediwi M, Rolff J. Metabolic pathways and antimicrobial peptide resistance in bacteria. J Antimicrob Chemother 2024; 79:1473-1483. [PMID: 38742645 DOI: 10.1093/jac/dkae128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024] Open
Abstract
Antimicrobial resistance is a pressing concern that poses a significant threat to global public health, necessitating the exploration of alternative strategies to combat drug-resistant microbial infections. Recently, antimicrobial peptides (AMPs) have gained substantial attention as possible replacements for conventional antibiotics. Because of their pharmacodynamics and killing mechanisms, AMPs display a lower risk of bacterial resistance evolution compared with most conventional antibiotics. However, bacteria display different mechanisms to resist AMPs, and the role of metabolic pathways in the resistance mechanism is not fully understood. This review examines the intricate relationship between metabolic genes and AMP resistance, focusing on the impact of metabolic pathways on various aspects of resistance. Metabolic pathways related to guanosine pentaphosphate (pppGpp) and guanosine tetraphosphate (ppGpp) [collectively (p)ppGpp], the tricarboxylic acid (TCA) cycle, haem biosynthesis, purine and pyrimidine biosynthesis, and amino acid and lipid metabolism influence in different ways metabolic adjustments, biofilm formation and energy production that could be involved in AMP resistance. By targeting metabolic pathways and their associated genes, it could be possible to enhance the efficacy of existing antimicrobial therapies and overcome the challenges exhibited by phenotypic (recalcitrance) and genetic resistance toward AMPs. Further research in this area is needed to provide valuable insights into specific mechanisms, uncover novel therapeutic targets, and aid in the fight against antimicrobial resistance.
Collapse
Affiliation(s)
- Mohammed Elbediwi
- Evolutionary Biology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
- Animal Health Research Institute, Agriculture Research Centre, 12618 Cairo, Egypt
| | - Jens Rolff
- Evolutionary Biology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
- Berlin-Brandenburg Institute of Advanced Biodiversity Research (BBIB), Berlin, Germany
| |
Collapse
|
47
|
Zhu W, Liu J, Zhang Y, Zhao D, Li S, Dou H, Wang H, Xia X. The role of rcpA gene in regulating biofilm formation and virulence in Vibrio parahaemolyticus. Int J Food Microbiol 2024; 418:110714. [PMID: 38677238 DOI: 10.1016/j.ijfoodmicro.2024.110714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 04/14/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
Vibrio parahaemolyticus (V. parahaemolyticus) is a common seafood-borne pathogen that can colonize the intestine of host and cause gastroenteritis. Biofilm formation by V. parahaemolyticus enhances its persistence in various environments, which poses a series of threats to food safety. This work aims to investigate the function of rcpA gene in biofilm formation and virulence of V. parahaemolyticus. Deletion of rcpA significantly reduced motility, biofilm biomass, and extracellular polymeric substances, and inhibited biofilm formation on a variety of food and food contact surfaces. In mice infection model, mice infected with ∆rcpA strain exhibited a decreased rate of pathogen colonization, a lower level of inflammatory cytokines, and less tissue damage when compared to mice infected with wild type strain. RNA-seq analysis revealed that 374 genes were differentially expressed in the rcpA deletion mutant, which include genes related to quorum sensing, flagellar system, ribosome, type VI secretion system, biotin metabolism and transcriptional regulation. In conclusion, rcpA plays a role in determining biofilm formation and virulence of V. parahaemolyticus and further research is necessitated to fully understand its function in V. parahaemolyticus.
Collapse
Affiliation(s)
- Wenxiu Zhu
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Jiaxiu Liu
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Yingying Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Dongyun Zhao
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Shugang Li
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Hanzheng Dou
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Haisong Wang
- Liaoning Key Lab of Lignocellulose Chemistry and BioMaterials, Liaoning Collaborative Innovation Center for Lignocellulosic Biorefinery, College of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Xiaodong Xia
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
| |
Collapse
|
48
|
Zhao Y, Zhang J, Ni M, Pan Y, Li L, Ding Y. Cultivation of phosphate-accumulating biofilm: Study of the effects of acyl-homoserine lactones (AHLs) and cyclic dimeric guanosine monophosphate (c-di-GMP) on the formation of biofilm and the enhancement of phosphate metabolism capacity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 928:172408. [PMID: 38608880 DOI: 10.1016/j.scitotenv.2024.172408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
This study investigated the mechanisms of microbial growth and metabolism during biofilm cultivation in the biofilm sequencing batch reactor (BSBR) process for phosphate (P) enrichment. The results showed that the sludge discharge was key to biofilm growth, as it terminated the competition for carbon (C) source between the nascent biofilm and the activated sludge. For the tested reactor, after the sludge discharge on 18 d, P metabolism and C source utilization improved significantly, and the biofilm grew rapidly. The P concentration of the recovery liquid reached up to 157.08 mg/L, which was sufficient for further P recovery via mineralization. Meta-omics methods were used to analyze metabolic pathways and functional genes in microbial growth during biofilm cultivation. It appeared that the sludge discharge activated the key genes of P metabolism and inhibited the key genes of C metabolism, which strengthened the polyphosphate-accumulating metabolism (PAM) as a result. The sludge discharge not only changed the types of polyphosphate-accumulating organisms (PAOs) but also promoted the growth of dominant PAOs. Before the sludge discharge, the necessary metabolic abilities that were spread among different microorganisms gradually concentrated into a small number of PAOs, and after the sludge discharge, they further concentrated into Candidatus_Contendobacter (P3) and Candidatus_Accumulibacter (P17). The messenger molecule C-di-GMP, produced mostly by P3 and P17, facilitated P enrichment by regulating cellular P and C metabolism. The glycogen-accumulating organism (GAO) Candidatus_Competibacter secreted N-Acyl homoserine lactones (AHLs), which stimulated the secretion of protein in extracellular polymeric substances (EPS), thus promoting the adhesion of microorganisms to biofilm and improving P metabolism via EPS-based P adsorption. Under the combined action of the dominant GAOs and PAOs, AHLs and C-di-GMP mediated QS to promote biofilm development and P enrichment. The research provides theoretical support for the cultivation of biofilm and its wider application.
