1
|
Pracser N, Zaiser A, Ciolacu L, Roch FF, Quijada NM, Thalguter S, Dzieciol M, Conrady B, Wagner M, Rychli K. The type of food influences the behaviour of Listeria monocytogenes in a food-gastrointestinal-infection model. NPJ Sci Food 2025; 9:79. [PMID: 40389416 PMCID: PMC12089613 DOI: 10.1038/s41538-025-00436-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 04/30/2025] [Indexed: 05/21/2025] Open
Abstract
Food contaminated with Listeria (L.) monocytogenes is the main source of human listeriosis, but how different food matrices affect the survival and invasion in the gastrointestinal (GI) tract is still unclear. This study examined three ready-to-eat foods - soft-cheese, smoked salmon, and sausage - using a food-GI-infection model. We observed strain-dependent growth rates, but food matrices did not significantly impact growth. However, nutrient sources altered gene expression. Passage through the GI model upregulated 23 stress genes and 29 virulence genes (e.g., clpE, hly, and plcB). L. monocytogenes survival was higher in cheese and fish compared to sausage, due to their lower buffer capacity. Invasion efficiency into Caco-2 cells was highest in fish, potentially linked to its fatty acid composition. Food matrices and GI conditions influenced the transcriptional profiles of stress-associated and virulence genes. This study highlights the significant role of food matrices in L. monocytogenes survival and infection.
Collapse
Grants
- P27920-B22 Austrian Science Fund (FWF)
- P27920-B22 Austrian Science Fund (FWF)
- 881882 The Austrian COMET-K1 competence centre for Feed and Food Quality, Safety and Innovation (FFoQSI) is funded by the Austrian federal ministries BMK, BMDW and the Austrian provinces Lower Austria, Upper Austria and Vienna within the scope of COMET.
- 881882 The Austrian COMET-K1 competence centre for Feed and Food Quality, Safety and Innovation (FFoQSI) is funded by the Austrian federal ministries BMK, BMDW and the Austrian provinces Lower Austria, Upper Austria and Vienna within the scope of COMET.
- 881882 The Austrian COMET-K1 competence centre for Feed and Food Quality, Safety and Innovation (FFoQSI) is funded by the Austrian federal ministries BMK, BMDW and the Austrian provinces Lower Austria, Upper Austria and Vienna within the scope of COMET.
- 881882 The Austrian COMET-K1 competence centre for Feed and Food Quality, Safety and Innovation (FFoQSI) is funded by the Austrian federal ministries BMK, BMDW and the Austrian provinces Lower Austria, Upper Austria and Vienna within the scope of COMET.
Collapse
Affiliation(s)
- Nadja Pracser
- Centre for Food Science and Veterinary Public Health, Clinical Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Vienna, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
| | - Andreas Zaiser
- Centre for Food Science and Veterinary Public Health, Clinical Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Luminita Ciolacu
- Centre for Food Science and Veterinary Public Health, Clinical Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Franz-Ferdinand Roch
- Centre for Food Science and Veterinary Public Health, Clinical Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Narciso M Quijada
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
- Department of Microbiology and Genetics, Institute for Agribiotechnology Research (CIALE), University of Salamanca, Salamanca, Spain
| | - Sarah Thalguter
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
| | - Monika Dzieciol
- Centre for Food Science and Veterinary Public Health, Clinical Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Beate Conrady
- Centre for Food Science and Veterinary Public Health, Clinical Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Vienna, Austria
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Copenhagen, Denmark
| | - Martin Wagner
- Centre for Food Science and Veterinary Public Health, Clinical Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Vienna, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
| | - Kathrin Rychli
- Centre for Food Science and Veterinary Public Health, Clinical Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, Vienna, Austria.
