1
|
Dai Y, Liu R, Yue Y, Song N, Jia H, Ma Z, Gao X, Zhang M, Yuan X, Liu Q, Liu X, Li B, Wang W. A c-di-GMP binding effector STM0435 modulates flagellar motility and pathogenicity in Salmonella. Virulence 2024; 15:2331265. [PMID: 38532247 PMCID: PMC10978029 DOI: 10.1080/21505594.2024.2331265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Flagella play a crucial role in the invasion process of Salmonella and function as a significant antigen that triggers host pyroptosis. Regulation of flagellar biogenesis is essential for both pathogenicity and immune escape of Salmonella. We identified the conserved and unknown function protein STM0435 as a new flagellar regulator. The ∆stm0435 strain exhibited higher pathogenicity in both cellular and animal infection experiments than the wild-type Salmonella. Proteomic and transcriptomic analyses demonstrated dramatic increases in almost all flagellar genes in the ∆stm0435 strain compared to wild-type Salmonella. In a surface plasmon resonance assay, purified STM0435 protein-bound c-di-GMP had an affinity of ~8.383 µM. The crystal structures of apo-STM0435 and STM0435&c-di-GMP complex were determined. Structural analysis revealed that R33, R137, and D138 of STM0435 were essential for c-di-GMP binding. A Salmonella with STM1987 (GGDEF protein) or STM4264 (EAL protein) overexpression exhibits completely different motility behaviours, indicating that the binding of c-di-GMP to STM0435 promotes its inhibitory effect on Salmonella flagellar biogenesis.
Collapse
Affiliation(s)
- Yuanji Dai
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ruirui Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yingying Yue
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Nannan Song
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Haihong Jia
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Zhongrui Ma
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xueyan Gao
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Min Zhang
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xilu Yuan
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Qing Liu
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaoyu Liu
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Bingqing Li
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Key Lab for Biotech-Drugs of National Health Commission, Shandong First Medical University, Jinan, Shandong, China
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong, China
| | - Weiwei Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
2
|
Tătulea-Codrean M, Lauga E. Physical mechanism reveals bacterial slowdown above a critical number of flagella. J R Soc Interface 2024; 21:20240283. [PMID: 39503268 PMCID: PMC11539103 DOI: 10.1098/rsif.2024.0283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/30/2024] [Accepted: 09/18/2024] [Indexed: 11/09/2024] Open
Abstract
Numerous studies have explored the link between bacterial swimming and the number of flagella, a distinguishing feature of motile multi-flagellated bacteria. We revisit this open question using augmented slender-body theory simulations, in which we resolve the full hydrodynamic interactions within a bundle of helical filaments rotating and translating in synchrony. Unlike previous studies, our model considers the full torque-speed relationship of the bacterial flagellar motor, revealing its significant impact on multi-flagellated swimming. Because the viscous load per motor decreases with the flagellar number, the bacterial flagellar motor transitions from the high-load to the low-load regime at a critical number of filaments, leading to bacterial slowdown as further flagella are added to the bundle. We explain the physical mechanism behind the observed slowdown as an interplay between the load-dependent generation of torque by the motor, and the load-reducing cooperativity between flagella, which consists of both hydrodynamic and non-hydrodynamic components. The theoretically predicted critical number of flagella is remarkably close to the values reported for the model organism Escherichia coli. Our model further predicts that the critical number of flagella increases with viscosity, suggesting that bacteria can enhance their swimming capacity by growing more flagella in more viscous environments, consistent with empirical observations.
Collapse
Affiliation(s)
- Maria Tătulea-Codrean
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, CambridgeCB3 0WA, UK
| | - Eric Lauga
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, CambridgeCB3 0WA, UK
| |
Collapse
|
3
|
Santos R, Mateus C, Oleastro M, Ferreira S. Exploring flagellar contributions to motility and virulence in Arcobacter butzleri. World J Microbiol Biotechnol 2024; 40:367. [PMID: 39455472 DOI: 10.1007/s11274-024-04175-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
Flagella is a well-known bacterial structure crucial for motility, which also plays pivotal roles in pathogenesis. Arcobacter butzleri, an enteropathogen, possesses a distinctive polar flagellum whose functional aspects remain largely unexplored. Upon investigating the factors influencing A. butzleri motility, we uncovered that environmental conditions like temperature, oxygen levels, and nutrient availability play a significant role. Furthermore, compounds that are found in human gut, such as short-chain fatty acids, mucins and bile salts, have a role in modulating the motility, and in turn, the pathogenicity of A. butzleri. Further investigation demonstrated that A. butzleri ΔflaA mutant showed a reduction in motility with a close to null average velocity, as well as a reduction on biofilm formation. In addition, compared with the wild-type, the ΔflaA mutant showed a decreased ability to invade Caco-2 cells and to adhere to mucins. Taken together, our findings support the role of environmental conditions and gut host associated compounds influencing key physiological aspects of the gastrointestinal pathogen A. butzleri, such as motility, and support the role of the flagellum on bacterial virulence.
Collapse
Affiliation(s)
- Raquel Santos
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Avenida Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Cristiana Mateus
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Avenida Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Mónica Oleastro
- National Reference Laboratory for Gastrointestinal Infections, Department of Infectious Diseases, National Institute of Health Dr. Ricardo Jorge, Lisbon, Portugal
| | - Susana Ferreira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Avenida Infante D. Henrique, Covilhã, 6200-506, Portugal.
| |
Collapse
|
4
|
Zhang X, Chen Y, Liu Y, Gang L, Yan T, Wang H, Peng Q, Li J, Song F. A novel regulator CdsR negatively regulates cell motility in Bacillus thuringiensis. Sci Rep 2024; 14:25270. [PMID: 39455735 PMCID: PMC11511885 DOI: 10.1038/s41598-024-76694-2] [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: 06/18/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Cell motility increases the fitness of bacterial cells. Previous research focused on the transcriptional regulator CdsR, which represses cellular autolysis and promotes spore formation in Bacillus thuringiensis. However, the targets of CdsR are mostly unknown. Here, we reported a new function of CdsR in regulating cell motility. Mutation of cdsR results in increase of cell mobility, and a number of related genes were upregulated compared to wild type HD73. Thus, we investigated the transcription of the fla/che gene cluster, which involves in cell mobility and comprises eight operons/genes, including motAB1, cheY-yrhK, lamB-cheR, yaaR-fliG2, cheV-mogR, hag1, hag2, and yjbJ-flgG. Additionally, the motAB2 operon was discovered, which consists of homologs genes motA2 and motB2 that are like motA1 and motB1. Through promoter-lacZ fusion assays and EMSA experiments, it was discovered that CdsR directly regulates the motAB1, cheY-yrhK, lamB-cheR, yaaR-fliG2, cheV-mogR, hag1, hag2, yjbJ-flgG, and motAB2 operons by binding to their promoter regions. Importantly, it was confirmed that CdsR is a metalloregulator and the binding to promoter can be inhibited by Cu (II) ions. This research enhances our understanding of the regulation of cell mobility in B. thuringiensis.
Collapse
Affiliation(s)
- Xin Zhang
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yuhan Chen
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yabin Liu
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Lili Gang
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Tinglu Yan
- Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Hengjie Wang
- Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Qi Peng
- Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jie Li
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Fuping Song
- Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
5
|
Warrell DL, Zarrella TM, Machalek C, Khare A. Interspecies surfactants serve as public goods enabling surface motility in Pseudomonas aeruginosa. J Bacteriol 2024; 206:e0028124. [PMID: 39235232 PMCID: PMC11500613 DOI: 10.1128/jb.00281-24] [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: 07/09/2024] [Accepted: 08/02/2024] [Indexed: 09/06/2024] Open
Abstract
In most natural environments, bacteria live in polymicrobial communities where secreted molecules from neighboring species alter bacterial behaviors, including motility, but such interactions are understudied. Pseudomonas aeruginosa is a motile opportunistic pathogen that exists in diverse multispecies environments, such as the soil, and is frequently found in human wound and respiratory tract co-infections with other bacteria, including Staphylococcus aureus. Here, we show that P. aeruginosa can co-opt secreted surfactants from other species for flagellar-based surface motility. We found that exogenous surfactants from S. aureus, other bacteria, and interkingdom species enabled P. aeruginosa to switch from swarming to an alternative surface spreading motility on semi-solid surfaces and allowed for the emergence of surface motility on hard agar where P. aeruginosa was otherwise unable to move. Although active flagellar function was required for surface spreading, known motility regulators were not essential, indicating that surface spreading may be regulated by an as yet unknown mechanism. This motility was distinct from the response of most other motile bacterial species in the presence of exogenous surfactants. Mutant analysis indicated that this P. aeruginosa motility was similar to a previously described mucin-based motility, "surfing," albeit with divergent regulation. Thus, our study demonstrates that secreted surfactants from the host as well as neighboring bacterial and interkingdom species act as public goods facilitating P. aeruginosa flagella-mediated surfing-like surface motility, thereby allowing it to access different environmental niches. IMPORTANCE Bacterial motility is an important determinant of bacterial fitness and pathogenesis, allowing expansion and invasion to access nutrients and adapt to new environments. Here, we demonstrate that secreted surfactants from a variety of foreign species, including other bacterial species, infection hosts, fungi, and plants, facilitate surface spreading motility in the opportunistic pathogen Pseudomonas aeruginosa that is distinct from established motility phenotypes. This response to foreign surfactants also occurs in Pseudomonas putida, but not in more distantly related bacterial species. Our systematic characterization of surfactant-based surface spreading shows that these interspecies surfactants serve as public goods to enable P. aeruginosa to move and explore environmental conditions when it would be otherwise immotile.
Collapse
Affiliation(s)
- Delayna L. Warrell
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Tiffany M. Zarrella
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
- Postdoctoral Research Associate Training Program, National Institute of General Medical Sciences, National Institutes of Health, Bethesda, Maryland, USA
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Christopher Machalek
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Anupama Khare
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
6
|
Cheng G, Jian S, Li W, Yan L, Chen T, Cheng T, Liu Z, Ye G, Tang H, Zhang L. Epigallocatechin gallate protects mice from Salmonella enterica ser. Typhimurium infection by modulating bacterial virulence through quorum sensing inhibition. Front Cell Infect Microbiol 2024; 14:1432111. [PMID: 39479281 PMCID: PMC11521958 DOI: 10.3389/fcimb.2024.1432111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Salmonella enterica ser. Typhimurium is a common pathogen that poses a considerable public health threat, contributing to severe gastrointestinal diseases and widespread foodborne illnesses. The virulence of S. Typhimurium is regulated by quorum sensing (QS) and the type III secretion system (T3SS). This study investigated the inhibitory effects and anti-QS activity of epigallocatechin gallate (EGCG), which is a bioactive ingredient found in green tea, on the virulence of S. Typhimurium. In vitro bacterial experiments demonstrated that EGCG inhibited the production of autoinducers, biofilm formation, and flagellar activity by downregulating the expression of AI-1, AI-2, Salmonella pathogenicity islands (SPI)-1, SPI-2, and genes related to flagella, fimbriae, and curli fibers. In a mouse model of S. Typhimurium-induced enteritis, EGCG considerably reduced intestinal colonization by S. Typhimurium and alleviated intestinal damage. In conclusion, EGCG protects the intestines of mice infected with S. Typhimurium by inhibiting QS-induced virulence gene expression, demonstrating its potential as a therapeutic agent for controlling S. Typhimurium infections.
Collapse
Affiliation(s)
- Guoqiang Cheng
- Animal Experiment Center, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Shanqiu Jian
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Wen Li
- Department of Science, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Liangchun Yan
- Animal Experiment Center, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Tiezhu Chen
- Animal Experiment Center, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Tingting Cheng
- Animal Experiment Center, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Zongxiu Liu
- Department of Innovation, Chengdu Qiankun Animal Pharmaceutical Co., Ltd, Chengdu, China
| | - Gang Ye
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Huaqiao Tang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Li Zhang
- Animal Experiment Center, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| |
Collapse
|
7
|
Chen KZM, Vu LM, Vollmer AC. Cultivation in long-term simulated microgravity is detrimental to pyocyanin production and subsequent biofilm formation ability of Pseudomonas aeruginosa. Microbiol Spectr 2024; 12:e0021124. [PMID: 39162544 PMCID: PMC11448113 DOI: 10.1128/spectrum.00211-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/27/2024] [Indexed: 08/21/2024] Open
Abstract
Pseudomonas aeruginosa forms aggregates known as biofilms. Previous studies have shown that when P. aeruginosa is cultivated in space, thicker and structurally different biofilms are formed than from those grown on Earth. We investigated how microgravity, simulated in a laboratory setting, influenced the growth, colonization, and virulence potentials of a P. aeruginosa PA14 wild-type strain, as well as two surface attachment-defective (sad) mutants altered at crucial biofilm-forming steps: flgK and pelA. Using high-aspect ratio rotating-wall vessel (HARV) bioreactors, P. aeruginosa bacteria were grown to stationary phase under prolonged (6 days) exposure to simulated microgravity or normal gravity conditions. After the exposure, the capacity of the culture to form biofilms was measured. Additionally, pigment (pyocyanin) formed by each culture during the incubation was extracted and quantified. We demonstrate that the first prolonged exposure to low-shear modeled microgravity (LSMMG) and without nutrient replenishment significantly diminishes wild-type P. aeruginosa PA14 biofilm formation abilities after exposure and pyocyanin production during exposure, while the mutant strains exhibit differing outcomes for both properties. IMPORTANCE Given plans for humans to engage in prolonged space travel, we investigated biofilm and pigment/virulence factor formation in Pseudomonas aeruginosa when cultivated in microgravity. These bacteria are opportunistic pathogens in immunocompromised individuals. Previous studies of space travelers have shown some immune system diminutions. Hence, our studies shed some light on how prolonged cultivation of bacteria in simulated microgravity conditions affect their growth characteristics.