Collapse
Affiliation(s)
- Yimeng Zhao
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Jun Zhang
- Suzhou Drainage Company Limited, Suzhou 215009, China
| | - Min Ni
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China; Jiangsu Key Laboratory of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Yang Pan
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China; National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, Suzhou University of Science and Technology, Suzhou 215009, China.
| | - Lu Li
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China; National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Yanyan Ding
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China; National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, Suzhou University of Science and Technology, Suzhou 215009, China
| |
Collapse
|
49
|
Vasenina A, Fu Y, O'Toole GA, Mucha PJ. Local control: a hub-based model for the c-di-GMP network. mSphere 2024; 9:e0017824. [PMID: 38591888 PMCID: PMC11237430 DOI: 10.1128/msphere.00178-24] [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: 04/10/2024] Open
Abstract
The genome of Pseudomonas fluorescens encodes >50 proteins predicted to play a role in bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP)-mediated biofilm formation. We built a network representation of protein-protein interactions and extracted key information via multidimensional scaling (i.e., principal component analysis) of node centrality measures, which measure features of proteins in a network. Proteins of different domain types (diguanylate cyclase, dual domain, phosphodiesterase, PilZ) exhibit unique network behavior and can be accurately classified by their network centrality values (i.e., roles in the network). The predictive power of protein-protein interactions in biofilm formation indicates the possibility of localized pools of c-di-GMP. A regression model showed a statistically significant impact of protein-protein interactions on the extent of biofilm formation in various environments. These results highlight the importance of a localized c-di-GMP signaling, extend our understanding of signaling by this second messenger beyond the current "Bow-tie Model," support a newly proposed "Hub Model," and suggest future avenues of investigation.
Collapse
Affiliation(s)
- Anna Vasenina
- Department of Mathematics, Dartmouth College, Hanover, New Hampshire, USA
| | - Yu Fu
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - George A. O'Toole
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Peter J. Mucha
- Department of Mathematics, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
50
|
McCaughey C, Trebino MA, McAtamney A, Isenberg RY, Mandel MJ, Yildiz FH, Sanchez LM. A Label-Free Approach for Relative Spatial Quantitation of c-di-GMP in Microbial Biofilms. Anal Chem 2024; 96:8308-8316. [PMID: 38752543 PMCID: PMC11140670 DOI: 10.1021/acs.analchem.3c04687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/27/2024] [Accepted: 05/07/2024] [Indexed: 05/19/2024]
Abstract
Microbial biofilms represent an important lifestyle for bacteria and are dynamic three-dimensional structures. Cyclic dimeric guanosine monophosphate (c-di-GMP) is a ubiquitous signaling molecule that is known to be tightly regulated with biofilm processes. While measurements of global levels of c-di-GMP have proven valuable toward understanding the genetic control of c-di-GMP production, there is a need for tools to observe the local changes of c-di-GMP production in biofilm processes. We have developed a label-free method for the direct detection of c-di-GMP in microbial colony biofilms using matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI). We applied this method to the enteric pathogen Vibrio cholerae, the marine symbiont V. fischeri, and the opportunistic pathogen Pseudomonas aeruginosa PA14 and detected spatial and temporal changes in c-di-GMP signal that accompanied genetic alterations in factors that synthesize and degrade the compound. We further demonstrated how this method can be simultaneously applied to detect additional metabolites of interest from a single sample.
Collapse
Affiliation(s)
- Catherine
S. McCaughey
- Department
of Chemistry and Biochemistry, University
of California, Santa Cruz, Santa Cruz, California 95064, United States
| | - Michael A. Trebino
- Department
of Microbiology and Environmental Toxicology, University of California, Santa Cruz, Santa Cruz, California 95064, United States
| | - Allyson McAtamney
- Department
of Chemistry and Biochemistry, University
of California, Santa Cruz, Santa Cruz, California 95064, United States
| | - Ruth Y. Isenberg
- Department
of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
- Microbiology
Doctoral Training Program, University of
Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Mark J. Mandel
- Department
of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
- Microbiology
Doctoral Training Program, University of
Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Fitnat H. Yildiz
- Department
of Microbiology and Environmental Toxicology, University of California, Santa Cruz, Santa Cruz, California 95064, United States
| | - Laura M. Sanchez
- Department
of Chemistry and Biochemistry, University
of California, Santa Cruz, Santa Cruz, California 95064, United States
| |
Collapse
|