| |
Collapse
|
2
|
Yeh CF, Chen CC, Lai CC, Liu JW, Tang HJ, Su WP. Synergistic effect of repurposed mitomycin C in combination with antibiotics against Aeromonas infection: In vitro and in vivo studies. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025; 58:189-197. [PMID: 39800585 DOI: 10.1016/j.jmii.2024.12.005] [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: 06/13/2024] [Revised: 12/03/2024] [Accepted: 12/19/2024] [Indexed: 03/18/2025]
Abstract
BACKGROUND Aeromonas infections pose a significant threat associated with high mortality rates. This study investigates the potential of mitomycin C (MMC), an anticancer drug, as a novel antimicrobial agent against Aeromonas infections. METHODS We evaluated the minimum inhibitory concentrations (MICs) of MMC and antibiotics against clinical Aeromonas isolates using broth microdilution. Synergistic effects of MMC with antibiotics were determined via time-kill studies. MMC's intracellular killing effects were analyzed using a representative Aeromonas isolate. Efficacy of combined therapies was assessed in a neutropenic mouse model. MMC-induced SOS response was evaluated using cell elongation method, and RNA extraction and quantitative real-time PCR. RESULTS Combining 1/8⨯ MIC of mitomycin C (MMC) with either 1⨯ or 1/2⨯ MIC of LVX demonstrated significant synergistic effects over 24 h in vitro. In a neutropenic mouse model, the combination of MMC (2 mg/kg or 1 mg/kg) with LVX achieved survival rates of 100 % and 80 %, respectively, compared to 0 % survival with monotherapy. MMC induced marked cell elongation and division inhibition in response to escalating doses. However, the combination therapy's enhancement did not surpass the effects of individual drug treatments. Notably, combination therapy reduced recA activator levels below those observed with either drug alone, suggesting rapid bacterial cell death curtailed further expression of recA and lexA. Alternatively, extensive DNA damage may have overwhelmed bacterial DNA repair mechanisms, rendering them ineffective. CONCLUSIONS These findings suggest that MMC may serve as a potential antimicrobial agent, particularly when used in combination with antibiotics. The integration of MMC with antibiotic therapy offers a promising approach for the treatment of Aeromonas infections and holds potential for future clinical applications.
Collapse
Affiliation(s)
- Cheng-Fa Yeh
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Chi-Chung Chen
- Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Chih-Cheng Lai
- Department of Intensive Care Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Jin-Wei Liu
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Hung-Jen Tang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan.
| | - Wen-Pin Su
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
3
|
Dou X, Liu Y, Koutsoumanis K, Song C, Li Z, Zhang H, Yang F, Zhu H, Dong Q. Employing genome-wide association studies to investigate acid adaptation mechanisms in Listeria monocytogenes. Food Res Int 2024; 196:115106. [PMID: 39614575 DOI: 10.1016/j.foodres.2024.115106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 12/01/2024]
Abstract
Listeria monocytogenes is a critical foodborne pathogen known to develop adaptation traits in mildly acidic food processing environments. This study analyzed the genomic data of 49 strains derived from clinical and food sources, utilizing genome-wide association studies (GWAS) to explore the correlation between the genotypic and phenotypic traits of L. monocytogenes, thereby identifying the genetic determinants of its acid adaptation capability. The findings revealed no significant association between the collected acid adaptation genes and the bacterial growth parameters. The GWAS results indicated that numerous single nucleotide polymorphism (SNP) sites were significantly correlated with the growth parameters of L. monocytogenes in a pH = 5.0 acidic environment, whereas the associations diminished as the pH approached neutrality at pH = 6.7. Analysis and annotation of synonymous mutation loci revealed that non-synonymous mutations primarily impact function. The phenotypes pH = 5.0, ΔpH (5.0-5.5), SNPλ, and SNPμmax show the strongest associations with non-synonymous mutation loci. The genes lmo0017, lmo1173, lmo0794, and lmo2783 are significant non-synonymous mutation loci influencing acid adaptation. These genes play critical roles in intracellular pH regulation, cell wall synthesis and environmental response control, directly or indirectly affecting bacterial acid tolerance. Future research could leverage existing data combined with machine learning and causal inference methods to further dissect the genotype-phenotype causal relationships, identifying causative genetic factors that govern the acid adaptation in L. monocytogenes, providing insights for risk assessment and management strategies in food safety.
Collapse
Affiliation(s)
- Xin Dou
- University of Shanghai for Science and Technology, 200098 Shanghai, China
| | - Yangtai Liu
- University of Shanghai for Science and Technology, 200098 Shanghai, China
| | - Kostas Koutsoumanis
- Department of Food Science and Technology, Faculty of Agriculture, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Chi Song
- University of Shanghai for Science and Technology, 200098 Shanghai, China
| | - Zhuosi Li
- University of Shanghai for Science and Technology, 200098 Shanghai, China
| | - Hui Zhang
- Jiangsu Academy of Agricultural Sciences, 210014 Nanjing, China
| | - Fan Yang
- Department of Pharmacy, Renji Hospital, School of Medicine Shanghai Jiao Tong University, 200127 Shanghai, China
| | - Huajian Zhu
- University of Shanghai for Science and Technology, 200098 Shanghai, China
| | - Qingli Dong
- University of Shanghai for Science and Technology, 200098 Shanghai, China.