Collapse
Affiliation(s)
| | - Linda My Vu
- Department of Biology, Swarthmore College, Swarthmore, Pennsylvania, USA
- Department of Microbial Pathogenesis, University of Maryland—Baltimore, Baltimore, Maryland, USA
| | - Amy Cheng Vollmer
- Department of Biology, Swarthmore College, Swarthmore, Pennsylvania, USA
| |
Collapse
|
8
|
Moustafa DA, Fantone KM, Tucker SL, McCarty NA, Stecenko AA, Goldberg JB, Rada B. Flagellum-deficient Pseudomonas aeruginosa is more virulent than non-motile but flagellated mutants in a cystic fibrosis mouse model. Microbiol Spectr 2024; 12:e0132524. [PMID: 39248473 PMCID: PMC11448114 DOI: 10.1128/spectrum.01325-24] [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: 06/12/2024] [Accepted: 07/25/2024] [Indexed: 09/10/2024] Open
Abstract
Loss of the flagellum marks the pathoadaptation of Pseudomonas aeruginosa to the cystic fibrosis (CF) airway environment during lung disease. Losing the flagellum is advantageous to the bacterium as the flagellum can be recognized by immune cells. The primary purpose of the flagellum is, however, to provide motility to the bacterium. Our goal was to determine whether the loss of flagellar motility or the loss of flagellum expression contributes to P. aeruginosa lung infection in CF. To address this, wild-type and gut-corrected FABP-human cystic fibrosis transmembrane conductance regulator (hCFTR) mice deficient in the murine Cftr gene were infected intratracheally with lethal doses of wild-type or flagellum-deficient P. aeruginosa. While there was no significant difference in the survival of wild-type mice after infection with either of the bacterial strains, a significantly higher mortality was observed in FABP-hCFTR mice infected with flagellum-deficient P. aeruginosa, compared to mice infected with their flagellated counterparts. When FABP-hCFTR mice were infected with isogenic, motility-deficient flagellated mutants, animal survival and lung bacterial titers were similar to those observed in mice infected with the wild-type bacterium. Airway levels of neutrophils and the amount neutrophil elastase were similar in mice infected with either the wild-type bacteria or the flagellum-deficient P. aeruginosa. Our results show that FABP-hCFTR mice have a different response to flagellum loss in P. aeruginosa compared to wild-type animals. The loss of flagellum expression, rather than the loss of motility, is the main driver behind the increased virulence of flagellum-deficient P. aeruginosa in CF. These observations provide new insight into P. aeruginosa virulence in CF.IMPORTANCEPseudomonas aeruginosa, a major respiratory pathogen in cystic fibrosis, is known to lose its flagellum during the course of infection in the airways. Here, we show that the loss of flagellum leads to a more enhanced virulence in Cftr-deficient cystic fibrosis mice than in control animals. Loss of flagellum expression, rather than the loss of flagellar swimming motility, represents the main driver behind this increased virulence suggesting that this appendage plays a specific role in P. aeruginosa virulence in cystic fibrosis airways.
Collapse
Affiliation(s)
- Dina A. Moustafa
- Division of Pulmonology, Asthma, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kayla M. Fantone
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, Georgia, USA
| | - Samantha L. Tucker
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, Georgia, USA
| | - Nael A. McCarty
- Division of Pulmonology, Asthma, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Arlene A. Stecenko
- Division of Pulmonology, Asthma, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Joanna B. Goldberg
- Division of Pulmonology, Asthma, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, Georgia, USA
| |
Collapse
|
9
|
Roncarati D, Vannini A, Scarlato V. Temperature sensing and virulence regulation in pathogenic bacteria. Trends Microbiol 2024:S0966-842X(24)00180-X. [PMID: 39164134 DOI: 10.1016/j.tim.2024.07.009] [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: 06/06/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024]
Abstract
Pathogenic bacteria can detect a variety of environmental signals, including temperature changes. While sudden and significant temperature variations act as danger signals that trigger a protective heat-shock response, minor temperature fluctuations typically signal to the pathogen that it has moved from one environment to another, such as entering a specific niche within a host during infection. These latter temperature fluctuations are utilized by pathogens to coordinate the expression of crucial virulence factors. Here, we elucidate the critical role of temperature in governing the expression of virulence factors in bacterial pathogens. Moreover, we outline the molecular mechanisms used by pathogens to detect temperature fluctuations, focusing on systems that employ proteins and nucleic acids as sensory devices. We also discuss the potential implications and the extent of the risk that climate change poses to human pathogenic diseases.
Collapse
Affiliation(s)
- Davide Roncarati
- Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, Bologna, Italy.
| | - Andrea Vannini
- Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Vincenzo Scarlato
- Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, Bologna, Italy
| |
Collapse
|
10
|
He L, Mao M, Zhao L, Li Q, Zhuang Z, Wang X, Huang H, Wang Q, Yan Q. A novel small non-coding RNA 562 mediates the virulence of Pseudomonas plecoglossicida by regulating the expression of fliP, a key component of flagella T3SS. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109752. [PMID: 38977112 DOI: 10.1016/j.fsi.2024.109752] [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: 05/15/2024] [Revised: 06/19/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024]
Abstract
Pseudomonas plecoglossicida is a vital pathogen that poses a substantial risk to aquaculture. Small RNAs (sRNAs) are non-coding regulatory molecules capable of sensing environmental changes and modulating virulence-associated signaling pathways, such as the assembly of flagella. However, the relevant researches on P. plecoglossicida are an urgent need. Here, we report a novel sRNA, sRNA562, which has potential to regulate the post-transcriptional of fliP, a key component of the lateral flagellar type III secretion system. In this study, the effects of sRNA562 on the virulence of P. plecoglossicida and its role in regulating the pathogenic process were investigated through the use of a constructed sRNA562 deletion strain. The deletion of sRNA562 resulted in an up-regulation of fliP in P. plecoglossicida, and leading to increased swarming motility and enhanced the ability of biofilm formation, adhesion and chemotaxis. Subsequent artificial infection experiment demonstrated that the deletion of sRNA562 increased the virulence of P. plecoglossicida towards hybrid grouper, as evidenced by a reduction in survival rate, elevation of tissue bacterial load, and the exacerbation of histopathological damage. Further studies have found that the deletion of sRNA562 lead to an up-regulation of fliP expression during hybrid grouper infection, thereby enhancing bacterial swarming ability and ultimately heightening pathogenicity, leading to a dysregulated host response to infection, tissue damage and eventually death. Our work revealed a sRNA that exerts negative regulation on the expression of lateral flagella in P. plecoglossicida, thereby impacting its virulence. These findings provide a new perspective on the virulence regulation mechanism of P. plecoglossicida, contributing to a more comprehensive understanding in the field of pathogenicity research.
Collapse
Affiliation(s)
- Li He
- Fisheries College, Jimei University, Xiamen, Fujian, 361021, China
| | - Meiqin Mao
- Fisheries College, Jimei University, Xiamen, Fujian, 361021, China
| | - Lingmin Zhao
- Fisheries College, Jimei University, Xiamen, Fujian, 361021, China
| | - Qi Li
- Fisheries College, Jimei University, Xiamen, Fujian, 361021, China
| | - Zhixia Zhuang
- College of Environment and Public Health, Xiamen Huaxia University, Xiamen, Fujian 361024, China
| | - Xiaoru Wang
- College of Environment and Public Health, Xiamen Huaxia University, Xiamen, Fujian 361024, China
| | - Huabin Huang
- College of Environment and Public Health, Xiamen Huaxia University, Xiamen, Fujian 361024, China
| | - Qiyao Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China; Shanghai Haosi Marine Biotechnology Co., Ltd, Shanghai, 200000, China.
| | - Qingpi Yan
- Fisheries College, Jimei University, Xiamen, Fujian, 361021, China.
| |
Collapse
|
11
|
Konyshev IV, Byvalov AA. The bacterial flagellum as an object for optical trapping. Biophys Rev 2024; 16:403-415. [PMID: 39309130 PMCID: PMC11415335 DOI: 10.1007/s12551-024-01212-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 07/16/2024] [Indexed: 09/25/2024] Open
Abstract
This letter considers the possibility of using the optical trap to study the structure and function of the microbial flagellum. The structure of the flagellum of a typical gram-negative bacterium is described in brief. A standard mathematical model based on the principle of superposition is used to describe the movement of an ellipsoidal microbial cell in a liquid medium. The basic principles of optical trapping based on the combined action of the light pressure and the gradient force are briefly clarified. Several problems related to thermal damage of living microscopic objects when the latter gets to the focus of a laser beam are shortly discussed. It is shown that the probability of cell damage depends nonlinearly on the wavelength of laser radiation. Finally, the model systems that would make it possible to study flagella of the free bacteria and the ones anchored or tethered on the surface of a solid material are discussed in detail.
Collapse
Affiliation(s)
- Ilya V. Konyshev
- Institute of Physiology of the Federal Research Centre, Komi Science Centre, Ural Branch of the Russian Academy of Sciences, Syktyvkar, 167982 Russia
- Vyatka State University, Kirov, 610000 Russia
| | - Andrey A. Byvalov
- Institute of Physiology of the Federal Research Centre, Komi Science Centre, Ural Branch of the Russian Academy of Sciences, Syktyvkar, 167982 Russia
- Vyatka State University, Kirov, 610000 Russia
| |
Collapse
|
12
|
Mei Z, Wang F, Bhosle A, Dong D, Mehta R, Ghazi A, Zhang Y, Liu Y, Rinott E, Ma S, Rimm EB, Daviglus M, Willett WC, Knight R, Hu FB, Qi Q, Chan AT, Burk RD, Stampfer MJ, Shai I, Kaplan RC, Huttenhower C, Wang DD. Strain-specific gut microbial signatures in type 2 diabetes identified in a cross-cohort analysis of 8,117 metagenomes. Nat Med 2024; 30:2265-2276. [PMID: 38918632 DOI: 10.1038/s41591-024-03067-7] [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: 12/01/2023] [Accepted: 05/14/2024] [Indexed: 06/27/2024]
Abstract
The association of gut microbial features with type 2 diabetes (T2D) has been inconsistent due in part to the complexity of this disease and variation in study design. Even in cases in which individual microbial species have been associated with T2D, mechanisms have been unable to be attributed to these associations based on specific microbial strains. We conducted a comprehensive study of the T2D microbiome, analyzing 8,117 shotgun metagenomes from 10 cohorts of individuals with T2D, prediabetes, and normoglycemic status in the United States, Europe, Israel and China. Dysbiosis in 19 phylogenetically diverse species was associated with T2D (false discovery rate < 0.10), for example, enriched Clostridium bolteae and depleted Butyrivibrio crossotus. These microorganisms also contributed to community-level functional changes potentially underlying T2D pathogenesis, for example, perturbations in glucose metabolism. Our study identifies within-species phylogenetic diversity for strains of 27 species that explain inter-individual differences in T2D risk, such as Eubacterium rectale. In some cases, these were explained by strain-specific gene carriage, including loci involved in various mechanisms of horizontal gene transfer and novel biological processes underlying metabolic risk, for example, quorum sensing. In summary, our study provides robust cross-cohort microbial signatures in a strain-resolved manner and offers new mechanistic insights into T2D.
Collapse
Affiliation(s)
- Zhendong Mei
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Fenglei Wang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Amrisha Bhosle
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Danyue Dong
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Raaj Mehta
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Andrew Ghazi
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yancong Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yuxi Liu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ehud Rinott
- Department of Medicine, Hebrew University and Hadassah Medical Center, Jerusalem, Israel
| | - Siyuan Ma
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric B Rimm
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Martha Daviglus
- Institute for Minority Health Research, University of Illinois Chicago, Chicago, IL, USA
| | - Walter C Willett
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Rob Knight
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Frank B Hu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Qibin Qi
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Andrew T Chan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Robert D Burk
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Obstetrics, Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Meir J Stampfer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Iris Shai
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Faculty of Health Sciences, The Health and Nutrition Innovative International Research Center, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Curtis Huttenhower
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Dong D Wang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
13
|
Baztarrika I, Wösten MMSM, Alonso R, Martínez-Ballesteros I, Martinez-Malaxetxebarria I. Genes involved in the adhesion and invasion of Arcobacter butzleri. Microb Pathog 2024; 193:106752. [PMID: 38880315 DOI: 10.1016/j.micpath.2024.106752] [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: 12/17/2023] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Arcobacter butzleri is a foodborne pathogen that mainly causes enteritis in humans, but the number of cases of bacteraemia has increased in recent years. However, there is still limited knowledge on the pathogenic mechanisms of this bacterium. To investigate how A. butzleri causes disease, single knockout mutants were constructed in the cadF, ABU_RS00335, ciaB, and flaAB genes, which might be involved in adhesion and invasion properties. These mutants and the isogenic wild-type (WT) were then tested for their ability to adhere and invade human Caco-2 and HT29-MTX cells. The adhesion and invasion of A. butzleri RM4018 strain was also visualized by a Leica CTR 6500 confocal microscope. The adhesion and invasion abilities of mutants lacking the invasion antigen CiaB or a functional flagellum were lower than those of the WTs. However, the extent of the decrease varied depending on the strain and/or cell line. Mutants lacking the fibronectin (FN)-binding protein CadF consistently exhibited reduced abilities, while the inactivation of the other studied FN-binding protein, ABU_RS00335, led to a reduction in only one of the two strains tested. Therefore, the ciaB and flaAB genes appear to be important for A. butzleri adhesion and invasion properties, while cadF appears to be indispensable.
Collapse
Affiliation(s)
- Itsaso Baztarrika
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Álava, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Álava, Spain
| | - Marc M S M Wösten
- Department Biomolecular Health Sciences, Utrecht University, Yalelaan 1, 3584, CL, Utrecht, the Netherlands
| | - Rodrigo Alonso
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Álava, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Álava, Spain
| | - Ilargi Martínez-Ballesteros
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Álava, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Álava, Spain
| | - Irati Martinez-Malaxetxebarria
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Álava, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Álava, Spain.
| |
Collapse
|
14
|
Samgane G, Karaçam S, Tunçer Çağlayan S. Unveiling the synergistic potency of chlorhexidine and azithromycin in combined action. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5975-5987. [PMID: 38376540 PMCID: PMC11329591 DOI: 10.1007/s00210-024-03010-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
The growing challenge of antibiotic resistance necessitates novel approaches for combating bacterial infections. This study explores the distinctive synergy between chlorhexidine, an antiseptic and disinfectant agent, and azithromycin, a macrolide antibiotic, in their impact on bacterial growth and virulence factors using Escherichia coli strain Crooks (ATCC 8739) as a model. Our findings reveal that the chlorhexidine and azithromycin combination demonstrates enhanced anti-bacterial effects compared to individual treatments. Intriguingly, the combination induced oxidative stress, decreased flagellin expression, impaired bacterial motility, and enhanced bacterial autoaggregation. Notably, the combined treatment also demonstrated a substantial reduction in bacterial adherence to colon epithelial cells and downregulated NF-κB in the epithelial cells. In conclusion, these results shed light on the potential of the chlorhexidine and azithromycin synergy as a compelling strategy to address the rising challenge of antibiotic resistance and may pave the way for innovative therapeutic interventions in tackling bacterial infections.
Collapse
Affiliation(s)
- Gizem Samgane
- Department of Biotechnology, Bilecik Şeyh Edebali University, Bilecik, 11100, Turkey
| | - Sevinç Karaçam
- Department of Biotechnology, Bilecik Şeyh Edebali University, Bilecik, 11100, Turkey
- Central Research and Application Laboratory, Bilecik Şeyh Edebali University, Bilecik, 11100, Turkey
| | - Sinem Tunçer Çağlayan
- Department of Medical Services and Techniques, Vocational School of Health Services, Bilecik Şeyh Edebali University, Pelitözü Mah. Fatih Sultan Mehmet Bulvarı No:27, Bilecik, 11100, Turkey.
| |
Collapse
|
15
|
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
|
16
|
Liu YL, Chen XW, Tian SQ, Tan XH, Peng B. Edwardsiella tarda Attenuates Virulence upon Oxytetracycline Resistance. J Proteome Res 2024; 23:2576-2586. [PMID: 38860290 DOI: 10.1021/acs.jproteome.4c00303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
The relationship between antibiotic resistance and bacterial virulence has not yet been fully explored. Here, we use Edwardsiella tarda as the research model to investigate the proteomic change upon oxytetracycline resistance (LTB4-ROTC). Compared to oxytetracycline-sensitive E. tarda (LTB4-S), LTB4-ROTC has 234 differentially expressed proteins, of which the abundance of 84 proteins is downregulated and 15 proteins are enriched to the Type III secretion system, Type VI secretion system, and flagellum pathways. Functional analysis confirms virulent phenotypes, including autoaggregation, biofilm formation, hemolysis, swimming, and swarming, are impaired in LTB4-ROTC. Furthermore, the in vivo bacterial challenge in both tilapia and zebrafish infection models suggests that the virulence of LTB4-ROTC is attenuated. Analysis of immune gene expression shows that LTB4-ROTC induces a stronger immune response in the spleen but a weaker response in the head kidney than that induced by LTB4-S, suggesting it's a potential vaccine candidate. Zebrafish and tilapia were challenged with a sublethal dose of LTB4-ROTC as a live vaccine followed by LTB4-S challenge. The relative percentage of survival of zebrafish is 60% and that of tilapia is 75% after vaccination. Thus, our study suggests that bacteria that acquire antibiotic resistance may attenuate virulence, which can be explored as a potential live vaccine to tackle bacterial infection in aquaculture.
Collapse
Affiliation(s)
- Ying-Li Liu
- State Key Laboratory of Biocontrol, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Xuan-Wei Chen
- State Key Laboratory of Biocontrol, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Si-Qi Tian
- State Key Laboratory of Biocontrol, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Xiao-Hua Tan
- State Key Laboratory of Biocontrol, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Bo Peng
- State Key Laboratory of Biocontrol, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou 510275, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266071, China
| |
Collapse
|
17
|
Kulshrestha S, Narad P, Singh B, Pai SS, Vijayaraghavan P, Tandon A, Gupta P, Modi D, Sengupta A. Biomarker Identification for Preterm Birth Susceptibility: Vaginal Microbiome Meta-Analysis Using Systems Biology and Machine Learning Approaches. Am J Reprod Immunol 2024; 92:e13905. [PMID: 39033501 DOI: 10.1111/aji.13905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024] Open
Abstract
PROBLEM The vaginal microbiome has a substantial role in the occurrence of preterm birth (PTB), which contributes substantially to neonatal mortality worldwide. However, current bioinformatics approaches mostly concentrate on the taxonomic classification and functional profiling of the microbiome, limiting their abilities to elucidate the complex factors that contribute to PTB. METHOD OF STUDY A total of 3757 vaginal microbiome 16S rRNA samples were obtained from five publicly available datasets. The samples were divided into two categories based on pregnancy outcome: preterm birth (PTB) (N = 966) and term birth (N = 2791). Additionally, the samples were further categorized based on the participants' race and trimester. The 16S rRNA reads were subjected to taxonomic classification and functional profiling using the Parallel-META 3 software in Ubuntu environment. The obtained abundances were analyzed using an integrated systems biology and machine learning approach to determine the key microbes, pathways, and genes that contribute to PTB. The resulting features were further subjected to statistical analysis to identify the top nine features with the greatest effect sizes. RESULTS We identified nine significant features, namely Shuttleworthia, Megasphaera, Sneathia, proximal tubule bicarbonate reclamation pathway, systemic lupus erythematosus pathway, transcription machinery pathway, lepA gene, pepX gene, and rpoD gene. Their abundance variations were observed through the trimesters. CONCLUSIONS Vaginal infections caused by Shuttleworthia, Megasphaera, and Sneathia and altered small metabolite biosynthesis pathways such as lipopolysaccharide folate and retinal may increase the susceptibility to PTB. The identified organisms, genes, pathways, and their networks may be specifically targeted for the treatment of bacterial infections that increase PTB risk.
Collapse
Affiliation(s)
- Sudeepti Kulshrestha
- Systems Biology and Data Analytics Research Lab, Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Priyanka Narad
- Division of Biomedical Informatics (BMI), Indian Council of Medical Research, Ansari Nagar, New Delhi, India
| | - Brojen Singh
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Somnath S Pai
- Amity Institute of Virology & Immunology, Amity University, Noida, Uttar Pradesh, India
| | - Pooja Vijayaraghavan
- Anti-mycotic Drug Susceptibility Laboratory, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Ansh Tandon
- Systems Biology and Data Analytics Research Lab, Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Payal Gupta
- Systems Biology and Data Analytics Research Lab, Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Deepak Modi
- Molecular and Cellular Biology Laboratory, National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Abhishek Sengupta
- Systems Biology and Data Analytics Research Lab, Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| |
Collapse
|
18
|
Halte M, Popp PF, Hathcock D, Severn J, Fischer S, Goosmann C, Ducret A, Charpentier E, Tu Y, Lauga E, Erhardt M, Renault TT. Bacterial motility depends on a critical flagellum length and energy-optimised assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.599820. [PMID: 38979141 PMCID: PMC11230379 DOI: 10.1101/2024.06.28.599820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The flagellum is the most complex macromolecular structure known in bacteria and comprised of around two dozen distinct proteins. The main building block of the long, external flagellar filament, flagellin, is secreted through the flagellar type-III secretion system at a remarkable rate of several tens of thousands amino acids per second, significantly surpassing the rates achieved by other pore-based protein secretion systems. The evolutionary implications and potential benefits of this high secretion rate for flagellum assembly and function, however, have remained elusive. In this study, we provide both experimental and theoretical evidence that the flagellar secretion rate has been evolutionarily optimized to facilitate rapid and efficient construction of a functional flagellum. By synchronizing flagellar assembly, we found that a minimal filament length of 2.5 µm was required for swimming motility. Biophysical modelling revealed that this minimal filament length threshold resulted from an elasto-hydrodynamic instability of the whole swimming cell, dependent on the filament length. Furthermore, we developed a stepwise filament labeling method combined with electron microscopy visualization to validate predicted flagellin secretion rates of up to 10,000 amino acids per second. A biophysical model of flagellum growth demonstrates that the observed high flagellin secretion rate efficiently balances filament elongation and energy consumption, thereby enabling motility in the shortest amount of time. Taken together, these insights underscore the evolutionary pressures that have shaped the development and optimization of the flagellum and type-III secretion system, illuminating the intricate interplay between functionality and efficiency in assembly of large macromolecular structures. Significance statement Our study demonstrates how protein secretion of the bacterial flagellum is finely tuned to optimize filament assembly rate and flagellum function while minimizing energy consumption. By measuring flagellar filament lengths and bacterial swimming after initiation of flag-ellum assembly, we were able to establish the minimal filament length necessary for swimming motility, which we rationalized physically as resulting from an elasto-hydrodynamic instability of the swimming cell. Our bio-physical model of flagellum growth further illustrates how the physiological flagellin secretion rate is optimized to maximize filament elongation while conserving energy. These findings illuminate the evolutionary pressures that have shaped the function of the bacterial flagellum and type-III secretion system, driving improvements in bacterial motility and overall fitness.
Collapse
|
19
|
Liu X, Ishak MI, Ma H, Su B, Nobbs AH. Bacterial Surface Appendages Modulate the Antimicrobial Activity Induced by Nanoflake Surfaces on Titanium. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310149. [PMID: 38233200 PMCID: PMC7616388 DOI: 10.1002/smll.202310149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/06/2024] [Indexed: 01/19/2024]
Abstract
Bioinspired nanotopography is a promising approach to generate antimicrobial surfaces to combat implant-associated infection. Despite efforts to develop bactericidal 1D structures, the antibacterial capacity of 2D structures and their mechanism of action remains uncertain. Here, hydrothermal synthesis is utilized to generate two 2D nanoflake surfaces on titanium (Ti) substrates and investigate the physiological effects of nanoflakes on bacteria. The nanoflakes impair the attachment and growth of Escherichia coli and trigger the accumulation of intracellular reactive oxygen species (ROS), potentially contributing to the killing of adherent bacteria. E. coli surface appendages type-1 fimbriae and flagella are not implicated in the nanoflake-mediated modulation of bacterial attachment but do influence the bactericidal effects of nanoflakes. An E. coli ΔfimA mutant lacking type-1 fimbriae is more susceptible to the bactericidal effects of nanoflakes than the parent strain, while E. coli cells lacking flagella (ΔfliC) are more resistant. The results suggest that type-1 fimbriae confer a cushioning effect that protects bacteria upon initial contact with the nanoflake surface, while flagella-mediated motility can lead to elevated membrane abrasion. This finding offers a better understanding of the antibacterial properties of nanoflake structures that can be applied to the design of antimicrobial surfaces for future medical applications.
Collapse
Affiliation(s)
- Xiayi Liu
- Bristol Dental School Research Laboratories, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1, 3NY, UK
| | - Mohd I Ishak
- Bristol Dental School Research Laboratories, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1, 3NY, UK
| | - Huan Ma
- School of Chemistry, Centre for Organized Matter Chemistry and Centre for Protolife Research, University of Bristol, Bristol, BS8 1TS, UK
| | - Bo Su
- Bristol Dental School Research Laboratories, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1, 3NY, UK
| | - Angela H Nobbs
- Bristol Dental School Research Laboratories, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1, 3NY, UK
| |
Collapse
|
20
|
Li Y, Wangjiang T, Sun Z, Shi L, Chen S, Chen L, Guo X, Wu W, Xiong G, Wang L. Inhibition mechanism of crude lipopeptide from Bacillus subtilis against Aeromonas veronii growth, biofilm formation, and spoilage of channel catfish flesh. Food Microbiol 2024; 120:104489. [PMID: 38431332 DOI: 10.1016/j.fm.2024.104489] [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: 11/28/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 03/05/2024]
Abstract
Aeromonas veronii is associated with food spoilage and some human diseases, such as diarrhea, gastroenteritis, hemorrhagic septicemia or asymptomatic and even death. This research investigated the mechanism of the growth, biofilm formation, virulence, stress resistance, and spoilage potential of Bacillus subtilis lipopeptide against Aeromonas veronii. Lipopeptides suppressed the transmembrane transport of Aeromonas veronii by changing the cell membrane's permeability, the structure of membrane proteins, and Na+/K+-ATPase. Lipopeptide significantly reduced the activities of succinate dehydrogenase (SDH) and malate dehydrogenase (MDH) by 86.03% and 56.12%, respectively, ultimately slowing Aeromonas veronii growth. Lipopeptides also restrained biofilm formation by inhibiting Aeromonas veronii motivation and extracellular polysaccharide secretion. Lipopeptides downregulated gene transcriptional levels related to the virulence and stress tolerance of Aeromonas veronii. Furthermore, lipopeptides treatment resulted in a considerable decrease in the extracellular protease activity of Aeromonas veronii, which restrained the decomposing of channel catfish flesh. This research provides new insights into lipopeptides for controlling Aeromonas veronii and improving food safety.