| |
Collapse
|
4
|
Ładziak M, Prochwicz E, Gut K, Gomza P, Jaworska K, Ścibek K, Młyńska-Witek M, Kadej-Zajączkowska K, Lillebaek EMS, Kallipolitis BH, Krawczyk-Balska A. Inactivation of lmo0946 ( sif) induces the SOS response and MGEs mobilization and silences the general stress response and virulence program in Listeria monocytogenes. Front Microbiol 2024; 14:1324062. [PMID: 38239729 PMCID: PMC10794523 DOI: 10.3389/fmicb.2023.1324062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/05/2023] [Indexed: 01/22/2024] Open
Abstract
Bacteria have evolved numerous regulatory pathways to survive in changing environments. The SOS response is an inducible DNA damage repair system that plays an indispensable role in bacterial adaptation and pathogenesis. Here we report a discovery of the previously uncharacterized protein Lmo0946 as an SOS response interfering factor (Sif) in the human pathogen Listeria monocytogenes. Functional genetic studies demonstrated that sif is indispensable for normal growth of L. monocytogenes in stress-free as well as multi-stress conditions, and sif contributes to susceptibility to β-lactam antibiotics, biofilm formation and virulence. Absence of Sif promoted the SOS response and elevated expression of mobilome genes accompanied by mobilization of the A118 prophage and ICELm-1 mobile genetic elements (MGEs). These changes were found to be associated with decreased expression of general stress response genes from the σB regulon as well as virulence genes, including the PrfA regulon. Together, this study uncovers an unexpected role of a previously uncharacterized factor, Sif, as an inhibitor of the SOS response in L. monocytogenes.
Collapse
Affiliation(s)
- Magdalena Ładziak
- Department of Molecular Microbiology, Biological and Chemical Research Centre, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Emilia Prochwicz
- Department of Molecular Microbiology, Biological and Chemical Research Centre, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Karina Gut
- Department of Molecular Microbiology, Biological and Chemical Research Centre, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Patrycja Gomza
- Department of Molecular Microbiology, Biological and Chemical Research Centre, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Karolina Jaworska
- Department of Molecular Microbiology, Biological and Chemical Research Centre, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Katarzyna Ścibek
- Department of Molecular Microbiology, Biological and Chemical Research Centre, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Marta Młyńska-Witek
- Department of Molecular Microbiology, Biological and Chemical Research Centre, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Katarzyna Kadej-Zajączkowska
- Department of Molecular Microbiology, Biological and Chemical Research Centre, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Eva M. S. Lillebaek
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Birgitte H. Kallipolitis
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Agata Krawczyk-Balska
- Department of Molecular Microbiology, Biological and Chemical Research Centre, Faculty of Biology, University of Warsaw, Warsaw, Poland
| |
Collapse
|
5
|
D'Onofrio F, Schirone M, Krasteva I, Tittarelli M, Iannetti L, Pomilio F, Torresi M, Paparella A, D'Alterio N, Luciani M. A comprehensive investigation of protein expression profiles in L. monocytogenes exposed to thermal abuse, mild acid, and salt stress conditions. Front Microbiol 2023; 14:1271787. [PMID: 37876777 PMCID: PMC10591339 DOI: 10.3389/fmicb.2023.1271787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/19/2023] [Indexed: 10/26/2023] Open
Abstract
Preventing L. monocytogenes infection is crucial for food safety, considering its widespread presence in the environment and its association with contaminated RTE foods. The pathogen's ability to persist under adverse conditions, for example, in food processing facilities, is linked to virulence and resistance mechanisms, including biofilm formation. In this study, the protein expression patterns of two L. monocytogenes 1/2a strains, grown under environmental stressors (mild acidic pH, thermal abuse, and high concentration of NaCl), were investigated. Protein identification and prediction were performed by nLC-ESI-MS/MS and nine different bioinformatic software programs, respectively. Gene enrichment analysis was carried out by STRING v11.05. A total of 1,215 proteins were identified, of which 335 were non-cytosolic proteins and 265 were immunogenic proteins. Proteomic analysis revealed differences in protein expression between L. monocytogenes strains in stressful conditions. The two strains exhibited unique protein expression profiles linked to stress response, virulence, and pathogenesis. Studying the proteomic profiles of such microorganisms provides information about adaptation and potential treatments, highlighting their genetic diversity and demonstrating the utility of bioinformatics and proteomics for a broader analysis of pathogens.