Collapse
Affiliation(s)
- Yali Li
- Key Laboratory of Agricultural Products Cold Chain Logistics, Ministry of Agriculture and Rural Affairs, Institute of Agro-Products Processing and Nuclear Agricultural Technology, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Tianqi Wangjiang
- Key Laboratory of Agricultural Products Cold Chain Logistics, Ministry of Agriculture and Rural Affairs, Institute of Agro-Products Processing and Nuclear Agricultural Technology, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Zhida Sun
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Liu Shi
- Key Laboratory of Agricultural Products Cold Chain Logistics, Ministry of Agriculture and Rural Affairs, Institute of Agro-Products Processing and Nuclear Agricultural Technology, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Sheng Chen
- Key Laboratory of Agricultural Products Cold Chain Logistics, Ministry of Agriculture and Rural Affairs, Institute of Agro-Products Processing and Nuclear Agricultural Technology, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Lang Chen
- Key Laboratory of Agricultural Products Cold Chain Logistics, Ministry of Agriculture and Rural Affairs, Institute of Agro-Products Processing and Nuclear Agricultural Technology, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Xiaojia Guo
- Key Laboratory of Agricultural Products Cold Chain Logistics, Ministry of Agriculture and Rural Affairs, Institute of Agro-Products Processing and Nuclear Agricultural Technology, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Wenjin Wu
- Key Laboratory of Agricultural Products Cold Chain Logistics, Ministry of Agriculture and Rural Affairs, Institute of Agro-Products Processing and Nuclear Agricultural Technology, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Guangquan Xiong
- Key Laboratory of Agricultural Products Cold Chain Logistics, Ministry of Agriculture and Rural Affairs, Institute of Agro-Products Processing and Nuclear Agricultural Technology, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Lan Wang
- Key Laboratory of Agricultural Products Cold Chain Logistics, Ministry of Agriculture and Rural Affairs, Institute of Agro-Products Processing and Nuclear Agricultural Technology, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China.
| |
Collapse
|
21
|
Manisha Y, Srinivasan M, Jobichen C, Rosenshine I, Sivaraman J. Sensing for survival: specialised regulatory mechanisms of Type III secretion systems in Gram-negative pathogens. Biol Rev Camb Philos Soc 2024; 99:837-863. [PMID: 38217090 DOI: 10.1111/brv.13047] [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: 10/20/2021] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/15/2024]
Abstract
For centuries, Gram-negative pathogens have infected the human population and been responsible for numerous diseases in animals and plants. Despite advancements in therapeutics, Gram-negative pathogens continue to evolve, with some having developed multi-drug resistant phenotypes. For the successful control of infections caused by these bacteria, we need to widen our understanding of the mechanisms of host-pathogen interactions. Gram-negative pathogens utilise an array of effector proteins to hijack the host system to survive within the host environment. These proteins are secreted into the host system via various secretion systems, including the integral Type III secretion system (T3SS). The T3SS spans two bacterial membranes and one host membrane to deliver effector proteins (virulence factors) into the host cell. This multifaceted process has multiple layers of regulation and various checkpoints. In this review, we highlight the multiple strategies adopted by these pathogens to regulate or maintain virulence via the T3SS, encompassing the regulation of small molecules to sense and communicate with the host system, as well as master regulators, gatekeepers, chaperones, and other effectors that recognise successful host contact. Further, we discuss the regulatory links between the T3SS and other systems, like flagella and metabolic pathways including the tricarboxylic acid (TCA) cycle, anaerobic metabolism, and stringent cell response.
Collapse
Affiliation(s)
- Yadav Manisha
- Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Mahalashmi Srinivasan
- Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Chacko Jobichen
- Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Ilan Rosenshine
- Department of Microbiology and Molecular Genetics, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem, 91120, Israel
| | - J Sivaraman
- Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| |
Collapse
|
22
|
Wang C, Ma Q, Zhang J, Meng N, Xu D. Co-culture of benzalkonium chloride promotes the biofilm formation and decreases the antibiotic susceptibility of a Pseudomonas aeruginosa strain. ENVIRONMENTAL SCIENCE. PROCESSES & IMPACTS 2024; 26:858-869. [PMID: 38687259 DOI: 10.1039/d4em00035h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Benzalkonium chloride (BAC) is a disinfectant with broad-spectrum antibacterial properties, yet despite its widespread use and detection in the environment, the effects of BAC exposure on microorganisms remain poorly documented. Herein, the impacts of BAC on a Pseudomonas aeruginosa strain Jade-X were systematically investigated. The results demonstrated that the minimum inhibitory concentration (MIC) of BAC against strain Jade-X was 64 mg L-1. Exposure to BAC concentrations of 8, 16, 32, and 64 mg L-1 significantly augmented biofilm formation by 2.03-, 2.43-, 2.96-, and 2.56-fold respectively. The swimming and twitching abilities, along with the virulence factor production, were inhibited. Consistently, quantitative reverse transcription PCR assays revealed significant downregulation of genes related to flagellate- and pili-mediated motilities (flgD, flgE, pilB, pilQ, and motB), as well as phzA and phzB genes involved in pyocyanin production. The results of disk diffusion and MIC assays demonstrated that BAC decreased the antibiotic susceptibility of ciprofloxacin, levofloxacin, norfloxacin, and tetracycline. Conversely, an opposite trend was observed for polymyxin B and ceftriaxone. Genomic analysis revealed that strain Jade-X harbored eleven resistance-nodulation-cell division efflux pumps, with mexCD-oprJ exhibiting significant upregulation while mexEF-oprN and mexGHI-opmD were downregulated. In addition, the quorum sensing-related regulators LasR and RhlR were also suppressed, implying that BAC might modulate the physiological and biochemical behaviors of strain Jade-X by attenuating the quorum sensing system. This study enhances our understanding of interactions between BAC and P. aeruginosa, providing valuable insights to guide the regulation and rational use of BAC.
Collapse
Affiliation(s)
- Caihong Wang
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian 116026, China.
| | - Qiao Ma
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian 116026, China.
| | - Jiaxin Zhang
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian 116026, China.
| | - Nan Meng
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian 116026, China.
| | - Dan Xu
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian 116026, China.
| |
Collapse
|
23
|
Wang X, Zhu H, Hu J, Zhang B, Guo W, Wang Z, Wang D, Qi J, Tian M, Bao Y, Si F, Wang S. Genetic distribution, characterization, and function of Escherichia coli type III secretion system 2 (ETT2). iScience 2024; 27:109763. [PMID: 38706860 PMCID: PMC11068852 DOI: 10.1016/j.isci.2024.109763] [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: 05/07/2024] Open
Abstract
Many Gram-negative bacteria use type Ⅲ secretion system (T3SS) to inject effector proteins and subvert host signaling pathways, facilitating the growth, survival, and virulence. Notably, some bacteria harbor multiple distinct T3SSs with different functions. An extraordinary T3SS, the Escherichia coli Type III Secretion System 2 (ETT2), is widespread among Escherichia coli (E. coli) strains. Since many ETT2 carry genetic mutations or deletions, it is thought to be nonfunctional. However, increasing studies highlight ETT2 contributes to E. coli pathogenesis. Here, we present a comprehensive overview of genetic distribution and characterization of ETT2. Subsequently, we outline its functional potential, contending that an intact ETT2 may retain the capacity to translocate effector proteins and manipulate the host's innate immune response. Given the potential zoonotic implications associated with ETT2-carrying bacteria, further investigations into the structure, function and regulation of ETT2 are imperative for comprehensive understanding of E. coli pathogenicity and the development of effective control strategies.
Collapse
Affiliation(s)
- Xinyu Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Hong Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Jiangang Hu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Beibei Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Weiqi Guo
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Zhiyang Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Di Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Jingjing Qi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Mingxing Tian
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Yanqing Bao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Fusheng Si
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
| | - Shaohui Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| |
Collapse
|
24
|
Wan X, Wang W, Zhu J, Xiao Y. Antibacterial peptide Reg4 ameliorates Pseudomonas aeruginosa-induced pulmonary inflammation and fibrosis. Microbiol Spectr 2024; 12:e0390523. [PMID: 38501823 PMCID: PMC11064540 DOI: 10.1128/spectrum.03905-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/08/2024] [Indexed: 03/20/2024] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is a Gram-negative facultative anaerobe that has become an important cause of severe infections in humans, particularly in patients with cystic fibrosis. The development of efficacious methods or mendicants against P. aeruginosa is still needed. We previously reported that regenerating islet-derived family member 4 (Reg4) has bactericidal activity against Salmonella Typhimurium, a Gram-negative flagellated bacterium. We herein explore whether Reg4 has bactericidal activity against P. aeruginosa. In the P. aeruginosa PAO1-chronic infection model, Reg4 significantly inhibits the colonization of PAO1 in the lung and subsequently ameliorates pulmonary inflammation and fibrosis. Reg4 recombinant protein suppresses the growth motility and biofilm formation capability of PAO1 in vitro. Mechanistically, Reg4 not only exerts bactericidal action via direct binding to the P. aeruginosa cell wall but also enhances the phagocytosis of alveolar macrophages in the host. Taken together, our study demonstrates that Reg4 may provide protection against P. aeruginosa-induced pulmonary inflammation and fibrosis via its antibacterial activity.IMPORTANCEChronic lung infection with Pseudomonas aeruginosa is a leading cause of morbidity and mortality in patients with cystic fibrosis. Due to the antibiotic resistance of Pseudomonas aeruginosa, antimicrobial peptides appear to be a potential alternative to combat its infection. In this study, we report an antimicrobial peptide, regenerating islet-derived 4 (Reg4), that showed killing activity against clinical strains of Pseudomonas aeruginosa PAO1 and ameliorated PAO1-induced pulmonary inflammation and fibrosis. Experimental data also showed Reg4 directly bound to the bacterial cell membrane and enhanced the phagocytosis of host alveolar macrophages. Our presented study will be a helpful resource in searching for novel antimicrobial peptides that could have the potential to replace conventional antibiotics.
Collapse
Affiliation(s)
- Xiaoyu Wan
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weipeng Wang
- Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Zhu
- Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongtao Xiao
- Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
25
|
Roh H, Kannimuthu D. Genomic and Transcriptomic Diversification of Flagellin Genes Provides Insight into Environmental Adaptation and Phylogeographic Characteristics in Aeromonas hydrophila. MICROBIAL ECOLOGY 2024; 87:65. [PMID: 38695873 PMCID: PMC11065939 DOI: 10.1007/s00248-024-02373-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/01/2024] [Indexed: 05/05/2024]
Abstract
Aeromonas hydrophila is an opportunistic motile pathogen with a broad host range, infecting both terrestrial and aquatic animals. Environmental and geographical conditions exert selective pressure on both geno- and phenotypes of pathogens. Flagellin, directly exposed to external environments and containing important immunogenic epitopes, may display significant variability in response to external conditions. In this study, we conducted a comparative analysis of ~ 150 A. hydrophila genomes, leading to the identification of six subunits of the flagellin gene (fla-1 to fla-4, flaA, and flaB). Individual strains harbored different composition of flagellin subunits and copies. The composition of subunits showed distinct patterns depending on environmental sources. Strains from aquatic environments were mainly comprised of fla-1 to fla-4 subunits, while terrestrial strains predominated in groups harboring flaA and flaB subunits. Each flagellin showed varying levels of expression, with flaA and flaB demonstrating significantly higher expression compared to others. One of the chemotaxis pathways that control flagellin movement through a two-component system was significantly upregulated in flaA(+ 1)/flaB(+ 1) group, whereas flaA and flaB showed different transcriptomic expressions. The genes positively correlated with flaA expression were relevant to biofilm formation and bacterial chemotaxis, but flaB showed a negative correlation with the genes in ABC transporters and quorum sensing pathway. However, the expression patterns of fla-2 to fla-4 were identical. This suggests various types of flagellin subunits may have different biological functions. The composition and expression levels of flagellin subunits could provide valuable insights into the adaptation of A. hydrophila and the differences among strains in response to various external environments.
Collapse
Affiliation(s)
- HyeongJin Roh
- Pathogen Transmission and Disease Research Group, Institute of Marine Research, PO Box 1870 Nordnes 5870, Bergen, Norway.
| | - Dhamotharan Kannimuthu
- Pathogen Transmission and Disease Research Group, Institute of Marine Research, PO Box 1870 Nordnes 5870, Bergen, Norway
| |
Collapse
|
26
|
Gambino M, Sørensen MCH. Flagellotropic phages: common yet diverse host interaction strategies. Curr Opin Microbiol 2024; 78:102451. [PMID: 38452595 DOI: 10.1016/j.mib.2024.102451] [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: 09/28/2023] [Revised: 01/14/2024] [Accepted: 02/10/2024] [Indexed: 03/09/2024]
Abstract
Many bacteriophages (phages) interact with flagella and rely on bacterial motility for successful infection of their hosts. Yet, limited information is available on how phages have evolved to recognize and bind both flagella and subsequent surface receptors for phage DNA injection. Here, we present an update on the current knowledge of flagellotropic phages using a few well-studied phages as examples to unravel the molecular details of bacterial host recognition. We discuss the recent advances in the role of globular exposed flagellin domains and flagella glycosylation in phage binding to the flagella. In addition, we present diverse types of surface receptors and phage components responsible for the interaction with the host. Finally, we point to questions remaining to be answered and new approaches to study this unique group of phages.
Collapse
Affiliation(s)
- Michela Gambino
- Institute of Conservation, Royal Danish Academy, Copenhagen, Denmark
| | - Martine C H Sørensen
- Section of Food Safety and Zoonoses, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark.
| |
Collapse
|
27
|
Seo B, Lee D, Jeon H, Ha J, Suh S. MotGen: a closed-loop bacterial motility control framework using generative adversarial networks. Bioinformatics 2024; 40:btae170. [PMID: 38552318 PMCID: PMC11031359 DOI: 10.1093/bioinformatics/btae170] [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: 12/20/2023] [Revised: 03/02/2024] [Accepted: 03/27/2024] [Indexed: 04/21/2024] Open
Abstract
MOTIVATION Many organisms' survival and behavior hinge on their responses to environmental signals. While research on bacteria-directed therapeutic agents has increased, systematic exploration of real-time modulation of bacterial motility remains limited. Current studies often focus on permanent motility changes through genetic alterations, restricting the ability to modulate bacterial motility dynamically on a large scale. To address this gap, we propose a novel real-time control framework for systematically modulating bacterial motility dynamics. RESULTS We introduce MotGen, a deep learning approach leveraging Generative Adversarial Networks to analyze swimming performance statistics of motile bacteria based on live cell imaging data. By tracking objects and optimizing cell trajectory mapping under environmentally altered conditions, we trained MotGen on a comprehensive statistical dataset derived from real image data. Our experimental results demonstrate MotGen's ability to capture motility dynamics from real bacterial populations with low mean absolute error in both simulated and real datasets. MotGen allows us to approach optimal swimming conditions for desired motility statistics in real-time. MotGen's potential extends to practical biomedical applications, including immune response prediction, by providing imputation of bacterial motility patterns based on external environmental conditions. Our short-term, in-situ interventions for controlling motility behavior offer a promising foundation for the development of bacteria-based biomedical applications. AVAILABILITY AND IMPLEMENTATION MotGen is presented as a combination of Matlab image analysis code and a machine learning workflow in Python. Codes are available at https://github.com/bgmseo/MotGen, for cell tracking and implementation of trained models to generate bacterial motility statistics.
Collapse
Affiliation(s)
- BoGeum Seo
- Department of Mechanical Engineering, Seoul National University, 08826 Seoul, Republic of Korea
| | - DoHee Lee
- Center for Healthcare Robotics, Korea Institute of Science & Technology, 02792 Seoul, Republic of Korea
| | - Heungjin Jeon
- Infection Control Convergence Research Center, Chungnam National University, 34134 Daejeon, Republic of Korea
| | - Junhyoung Ha
- Center for Healthcare Robotics, Korea Institute of Science & Technology, 02792 Seoul, Republic of Korea
| | - SeungBeum Suh
- Center for Healthcare Robotics, Korea Institute of Science & Technology, 02792 Seoul, Republic of Korea
| |
Collapse
|
28
|
Williams TJ, Allen MA, Ray AE, Benaud N, Chelliah DS, Albanese D, Donati C, Selbmann L, Coleine C, Ferrari BC. Novel endolithic bacteria of phylum Chloroflexota reveal a myriad of potential survival strategies in the Antarctic desert. Appl Environ Microbiol 2024; 90:e0226423. [PMID: 38372512 PMCID: PMC10952385 DOI: 10.1128/aem.02264-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/02/2024] [Indexed: 02/20/2024] Open
Abstract
The ice-free McMurdo Dry Valleys of Antarctica are dominated by nutrient-poor mineral soil and rocky outcrops. The principal habitat for microorganisms is within rocks (endolithic). In this environment, microorganisms are provided with protection against sub-zero temperatures, rapid thermal fluctuations, extreme dryness, and ultraviolet and solar radiation. Endolithic communities include lichen, algae, fungi, and a diverse array of bacteria. Chloroflexota is among the most abundant bacterial phyla present in these communities. Among the Chloroflexota are four novel classes of bacteria, here named Candidatus Spiritibacteria class. nov. (=UBA5177), Candidatus Martimicrobia class. nov. (=UBA4733), Candidatus Tarhunnaeia class. nov. (=UBA6077), and Candidatus Uliximicrobia class. nov. (=UBA2235). We retrieved 17 high-quality metagenome-assembled genomes (MAGs) that represent these four classes. Based on genome predictions, all these bacteria are inferred to be aerobic heterotrophs that encode enzymes for the catabolism of diverse sugars. These and other organic substrates are likely derived from lichen, algae, and fungi, as metabolites (including photosynthate), cell wall components, and extracellular matrix components. The majority of MAGs encode the capacity for trace gas oxidation using high-affinity uptake hydrogenases, which could provide energy and metabolic water required for survival and persistence. Furthermore, some MAGs encode the capacity to couple the energy generated from H2 and CO oxidation to support carbon fixation (atmospheric chemosynthesis). All encode mechanisms for the detoxification and efflux of heavy metals. Certain MAGs encode features that indicate possible interactions with other organisms, such as Tc-type toxin complexes, hemolysins, and macroglobulins.IMPORTANCEThe ice-free McMurdo Dry Valleys of Antarctica are the coldest and most hyperarid desert on Earth. It is, therefore, the closest analog to the surface of the planet Mars. Bacteria and other microorganisms survive by inhabiting airspaces within rocks (endolithic). We identify four novel classes of phylum Chloroflexota, and, based on interrogation of 17 metagenome-assembled genomes, we predict specific metabolic and physiological adaptations that facilitate the survival of these bacteria in this harsh environment-including oxidation of trace gases and the utilization of nutrients (including sugars) derived from lichen, algae, and fungi. We propose that such adaptations allow these endolithic bacteria to eke out an existence in this cold and extremely dry habitat.
Collapse
Affiliation(s)
- Timothy J Williams
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Michelle A Allen
- School of Biological, Earth and Environmental Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Angelique E Ray
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Nicole Benaud
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Devan S Chelliah
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Davide Albanese
- Research and Innovation Center, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | - Claudio Donati
- Research and Innovation Center, Fondazione Edmund Mach, San Michele all'Adige, Italy
| | - Laura Selbmann
- Department of Ecological and Biological Sciences, University of Tuscia, Largo dell'Università, Viterbo, Italy
- Mycological Section, Italian Antarctic National Museum (MNA), Genova, Italy
| | - Claudia Coleine
- Department of Ecological and Biological Sciences, University of Tuscia, Largo dell'Università, Viterbo, Italy
| | - Belinda C Ferrari
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
29
|
Fabian B, Foster C, Asher A, Hassan K, Paulsen I, Tetu S. Identifying the suite of genes central to swimming in the biocontrol bacterium Pseudomonas protegens Pf-5. Microb Genom 2024; 10:001212. [PMID: 38546328 PMCID: PMC11004494 DOI: 10.1099/mgen.0.001212] [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: 01/01/2024] [Accepted: 02/20/2024] [Indexed: 04/12/2024] Open
Abstract
Swimming motility is a key bacterial trait, important to success in many niches. Biocontrol bacteria, such as Pseudomonas protegens Pf-5, are increasingly used in agriculture to control crop diseases, where motility is important for colonization of the plant rhizosphere. Swimming motility typically involves a suite of flagella and chemotaxis genes, but the specific gene set employed for both regulation and biogenesis can differ substantially between organisms. Here we used transposon-directed insertion site sequencing (TraDIS), a genome-wide approach, to identify 249 genes involved in P. protegens Pf-5 swimming motility. In addition to the expected flagella and chemotaxis, we also identified a suite of additional genes important for swimming, including genes related to peptidoglycan turnover, O-antigen biosynthesis, cell division, signal transduction, c-di-GMP turnover and phosphate transport, and 27 conserved hypothetical proteins. Gene knockout mutants and TraDIS data suggest that defects in the Pst phosphate transport system lead to enhanced swimming motility. Overall, this study expands our knowledge of pseudomonad motility and highlights the utility of a TraDIS-based approach for analysing the functions of thousands of genes. This work sets a foundation for understanding how swimming motility may be related to the inconsistency in biocontrol bacteria performance in the field.
Collapse
Affiliation(s)
- B.K. Fabian
- School of Natural Sciences, Macquarie University, Sydney, Australia
- ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, Australia
| | - C. Foster
- School of Natural Sciences, Macquarie University, Sydney, Australia
| | - A. Asher
- School of Natural Sciences, Macquarie University, Sydney, Australia
| | - K.A. Hassan
- ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, Australia
- School of Environmental and Life Sciences, University of Newcastle, Newcastle, Australia
| | - I.T. Paulsen
- School of Natural Sciences, Macquarie University, Sydney, Australia
- ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, Australia
| | - S.G. Tetu
- School of Natural Sciences, Macquarie University, Sydney, Australia
- ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, Australia
| |
Collapse
|
30
|
Xiong C, Xiong C, Lu J, Long R, Jiao H, Li Y, Wang B, Lin Y, Ye H, Lin L, Wu R. flgL mutation reduces pathogenicity of Aeromonas hydrophila by negatively regulating swimming ability, biofilm forming ability, adherence and virulence gene expression. Int J Biol Macromol 2024; 261:129676. [PMID: 38272420 DOI: 10.1016/j.ijbiomac.2024.129676] [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: 12/10/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 01/27/2024]
Abstract
Aeromonas hydrophila is a serious human and animal co-pathogenic bacterium. Flagellum, a key virulence factor, is vital for bacterium tissue colonization and invasion. flgL is a crucial gene involved in the composition of flagellum. However, the impact of flgL on virulence is not yet clear. In this study, we constructed a stable mutant strain (△flgL-AH) using homologous recombination. The results of the attack experiments indicated a significant decrease in the virulence of △flgL-AH. The biological properties analysis revealed a significant decline in swimming ability and biofilm formation capacity in △flgL-AH and the transmission electron microscope results showed that the ∆flgL-AH strain did not have a flagellar structure. Moreover, a significant decrease in the adhesion capacity of ∆flgL-AH was found using absolute fluorescence quantitative polymerase chain reaction (PCR). The quantitative real-time PCR results showed that the expression of omp and the eight flagellum-related genes were down-regulated. In summary, flgL mutation leads to a reduction in pathogenicity possibly via decreasing the swimming ability, biofilm formation capacity and adhesion capacity, these changes might result from the down expression of omp and flagellar-related genes.
Collapse
Affiliation(s)
- Caijiang Xiong
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Chuanyu Xiong
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Jiahui Lu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Rui Long
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Hanyang Jiao
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Yun Li
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Bei Wang
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
| | - Ying Lin
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Hua Ye
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China
| | - Lingyun Lin
- Agriculture Ministry Key Laboratory of Healthy Freshwater Aquaculture, Key Laboratory of Fish Health and Nutrition of Zhejiang Province, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, China.
| | - Ronghua Wu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China.
| |
Collapse
|
31
|
Zhai Y, Tian W, Chen K, Lan L, Kan J, Shi H. Flagella-mediated adhesion of Escherichia coli O157:H7 to surface of stainless steel, glass and fresh produces during sublethal injury and recovery. Food Microbiol 2024; 117:104383. [PMID: 37918998 DOI: 10.1016/j.fm.2023.104383] [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: 07/24/2023] [Revised: 09/06/2023] [Accepted: 09/10/2023] [Indexed: 11/04/2023]
Abstract
E. coli O157:H7 can be induced into sublethally injured (SI) state by lactic acid (LA) and regain activity in nutrient environments. This research clarified the role of flagella-related genes (fliD, fliS, cheA and motA) in adhesion of E. coli O157:H7 onto stainless steel, glass, lettuce, spinach, red cabbage and cucumber during LA-induced SI and recovery by plate counting. Results of adhesion showed improper flagellar rotation caused by the deletion of motA resulting in the decreased adhesion. Motility of wildtype determined by diameter of motility halo decreased in SI state and repaired with recovery time increasing, lagging behind changes in expression of flagella-related genes. Flagellar function-impaired strains all exhibited non-motile property. Thus, we speculated that flagella-mediated motility is critical in early stage of adhesion. We also found the effects of Fe2+, Ca2+ and Mn2+ on adhesion or motility of wildtype was independent of bacterial states. However, the addition of Ca2+ and Mn2+ did not affect motility of flagellar function-impaired strains as they did on wildtype. This research provides new insights to understand the role of flagella and cations in bacterial adhesion, which will aid in development of anti-adhesion agents to reduce bio-contamination in food processing.
Collapse
Affiliation(s)
- Yujun Zhai
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Weina Tian
- College of Bioengineering, Beijing Polytechnic, Beijing, 100176, China
| | - Kewei Chen
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Linshu Lan
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Jianquan Kan
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Hui Shi
- College of Food Science, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
32
|
Li K, Yu K, Huang Z, Liu X, Mei L, Ren X, Bai X, Gao H, Sun Z, Liu X, Wang D. Stenotrophomonas maltophilia complex: insights into evolutionary relationships, global distribution and pathogenicity. Front Cell Infect Microbiol 2024; 13:1325379. [PMID: 38268792 PMCID: PMC10806987 DOI: 10.3389/fcimb.2023.1325379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 12/21/2023] [Indexed: 01/26/2024] Open
Abstract
Introduction Stenotrophomonas maltophilia complex (Smc) comprises opportunistic Gram-negative bacilli responsible for various nosocomial infections. Limited data exists concerning its evolutionary lineage, global prevalence and pathogenicity. Methods We conducted an extensive genomic analysis on 734 Smc genomes, of which 90 were newly sequenced and isolated from different patients. The species composition and evolutionary relationships of Smc were examined using core protein sequence analysis. Pathogenicity evaluation was used by assays for swimming motility, biofilm formation and identification of virulence factors. The broth microdilution method was used to evaluate the drug resistance spectrum of clinical isolates. Results Phylogenetic analyses delineated 24 species-level clades, dominated by S. maltophilia (42.8%), S. sepilia (13.6%) and S. geniculata (9.9%). Geographically, strains were primarily distributed in Europe (34.2%), Asia (33.7%) and North America (24.0%), with intricate global distribution patterns. Meanwhile, 154 virulence-associated genes and 46 antimicrobial resistance genes within Smc were identified. These genes encoded span various functions, including motility, adherence, toxin, RND antibiotic efflux pumps, beta-lactamases and aminoglycoside-modifying enzymes. Moreover, significant variations were indicated in swimming motility and biofilm-forming capability across the different species, with S. sepilia exhibiting superior levels of both traits. Additionally, no statistically significant discrepancy was detected among Smc species to other antibiotics, despite the fact that all S. geniculata isolates were resistant to Ceftazidime and much higher than other species. Conclusion Our findings indicate the need to pay increased attention to other mainstream species of Smc besides S. maltophilia in order to better manage Smc-related infections and tailor effective treatment strategies.
Collapse
Affiliation(s)
- Kun Li
- School of Public Health, Lanzhou University, Lanzhou, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Keyi Yu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhenzhou Huang
- Microbiology Laboratory, Hangzhou Center for Disease Control and Prevention, Hangzhou, China
| | - Xiao Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Li Mei
- National Pathogen Resource Center, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaodong Ren
- School of Public Health, Lanzhou University, Lanzhou, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xuemei Bai
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - He Gao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhiwen Sun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaoning Liu
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Duochun Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
33
|
Han S, Byun JW, Lee M. Comparative Transcriptomic Analysis of Flagellar-Associated Genes in Salmonella Typhimurium and Its rnc Mutant. J Microbiol 2024; 62:33-48. [PMID: 38182942 DOI: 10.1007/s12275-023-00099-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 01/07/2024]
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is a globally recognized foodborne pathogen that affects both animals and humans. Endoribonucleases mediate RNA processing and degradation in the adaptation of bacteria to environmental changes and have been linked to the pathogenicity of S. Typhimurium. Not much is known about the specific regulatory mechanisms of these enzymes in S. Typhimurium, particularly in the context of environmental adaptation. Thus, this study carried out a comparative transcriptomic analysis of wild-type S. Typhimurium SL1344 and its mutant (∆rnc), which lacks the rnc gene encoding RNase III, thereby elucidating the detailed regulatory characteristics that can be attributed to the rnc gene. Global gene expression analysis revealed that the ∆rnc strain exhibited 410 upregulated and 301 downregulated genes (fold-change > 1.5 and p < 0.05), as compared to the wild-type strain. Subsequent bioinformatics analysis indicated that these differentially expressed genes are involved in various physiological functions, in both the wild-type and ∆rnc strains. This study provides evidence for the critical role of RNase III as a general positive regulator of flagellar-associated genes and its involvement in the pathogenicity of S. Typhimurium.
Collapse
Affiliation(s)
- Seungmok Han
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Ji-Won Byun
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Minho Lee
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea.
- Institute of Medical Science, College of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea.
| |
Collapse
|
34
|
Claushuis B, de Ru AH, Rotman SA, van Veelen PA, Dawson LF, Wren BW, Corver J, Smits WK, Hensbergen PJ. Revised Model for the Type A Glycan Biosynthetic Pathway in Clostridioides difficile Strain 630Δ erm Based on Quantitative Proteomics of cd0241- cd0244 Mutant Strains. ACS Infect Dis 2023; 9:2665-2674. [PMID: 37965964 PMCID: PMC10714395 DOI: 10.1021/acsinfecdis.3c00485] [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: 09/12/2023] [Revised: 10/20/2023] [Accepted: 10/30/2023] [Indexed: 11/16/2023]
Abstract
The bacterial flagellum is involved in a variety of processes including motility, adherence, and immunomodulation. In the Clostridioides difficile strain 630Δerm, the main filamentous component, FliC, is post-translationally modified with an O-linked Type A glycan structure. This modification is essential for flagellar function, since motility is seriously impaired in gene mutants with improper biosynthesis of the Type A glycan. The cd0240-cd0244 gene cluster encodes the Type A biosynthetic proteins, but the role of each gene, and the corresponding enzymatic activity, have not been fully elucidated. Using quantitative mass spectrometry-based proteomics analyses, we determined the relative abundance of the observed glycan variations of the Type A structure in cd0241, cd0242, cd0243, and cd0244 mutant strains. Our data not only confirm the importance of CD0241, CD0242, and CD0243 but, in contrast to previous data, also show that CD0244 is essential for the biosynthesis of the Type A modification. Combined with additional bioinformatic analyses, we propose a revised model for Type A glycan biosynthesis.
Collapse
Affiliation(s)
- Bart Claushuis
- Center
for Proteomics and Metabolomics, Leiden
University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Arnoud H. de Ru
- Center
for Proteomics and Metabolomics, Leiden
University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Sarah A. Rotman
- Center
for Proteomics and Metabolomics, Leiden
University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Peter A. van Veelen
- Center
for Proteomics and Metabolomics, Leiden
University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Lisa F. Dawson
- Faculty
of Infectious and Tropical Diseases, London
School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Brendan W. Wren
- Faculty
of Infectious and Tropical Diseases, London
School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Jeroen Corver
- Department
of Medical Microbiology, Leiden University
Medical Center, Leiden 2333 ZA, The Netherlands
| | - Wiep Klaas Smits
- Department
of Medical Microbiology, Leiden University
Medical Center, Leiden 2333 ZA, The Netherlands
| | - Paul J. Hensbergen
- Center
for Proteomics and Metabolomics, Leiden
University Medical Center, Leiden 2333 ZA, The Netherlands
| |
Collapse
|
35
|
Zhang J, Liu K, Gong X, Zhang N, Zeng Y, Ren W, Huang A, Long H, Xie Z. Transcriptome analysis of the hepatopancreas from the Litopenaeus vannamei infected with different flagellum types of Vibrio alginolyticus strains. Front Cell Infect Microbiol 2023; 13:1265917. [PMID: 38076457 PMCID: PMC10703188 DOI: 10.3389/fcimb.2023.1265917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Vibrio alginolyticus, one of the prevalently harmful Vibrio species found in the ocean, causes significant economic damage in the shrimp farming industry. Its flagellum serves as a crucial virulence factor in the invasion of host organisms. However, the processes of bacteria flagella recognition and activation of the downstream immune system in shrimp remain unclear. To enhance comprehension of this, a ΔflhG strain was created by in-frame deletion of the flhG gene in V. alginolyticus strain HN08155. Then we utilized the transcriptome analysis to examine the different immune responses in Litopenaeus vannamei hepatopancreas after being infected with the wild type and the mutant strains. The results showed that the ΔflhG strain, unlike the wild type, lost its ability to regulate flagella numbers negatively and displayed multiple flagella. When infected with the hyperflagella-type strain, the RNA-seq revealed the upregulation of several immune-related genes in the shrimp hepatopancreas. Notably, two C-type lectins (CTLs), namely galactose-specific lectin nattectin and macrophage mannose receptor 1, and the TNF receptor-associated factor (TRAF) 6 gene were upregulated significantly. These findings suggested that C-type lectins were potentially involved in flagella recognition in shrimp and the immune system was activated through the TRAF6 pathway after flagella detection by CTLs.
Collapse
Affiliation(s)
- Jingwen Zhang
- Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, Hainan University, Haikou, China
- Laboratory of Development and Utilization of Marine Microbial Resource, Hainan University, Haikou, China
| | - Kaifang Liu
- School of Fisheries, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Xiaoxiao Gong
- Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, Hainan University, Haikou, China
- Laboratory of Development and Utilization of Marine Microbial Resource, Hainan University, Haikou, China
| | - Na Zhang
- Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, Hainan University, Haikou, China
- Laboratory of Development and Utilization of Marine Microbial Resource, Hainan University, Haikou, China
| | - Yanhua Zeng
- Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, Hainan University, Haikou, China
- Laboratory of Development and Utilization of Marine Microbial Resource, Hainan University, Haikou, China
- Collaborative Innovation Center of Marine Science and Technology, Hainan University, Haikou, China
| | - Wei Ren
- Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, Hainan University, Haikou, China
- Laboratory of Development and Utilization of Marine Microbial Resource, Hainan University, Haikou, China
- Collaborative Innovation Center of Marine Science and Technology, Hainan University, Haikou, China
| | - Aiyou Huang
- Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, Hainan University, Haikou, China
- Laboratory of Development and Utilization of Marine Microbial Resource, Hainan University, Haikou, China
- Collaborative Innovation Center of Marine Science and Technology, Hainan University, Haikou, China
| | - Hao Long
- Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, Hainan University, Haikou, China
- Laboratory of Development and Utilization of Marine Microbial Resource, Hainan University, Haikou, China
- Collaborative Innovation Center of Marine Science and Technology, Hainan University, Haikou, China
| | - Zhenyu Xie
- Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, Hainan University, Haikou, China
- Laboratory of Development and Utilization of Marine Microbial Resource, Hainan University, Haikou, China
- Collaborative Innovation Center of Marine Science and Technology, Hainan University, Haikou, China
| |
Collapse
|
36
|
Partridge JD, Dufour Y, Hwang Y, Harshey RM. Flagellar motor remodeling during swarming requires FliL. Mol Microbiol 2023; 120:670-683. [PMID: 37675594 PMCID: PMC10942728 DOI: 10.1111/mmi.15148] [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: 07/14/2023] [Revised: 08/18/2023] [Accepted: 08/20/2023] [Indexed: 09/08/2023]
Abstract
FliL is an essential component of the flagellar machinery in some bacteria, but a conditional one in others. The conditional role is for optimal swarming in some bacteria. During swarming, physical forces associated with movement on a surface are expected to exert a higher load on the flagellum, requiring more motor torque to move. FliL was reported to enhance motor output in several bacteria and observed to assemble as a ring around ion-conducting stators that power the motor. In this study we identify a common new function for FliL in diverse bacteria-Escherichia coli, Bacillus subtilis, and Proteus mirabilis. During swarming, all these bacteria show increased cell speed and a skewed motor bias that suppresses cell tumbling. We demonstrate that these altered motor parameters, or "motor remodeling," require FliL. Both swarming and motor remodeling can be restored in an E. coli fliL mutant by complementation with fliL genes from P. mirabilis and B. subtilis, showing conservation of a swarming-associated FliL function across phyla. In addition, we demonstrate that the strong interaction we reported earlier between FliL and the flagellar MS-ring protein FliF is confined to the RBM-3 domain of FliF that links the periplasmic rod to the cytoplasmic C-ring. This interaction may explain several phenotypes associated with the absence of FliL.
Collapse
Affiliation(s)
- Jonathan D. Partridge
- Department of Molecular Biosciences and the LaMontagne Center for Infectious Diseases, The University of Texas at Austin, Austin, Texas, USA
| | - Yann Dufour
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - YuneSahng Hwang
- Department of Molecular Biosciences and the LaMontagne Center for Infectious Diseases, The University of Texas at Austin, Austin, Texas, USA
| | - Rasika M. Harshey
- Department of Molecular Biosciences and the LaMontagne Center for Infectious Diseases, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
37
|
Maldonado J, Czarnecka B, Harmon DE, Ruiz C. The multidrug efflux pump regulator AcrR directly represses motility in Escherichia coli. mSphere 2023; 8:e0043023. [PMID: 37787551 PMCID: PMC10597343 DOI: 10.1128/msphere.00430-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/18/2023] [Indexed: 10/04/2023] Open
Abstract
Efflux and motility are two key biological functions in bacteria. Recent findings have shown that efflux impacts flagellum biosynthesis and motility in Escherichia coli and other bacteria. AcrR is known to be the major transcriptional repressor of AcrAB-TolC, the main multidrug efflux pump in E. coli and other Enterobacteriaceae. However, the underlying molecular mechanisms of how efflux and motility are co-regulated remain poorly understood. Here, we have studied the role of AcrR in direct regulation of motility in E. coli. By combining bioinformatics, electrophoretic mobility shift assays (EMSAs), gene expression, and motility experiments, we have found that AcrR represses motility in E. coli by directly repressing transcription of the flhDC operon, but not the other flagellum genes/operons tested. flhDC encodes the master regulator of flagellum biosynthesis and motility genes. We found that such regulation primarily occurs by direct binding of AcrR to the flhDC promoter region containing the first of the two predicted AcrR-binding sites identified in this promoter. This is the first report of direct regulation by AcrR of genes unrelated to efflux or detoxification. Moreover, we report that overexpression of AcrR restores to parental levels the increased swimming motility previously observed in E. coli strains without a functional AcrAB-TolC pump, and that such effect by AcrR is prevented by the AcrR ligand and AcrAB-TolC substrate ethidium bromide. Based on these and prior findings, we provide a novel model in which AcrR senses efflux and then co-regulates efflux and motility in E. coli to maintain homeostasis and escape hazards. IMPORTANCE Efflux and motility play a major role in bacterial growth, colonization, and survival. In Escherichia coli, the transcriptional repressor AcrR is known to directly repress efflux and was later found to also repress flagellum biosynthesis and motility by Kim et al. (J Microbiol Biotechnol 26:1824-1828, 2016, doi: 10.4014/jmb.1607.07058). However, it remained unknown whether AcrR represses flagellum biosynthesis and motility directly and through which target genes, or indirectly because of altering the amount of efflux. This study reveals that AcrR represses flagellum biosynthesis and motility by directly repressing the expression of the flhDC master regulator of flagellum biosynthesis and motility genes, but not the other flagellum genes tested. We also show that the antimicrobial, efflux pump substrate, and AcrR ligand ethidium bromide regulates motility via AcrR. Overall, these findings support a novel model of direct co-regulation of efflux and motility mediated by AcrR in response to stress in E. coli.
Collapse
Affiliation(s)
- Jessica Maldonado
- Department of Biology, California State University Northridge, Northridge, California, USA
| | - Barbara Czarnecka
- Department of Biology, California State University Northridge, Northridge, California, USA
| | - Dana E. Harmon
- Department of Biology, California State University Northridge, Northridge, California, USA
| | - Cristian Ruiz
- Department of Biology, California State University Northridge, Northridge, California, USA
| |
Collapse
|
38
|
Huang W, Chen W, Chen Y, Fang S, Huang T, Chang P, Chang Y. Salmonella YqiC exerts its function through an oligomeric state. Protein Sci 2023; 32:e4749. [PMID: 37555831 PMCID: PMC10503411 DOI: 10.1002/pro.4749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/18/2023] [Accepted: 08/07/2023] [Indexed: 08/10/2023]
Abstract
Protein oligomerization occurs frequently both in vitro and in vivo, with specific functionalities associated with different oligomeric states. The YqiC protein from Salmonella Typhimurium forms a homotrimer through its C-terminal coiled-coil domain, and the protein is closely linked to the colonization and invasion of the bacteria to the host cells. To elucidate the importance of the oligomeric state of YqiC in vivo and its relation with bacterial infection, we mutated crucial residues in YqiC's coiled-coil region and confirmed the loss of trimer formation using chemical crosslinking and size exclusion chromatography coupled with multiple angle light scattering (SEC-MALS) techniques. The yqiC-knockout strain complemented with mutant YqiC showed significantly reduced colonization and invasion of Salmonella to host cells, demonstrating the critical role of YqiC oligomerization in bacterial pathogenesis. Furthermore, we conducted a protein-protein interaction study of YqiC using a pulled-down assay coupled with mass spectrometry analysis to investigate the protein's role in bacterial virulence. The results reveal that YqiC interacts with subunits of Complex II of the electron transport chain (SdhA and SdhB) and the β-subunit of F0 F1 -ATP synthase. These interactions suggest that YqiC may modulate the energy production of Salmonella and subsequently affect the assembly of crucial virulence factors, such as flagella. Overall, our findings provide new insights into the molecular mechanisms of YqiC's role in S. Typhimurium pathogenesis and suggest potential therapeutic targets for bacterial infections.
Collapse
Affiliation(s)
- Wei‐Chun Huang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Wai‐Ting Chen
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Yueh‐Chen Chen
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Shiuh‐Bin Fang
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, Shuang Ho HospitalTaipei Medical UniversityTaipeiTaiwan
- Department of Pediatrics, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Master Program for Clinical Genomics and Proteomics, College of PharmacyTaipei Medical UniversityTaipeiTaiwan
| | - Tzu‐Wen Huang
- Department of Microbiology and Immunology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Pei‐Ru Chang
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, Shuang Ho HospitalTaipei Medical UniversityTaipeiTaiwan
- Department of Pediatrics, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Yu‐Chu Chang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- International PhD Program in Cell Therapy and Regenerative Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
39
|
Worrall LJ, Majewski DD, Strynadka NCJ. Structural Insights into Type III Secretion Systems of the Bacterial Flagellum and Injectisome. Annu Rev Microbiol 2023; 77:669-698. [PMID: 37713458 DOI: 10.1146/annurev-micro-032521-025503] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Two of the most fascinating bacterial nanomachines-the broadly disseminated rotary flagellum at the heart of cellular motility and the eukaryotic cell-puncturing injectisome essential to specific pathogenic species-utilize at their core a conserved export machinery called the type III secretion system (T3SS). The T3SS not only secretes the components that self-assemble into their extracellular appendages but also, in the case of the injectisome, subsequently directly translocates modulating effector proteins from the bacterial cell into the infected host. The injectisome is thought to have evolved from the flagellum as a minimal secretory system lacking motility, with the subsequent acquisition of additional components tailored to its specialized role in manipulating eukaryotic hosts for pathogenic advantage. Both nanomachines have long been the focus of intense interest, but advances in structural and functional understanding have taken a significant step forward since 2015, facilitated by the revolutionary advances in cryo-electron microscopy technologies. With several seminal structures of each nanomachine now captured, we review here the molecular similarities and differences that underlie their diverse functions.
Collapse
Affiliation(s)
- Liam J Worrall
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, The University of British Columbia, Vancouver, British Columbia, Canada; , ,
| | - Dorothy D Majewski
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, The University of British Columbia, Vancouver, British Columbia, Canada; , ,
- Current affiliation: Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Natalie C J Strynadka
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, The University of British Columbia, Vancouver, British Columbia, Canada; , ,
| |
Collapse
|
40
|
Xie F, Zhu C, Gong L, Zhu N, Ma Q, Yang Y, Zhao X, Qin M, Lin Z, Wang Y. Engineering core-shell chromium nanozymes with inflammation-suppressing, ROS-scavenging and antibacterial properties for pulpitis treatment. NANOSCALE 2023; 15:13971-13986. [PMID: 37606502 DOI: 10.1039/d3nr02930a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Oral diseases are usually caused by inflammation and bacterial infection. Reactive oxygen species (ROS), which come from both autologous inflammation tissue and bacterial infection, play an important role in this process. Thus, the elimination of excessive intracellular ROS can be a promising strategy for anti-inflammatory treatment. With the rapid development of nanomedicines, nanozymes, which can maintain the intracellular redox balance and protect cells against oxidative damage, have shown great application prospects in the treatment of inflammation-related diseases. However, their performance in pulpitis and their related mechanisms have yet to be explored. Herein, we prepared dozens of metallic nanoparticles with core-shell structures, and among them, chromium nanoparticles (NanoCr) were selected for their great therapeutic potential for pulpitis disease. NanoCr showed a broad antibacterial spectrum and strong anti-inflammatory function. Antibacterial assays showed that NanoCr could effectively inhibit a variety of common pathogens of oral infection. In vitro experiments offered evidence of the multienzyme activity of NanoCr and its function in suppressing ROS-induced inflammation reactions. The experimental results show that NanoCr has optimal antibacterial and anti-inflammatory properties in in vitro cell models, showing great potential for the treatment of pulpitis. Therefore, the use of NanoCr could become a new therapeutic strategy for clinical pulpitis.
Collapse
Affiliation(s)
- Fei Xie
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China.
| | - Chuanda Zhu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, P.R. China.
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Lidong Gong
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, P.R. China.
| | - Ningxin Zhu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China.
| | - Qiang Ma
- Institute of Environment and Sustainable Development in Agriculture, Chinese academy of Agriculture, Beijing 100081, P.R. China
| | - Yuanyuan Yang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, P.R. China.
| | - Xinrong Zhao
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Man Qin
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China.
| | - Zhiqiang Lin
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, P.R. China.
| | - Yuanyuan Wang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China.
| |
Collapse
|
41
|
Abdulkadieva MM, Sysolyatina EV, Vasilieva EV, Litvinenko VV, Kalinin EV, Zhukhovitsky VG, Shevlyagina NV, Andreevskaya SG, Stanishevskyi YM, Vasiliev MM, Petrov OF, Ermolaeva SA. Motility provides specific adhesion patterns and improves Listeria monocytogenes invasion into human HEp-2 cells. PLoS One 2023; 18:e0290842. [PMID: 37651463 PMCID: PMC10470941 DOI: 10.1371/journal.pone.0290842] [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: 05/24/2023] [Accepted: 08/17/2023] [Indexed: 09/02/2023] Open
Abstract
Listeria monocytogenes is motile at 22°C and non-motile at 37°C. In contrast, expression of L. monocytogenes virulence factors is low at 22°C and up-regulated at 37°C. Here, we studied a character of L. monocytogenes near surface swimming (NSS) motility and its effects on adhesion patterns and invasion into epithelial cells. L. monocytogenes and its saprophytic counterpart L. innocua both grown at 22°C showed similar NSS characteristics including individual velocities, trajectory lengths, residence times, and an asymmetric distribution of velocity directions. Similar NSS patterns correlated with similar adhesion patterns. Motile bacteria, including both pathogenic and saprophytic species, showed a preference for adhering to the periphery of epithelial HEp-2 cells. In contrast, non-motile bacteria were evenly distributed across the cell surface, including areas over the nucleus. However, the uneven distribution of motile bacteria did not enhance the invasion into HEp-2 cells unless virulence factor production was up-regulated by the transient shift of the culture to 37°C. Motile L. monocytogenes grown overnight at 22°C and then shifted to 37°C for 2 h expressed invasion factors at the same level and invaded human cells up to five times more efficiently comparatively with non-motile bacteria grown overnight at 37°C. Taken together, obtained results demonstrated that (i) NSS motility and correspondent peripheral location over the cell surface did not depend on L. monocytogenes virulence traits; (ii) motility improved L. monocytogenes invasion into human HEp-2 cells within a few hours after the transition from the ambient temperature to the human body temperature.
Collapse
Affiliation(s)
- Mariam M. Abdulkadieva
- Department of Infections with Natural Foci, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
- Department of Dusty Plasmas, Joint Institute of High Temperatures RAS, Moscow, Russia
- Institute of Biochemical Technology and Nanotechnology, People’s Friendship University RUDN, Moscow, Russia
| | - Elena V. Sysolyatina
- Department of Infections with Natural Foci, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
| | - Elena V. Vasilieva
- Department of Infections with Natural Foci, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
- Department of Dusty Plasmas, Joint Institute of High Temperatures RAS, Moscow, Russia
| | - Veronika V. Litvinenko
- Department of Infections with Natural Foci, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
- Department of Dusty Plasmas, Joint Institute of High Temperatures RAS, Moscow, Russia
| | - Egor V. Kalinin
- Department of Infections with Natural Foci, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
- Institute of Biochemical Technology and Nanotechnology, People’s Friendship University RUDN, Moscow, Russia
| | - Vladimir G. Zhukhovitsky
- Department of Bacterial Infections, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
- Russian Medical Academy of Continuing Professional Education (RMANPO), Ministry of Public Health, Moscow, Russia
| | - Natalia V. Shevlyagina
- Department of Bacterial Infections, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
| | - Svetlana G. Andreevskaya
- Department of Bacterial Infections, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
| | - Yaroslav M. Stanishevskyi
- Institute of Biochemical Technology and Nanotechnology, People’s Friendship University RUDN, Moscow, Russia
| | - Mikhail M. Vasiliev
- Department of Dusty Plasmas, Joint Institute of High Temperatures RAS, Moscow, Russia
| | - Oleg F. Petrov
- Department of Dusty Plasmas, Joint Institute of High Temperatures RAS, Moscow, Russia
| | - Svetlana A. Ermolaeva
- Department of Infections with Natural Foci, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
| |
Collapse
|
42
|
Wang L, Zhu T. Strong Opponent of Walnut Anthracnose- Bacillus velezensis and Its Transcriptome Analysis. Microorganisms 2023; 11:1885. [PMID: 37630445 PMCID: PMC10456653 DOI: 10.3390/microorganisms11081885] [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: 06/21/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Walnut is a significant economic tree species worldwide. Walnut anthracnose, caused by the pathogen Colletotrichum gloeosporioides, greatly reduces walnut production and economic benefits. Our study showed that Bacillus velezensis effectively halted the growth of C. gloeosporioides, inducing noticeable abnormalities such as hyphal breakage and distortion, thereby curtailing the pathogen's virulence. A 50-100 times dilution of B. velezensis fermentation broth, applied every two to three days, served as an efficient protective layer for walnut leaves and fruits against C. gloeosporioides infection. Transcriptomic analysis of B. velezensis unveiled its dynamic response against C. gloeosporioides. On the second day, B. velezensis upregulated a significant number of differentially expressed genes related to the synthesis of metabolic products, amino acid biosynthesis, and motility. On the fourth day, continuous synthesis of metabolic products and amino acids, along with differential expression of spore-related genes, was observed. By the sixth day, the focus shifted towards environmental adaptation and carbon source utilization. Throughout the process, B. velezensis likely employed strategies such as the release of metabolic products, increased chemotaxis, and nutrient competition to exert its antagonistic effect on C. gloeosporioides. Fluorescence quantitative results showed that 15 primer pairs were up-regulated and 15 were down-regulated, with a 100% similarity rate to transcriptome sequencing results, confirming their authenticity. These findings provided a foundation for the widespread application of B. velezensis as a biocontrol agent in agriculture and forestry.
Collapse
Affiliation(s)
| | - Tianhui Zhu
- College of Forestry, Sichuan Agricultural University, Yaan 625000, China
| |
Collapse
|
43
|
Partridge JD, Dufour Y, Hwang Y, Harshey RM. Flagellar motor remodeling during swarming requires FliL. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.14.549092. [PMID: 37503052 PMCID: PMC10370021 DOI: 10.1101/2023.07.14.549092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
FliL is an essential component of the flagellar machinery in some bacteria, but a conditional one in others. The conditional role is for optimal swarming in some bacteria. During swarming, physical forces associated with movement on a surface are expected to exert a higher load on the flagellum, requiring more motor torque to move. Bacterial physiology and morphology are also altered during swarming to cope with the challenges of surface navigation. FliL was reported to enhance motor output in several bacteria and observed to assemble as a ring around ion-conducting stators that power the motor. In this study we identify a common new function for FliL in diverse bacteria - Escherichia coli, Bacillus subtilis and Proteus mirabilis . During swarming, all these bacteria show increased cell speed and a skewed motor bias that suppresses cell tumbling. We demonstrate that these altered motor parameters, or 'motor remodeling', require FliL. Both swarming and motor remodeling can be restored in an E. coli fliL mutant by complementation with fliL genes from P. mirabilis and B. subtilis , showing conservation of swarming-associated FliL function across phyla. In addition, we demonstrate that the strong interaction we reported earlier between FliL and the flagellar MS-ring protein FliF is confined to the RBM-3 domain of FliF that links the periplasmic rod to the cytoplasmic C-ring. This interaction may explain several phenotypes associated with the absence of FliL.
Collapse
Affiliation(s)
- Jonathan D Partridge
- Department of Molecular Biosciences and the LaMontagne Center for Infectious Diseases The University of Texas at Austin, Austin, Texas, 78712, USA
| | - Yann Dufour
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - YuneSahng Hwang
- Department of Molecular Biosciences and the LaMontagne Center for Infectious Diseases The University of Texas at Austin, Austin, Texas, 78712, USA
| | - Rasika M Harshey
- Department of Molecular Biosciences and the LaMontagne Center for Infectious Diseases The University of Texas at Austin, Austin, Texas, 78712, USA
| |
Collapse
|
44
|
Gonzales M, Plener L, Armengaud J, Armstrong N, Chabrière É, Daudé D. Lactonase-mediated inhibition of quorum sensing largely alters phenotypes, proteome, and antimicrobial activities in Burkholderia thailandensis E264. Front Cell Infect Microbiol 2023; 13:1190859. [PMID: 37333853 PMCID: PMC10272358 DOI: 10.3389/fcimb.2023.1190859] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Burkholderia thailandensis is a study model for Burkholderia pseudomallei, a highly virulent pathogen, known to be the causative agent of melioidosis and a potential bioterrorism agent. These two bacteria use an (acyl-homoserine lactone) AHL-mediated quorum sensing (QS) system to regulate different behaviors including biofilm formation, secondary metabolite productions, and motility. Methods Using an enzyme-based quorum quenching (QQ) strategy, with the lactonase SsoPox having the best activity on B. thailandensis AHLs, we evaluated the importance of QS in B. thailandensis by combining proteomic and phenotypic analyses. Results We demonstrated that QS disruption largely affects overall bacterial behavior including motility, proteolytic activity, and antimicrobial molecule production. We further showed that QQ treatment drastically decreases B. thailandensis bactericidal activity against two bacteria (Chromobacterium violaceum and Staphylococcus aureus), while a spectacular increase in antifungal activity was observed against fungi and yeast (Aspergillus niger, Fusarium graminearum and Saccharomyces cerevisiae). Discussion This study provides evidence that QS is of prime interest when it comes to understanding the virulence of Burkholderia species and developing alternative treatments.
Collapse
Affiliation(s)
- Mélanie Gonzales
- Aix Marseille Université, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
- Gene&GreenTK, Marseille, France
| | | | - Jean Armengaud
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Bagnols-sur-Cèze, France
| | | | - Éric Chabrière
- Aix Marseille Université, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | | |
Collapse
|
45
|
Rossi P, Xing Q, Bini E, Portaliou AG, Clay MC, Warren EM, Khanra NK, Economou A, Kalodimos CG. Chaperone Recycling in Late-Stage Flagellar Assembly. J Mol Biol 2023; 435:167954. [PMID: 37330284 PMCID: PMC10471782 DOI: 10.1016/j.jmb.2023.167954] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 06/19/2023]
Abstract
The flagellum is a sophisticated nanomachine responsible for motility in Gram-negative bacteria. Flagellar assembly is a strictly choreographed process, in which the motor and export gate are formed first, followed by the extracellular propeller structure. Extracellular flagellar components are escorted to the export gate by dedicated molecular chaperones for secretion and self-assembly at the apex of the emerging structure. The detailed mechanisms of chaperone-substrate trafficking at the export gate remain poorly understood. Here, we structurally characterized the interaction of Salmonella enterica late-stage flagellar chaperones FliT and FlgN with the export controller protein FliJ. Previous studies showed that FliJ is absolutely required for flagellar assembly since its interaction with chaperone-client complexes controls substrate delivery to the export gate. Our biophysical and cell-based data show that FliT and FlgN bind FliJ cooperatively, with high affinity and on specific sites. Chaperone binding completely disrupts the FliJ coiled-coil structure and alters its interactions with the export gate. We propose that FliJ aids the release of substrates from the chaperone and forms the basis of chaperone recycling during late-stage flagellar assembly.
Collapse
Affiliation(s)
- Paolo Rossi
- Deparment of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States
| | - Qiong Xing
- State Key Laboratory of Bio-catalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan 430074, China
| | - Elisabetta Bini
- Deparment of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States
| | - Athina G Portaliou
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Mary C Clay
- Deparment of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States
| | - Eric M Warren
- Deparment of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States
| | - Nandish K Khanra
- Deparment of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States
| | - Anastassios Economou
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Charalampos G Kalodimos
- Deparment of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States.
| |
Collapse
|
46
|
Uljanovas D, Gölz G, Fleischmann S, Kudirkiene E, Kasetiene N, Grineviciene A, Tamuleviciene E, Aksomaitiene J, Alter T, Malakauskas M. Genomic Characterization of Arcobacter butzleri Strains Isolated from Various Sources in Lithuania. Microorganisms 2023; 11:1425. [PMID: 37374927 DOI: 10.3390/microorganisms11061425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/09/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Arcobacter (A.) butzleri, the most widespread species within the genus Arcobacter, is considered as an emerging pathogen causing gastroenteritis in humans. Here, we performed a comparative genome-wide analysis of 40 A. butzleri strains from Lithuania to determine the genetic relationship, pangenome structure, putative virulence, and potential antimicrobial- and heavy-metal-resistance genes. Core genome single nucleotide polymorphism (cgSNP) analysis revealed low within-group variability (≤4 SNPs) between three milk strains (RCM42, RCM65, RCM80) and one human strain (H19). Regardless of the type of input (i.e., cgSNPs, accessory genome, virulome, resistome), these strains showed a recurrent phylogenetic and hierarchical grouping pattern. A. butzleri demonstrated a relatively large and highly variable accessory genome (comprising of 6284 genes with around 50% of them identified as singletons) that only partially correlated to the isolation source. Downstream analysis of the genomes resulted in the detection of 115 putative antimicrobial- and heavy-metal-resistance genes and 136 potential virulence factors that are associated with the induction of infection in host (e.g., cadF, degP, iamA), survival and environmental adaptation (e.g., flagellar genes, CheA-CheY chemotaxis system, urease cluster). This study provides additional knowledge for a better A. butzleri-related risk assessment and highlights the need for further genomic epidemiology studies in Lithuania and other countries.
Collapse
Affiliation(s)
- Dainius Uljanovas
- Department of Food Safety and Quality, Faculty of Veterinary Medicine, Veterinary Academy, Lithuanian University of Health Sciences, Tilzes St. 18, LT-47181 Kaunas, Lithuania
| | - Greta Gölz
- Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Königsweg 69, 14163 Berlin, Germany
| | - Susanne Fleischmann
- Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Königsweg 69, 14163 Berlin, Germany
| | - Egle Kudirkiene
- Statens Serum Institut, Artillerivej 5, DK-2300 Copenhagen, Denmark
| | - Neringa Kasetiene
- Department of Food Safety and Quality, Faculty of Veterinary Medicine, Veterinary Academy, Lithuanian University of Health Sciences, Tilzes St. 18, LT-47181 Kaunas, Lithuania
| | - Audrone Grineviciene
- Kaunas Clinical Hospital Microbiology Laboratory, Medical Academy, Lithuanian University of Health Sciences, Josvainiu St. 2, LT-47144 Kaunas, Lithuania
| | - Egle Tamuleviciene
- Department of Pediatrics, Medical Academy, Lithuanian University of Health Sciences, Eiveniu St. 2, LT-50161 Kaunas, Lithuania
| | - Jurgita Aksomaitiene
- Department of Food Safety and Quality, Faculty of Veterinary Medicine, Veterinary Academy, Lithuanian University of Health Sciences, Tilzes St. 18, LT-47181 Kaunas, Lithuania
| | - Thomas Alter
- Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Königsweg 69, 14163 Berlin, Germany
| | - Mindaugas Malakauskas
- Department of Food Safety and Quality, Faculty of Veterinary Medicine, Veterinary Academy, Lithuanian University of Health Sciences, Tilzes St. 18, LT-47181 Kaunas, Lithuania
| |
Collapse
|
47
|
Ling C, Liang S, Li Y, Cao Q, Ye H, Zhang C, Dong Z, Feng D, Wang W, Zuo J. A Potential Adhesin/Invasin STM0306 Participates in Host Cell Inflammation Induced by Salmonella enterica Serovar Typhimurium. Int J Mol Sci 2023; 24:ijms24098170. [PMID: 37175877 PMCID: PMC10179656 DOI: 10.3390/ijms24098170] [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: 02/17/2023] [Revised: 04/04/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Salmonella enterica serovar typhimurium (S. Typhimurium) is a common Gram-negative foodborne pathogenic bacterium that causes gastrointestinal disease in humans and animals. It is well known that adhesins and invasins play crucial roles in the infection mechanism of S. Typhimurium. S. Typhimurium STM0306 has been denoted as a putative protein and its functions have rarely been reported. In this study, we constructed the STM0306 gene mutant strain of S. Typhimurium and purified the recombinant STM0306 from Escherichia coli. Deletion of the STM0306 gene resulted in reduced adhesion and invasion of S. Typhimurium to IPEC-J2, Caco-2, and RAW264.7 cells. In addition, STM0306 could bind to intestinal epithelial cells and induced F-actin modulation in IPEC-J2 cells. Furthermore, we found that STM0306 activated the nuclear factor kappa B (NF-κB) signaling pathway and increased the mRNA expression of pro-inflammatory cytokines such as IL-1β, TNF-α, as well as chemokine CXCL2, thus resulting in cellular inflammation in host cells. In vivo, the deletion of the STM0306 gene led to reduced pathogenicity of S. Typhimurium, as evidenced by lower fecal bacterial counts and reduced body weight loss in S. Typhimurium infected mice. In conclusion, the STM0306 of S. Typhimurium is an important adhesin/invasin involved in the pathogenic process and cellular inflammation of the host.
Collapse
Affiliation(s)
- Chong Ling
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shujie Liang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yan Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qingyun Cao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hui Ye
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Changming Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Zemin Dong
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Dingyuan Feng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Weiwei Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jianjun Zuo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
48
|
Yao B, Huang R, Zhang Z, Shi S. Diverse Virulence Attributes of Pantoea alfalfae sp. nov. CQ10 Responsible for Bacterial Leaf Blight in Alfalfa Revealed by Genomic Analysis. Int J Mol Sci 2023; 24:ijms24098138. [PMID: 37175847 PMCID: PMC10179099 DOI: 10.3390/ijms24098138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
Alfalfa is widely grown worldwide for its excellent nutritional value. Pantoea species living in alfalfa seeds can easily spread over great distances with frequent trade. However, the pathogenic properties of this dangerous hitchhiker on alfalfa have not been evaluated. Here, we identified the taxonomic status of Pantoea strain CQ10 isolated from the interior of alfalfa seeds based on the whole genome sequence. The diverse virulence attributes of strain CQ10 during host infection were characterized through pathogenicity assays and functional and genomic analyses. We report that strain CQ10 belongs to a novel species in the genus Pantoea, which was phylogenetically close to Pantoea vagans and Pantoea agglomerans. Strain CQ10 caused bacterial leaf blight of alfalfa after inoculation from the roots. We found that strain CQ10 possesses a large number of pathogenic genes involved in shaping the virulence properties during bacteria-host interactions, including motility, biofilm, type VI secretion system, and nutrient acquisition. Compared with P. vagans and P. agglomerans, the unique virulence factors of strain CQ10 were mainly involved in motility and biofilm, which were confirmed by in vitro experiments. Taken together, our results suggest that strain CQ10 is the first Pantoea species to infect alfalfa, and it possesses diverse virulence attributes among which motility and biofilm may be the best weapons.
Collapse
Affiliation(s)
- Bo Yao
- Key Laboratory of Grassland Ecosystem, Ministry of Education, College of Grassland Science, Gansu Agricultural University, Lanzhou 730070, China
- Sino-U.S. Centers for Grazing Land Ecosystem Sustainability, Ministry of Science and Technology, Lanzhou 730070, China
| | - Rong Huang
- Key Laboratory of Grassland Ecosystem, Ministry of Education, College of Grassland Science, Gansu Agricultural University, Lanzhou 730070, China
- Sino-U.S. Centers for Grazing Land Ecosystem Sustainability, Ministry of Science and Technology, Lanzhou 730070, China
| | - Zhenfen Zhang
- Key Laboratory of Grassland Ecosystem, Ministry of Education, College of Grassland Science, Gansu Agricultural University, Lanzhou 730070, China
- Sino-U.S. Centers for Grazing Land Ecosystem Sustainability, Ministry of Science and Technology, Lanzhou 730070, China
| | - Shangli Shi
- Key Laboratory of Grassland Ecosystem, Ministry of Education, College of Grassland Science, Gansu Agricultural University, Lanzhou 730070, China
- Sino-U.S. Centers for Grazing Land Ecosystem Sustainability, Ministry of Science and Technology, Lanzhou 730070, China
| |
Collapse
|
49
|
Da F, Wan X, Lin G, Jian J, Cai S. Characterization of fliR-deletion mutant Δ fliR from Vibrio alginolyticus and the evaluation as a live attenuated vaccine. Front Cell Infect Microbiol 2023; 13:1162299. [PMID: 37180437 PMCID: PMC10166871 DOI: 10.3389/fcimb.2023.1162299] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/05/2023] [Indexed: 05/16/2023] Open
Abstract
Vibrio alginolyticus is the common pathogen affecting various species of marine organisms. It has been demonstrated that fliR is a necessary virulence factor to adhere and infect their hosts for pathogenic bacteria. Frequent disease outbreaks in aquaculture have highlighted the necessity of developing effective vaccines. In the present study, in order to investigate the function of fliR in V.alginolyticus, the fliR deletion mutant ΔfliR was constructed and its biological properties were evaluated, additionally, the differences in gene expression levels between wild-type and ΔfliR were analyzed by transcriptomics. Finally, ΔfliR was used as a live attenuated vaccine to immunize grouper via the intraperitoneal route to evaluate its protective effect. Results show that fliR gene of V. alginolyticus was identified as being 783 bp in length, encoding 260 amino acids, and showing significant similarity to homologs of other Vibrio species. The fliR-deletion mutant ΔfliR of V. alginolyticus was successfully constructed, and its biological phenotype analysis showed no significant differences in growth capacity and extracellular enzyme activity compared to the wild-type. However, a substantial reduction of motility ability was detected in ΔfliR. Transcriptomic analysis revealed that the absence of fliR gene is responsible for a significantly decreased expression of flagellar genes, including flaA, flaB, fliS, flhB and fliM. The fliR-deletion mainly affects the related pathways involved in cell motility, membrane transport, signal transduction, carbohydrate metabolism, and amino acid metabolism in V. alginolyticus. The efficacy of ΔfliR as a candidate of live attenuated vaccine were evaluated by intraperitoneal injection in grouper. The ΔfliR provided the RPS (Relative protection rate) of 67.2% against V. alginolyticus in groupers. The ΔfliR efficiently stimulated antibody production with specific IgM still detected at 42 d post-vaccination, and significantly elevated the activity of antioxidant enzymes like Catalase (CAT), Superoxide dismutase (SOD), and lactate dehydrogenase (LDH) in the serum. The higher expression levels of immune-related genes were observed in the immune tissues of inoculated grouper compared to the control. In conclusion, ΔfliR effectively improved the immunity of inoculated fish. The results suggest that ΔfliR is an effective live attenuated vaccine against vibriosis in in grouper.
Collapse
Affiliation(s)
| | | | | | | | - Shuanghu Cai
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Shenzhen Institute of Guangdong Ocean University, Fisheries College of Guangdong Ocean University, Guangdong, China
| |
Collapse
|
50
|
Kuipou W, Mohamadou A. Surface tension coupled non-uniformly imposed flows modulate the activity of reproducing chemotactic bacteria in porous media. Sci Rep 2023; 13:5561. [PMID: 37019986 PMCID: PMC10076314 DOI: 10.1038/s41598-023-31753-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/16/2023] [Indexed: 04/07/2023] Open
Abstract
This paper investigates a non-homogeneous two-dimensional model for reproducing chemotactic bacteria, immersed in a porous medium that experiences non-uniformly imposed flows. It is shown that independently of the form of the fluid velocity field, the compressible/incompressible nature of the fluid significantly shifts the Turing stability-instability transition line. In dry media, Gaussian perturbations travel faster than the hyperbolic secant ones, yet the latter exhibit better stability properties. The system becomes highly unstable under strong flows and high surface tension. Approximated solutions recovered by injecting Gaussian perturbations overgrow, in addition to triggering concentric breathing features that split the medium into high and low-density domains. Secant perturbations on the other hand scatter slowly and form patterns of non-uniformly distributed peaks for strong flows and high surface tension. These results emphasize that Gaussian perturbations strongly modulate the activity of bacteria, hence can be exploited to perform fast spreading in environments with changing properties. In this sense, Gaussian profiles are better candidates to explain quick bacterial responses to external factors. Secant-type approximated solutions slowly modulate the bacterial activity, hence are better alternatives to dive into weak bacterial progressions in heterogeneous media.
Collapse
Affiliation(s)
- William Kuipou
- African Centre for Advanced Studies, P.O. Box 4477, Yaoundé, Cameroon.
- Centre for Research in Infectious Disease, P.O. Box 13591, Yaoundé, Cameroon.
| | - Alidou Mohamadou
- National Advanced School of Engineering of Maroua, University of Maroua, P.O. Box 46, Maroua, Cameroon
| |
Collapse
|