Collapse
Affiliation(s)
- Federica D'Onofrio
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise “G. Caporale”, Teramo, Italy
| | - Maria Schirone
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Ivanka Krasteva
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise “G. Caporale”, Teramo, Italy
| | - Manuela Tittarelli
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise “G. Caporale”, Teramo, Italy
| | - Luigi Iannetti
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise “G. Caporale”, Teramo, Italy
| | - Francesco Pomilio
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise “G. Caporale”, Teramo, Italy
| | - Marina Torresi
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise “G. Caporale”, Teramo, Italy
| | - Antonello Paparella
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Nicola D'Alterio
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise “G. Caporale”, Teramo, Italy
| | - Mirella Luciani
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise “G. Caporale”, Teramo, Italy
| |
Collapse
|
6
|
Myintzaw P, Pennone V, McAuliffe O, Begley M, Callanan M. Association of Virulence, Biofilm, and Antimicrobial Resistance Genes with Specific Clonal Complex Types of Listeria monocytogenes. Microorganisms 2023; 11:1603. [PMID: 37375105 DOI: 10.3390/microorganisms11061603] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Precise classification of foodborne pathogen Listeria monocytogenes is a necessity in efficient foodborne disease surveillance, outbreak detection, and source tracking throughout the food chain. In this study, a total of 150 L. monocytogenes isolates from various food products, food processing environments, and clinical sources were investigated for variations in virulence, biofilm formation, and the presence of antimicrobial resistance genes based on their Whole-Genome Sequences. Clonal complex (CC) determination based on Multi-Locus Sequence Typing (MLST) revealed twenty-eight CC-types including eight isolates representing novel CC-types. The eight isolates comprising the novel CC-types share the majority of the known (cold and acid) stress tolerance genes and are all genetic lineage II, serogroup 1/2a-3a. Pan-genome-wide association analysis by Scoary using Fisher's exact test identified eleven genes specifically associated with clinical isolates. Screening for the presence of antimicrobial and virulence genes using the ABRicate tool uncovered variations in the presence of Listeria Pathogenicity Islands (LIPIs) and other known virulence genes. Specifically, the distributions of actA, ecbA, inlF, inlJ, lapB, LIPI-3, and vip genes across isolates were found to be significantly CC-dependent while the presence of ami, inlF, inlJ, and LIPI-3 was associated with clinical isolates specifically. In addition, Roary-derived phylogenetic grouping based on Antimicrobial-Resistant Genes (AMRs) revealed that the thiol transferase (FosX) gene was present in all lineage I isolates, and the presence of the lincomycin resistance ABC-F-type ribosomal protection protein (lmo0919_fam) was also genetic-lineage-dependent. More importantly, the genes found to be specific to CC-type were consistent when a validation analysis was performed with fully assembled, high-quality complete L. monocytogenes genome sequences (n = 247) extracted from the National Centre for Biotechnology Information (NCBI) microbial genomes database. This work highlights the usefulness of MLST-based CC typing using the Whole-Genome Sequence as a tool in classifying isolates.
Collapse
Affiliation(s)
- Peter Myintzaw
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland
| | - Vincenzo Pennone
- Teagasc Food Research Centre, Moorepark, Fermoy, Co., P61 C996 Cork, Ireland
| | - Olivia McAuliffe
- Teagasc Food Research Centre, Moorepark, Fermoy, Co., P61 C996 Cork, Ireland
| | - Máire Begley
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland
| | - Michael Callanan
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland
| |
Collapse
|
7
|
Inactivation of Escherichia coli O157:H7, Salmonella Typhimurium, and Listeria monocytogenes on stainless steel by synergistic effects of tap water-based neutral electrolyzed water and lactic acid. Food Microbiol 2023; 112:104233. [PMID: 36906304 DOI: 10.1016/j.fm.2023.104233] [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: 06/07/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
Contaminated food contact surface is one of the most important transmission routes for foodborne pathogens. Stainless steel is one such food-contact surface that is widely used in food-processing environments. The present study aimed to evaluate the synergistic antimicrobial efficacy of a combination of tap water-based neutral electrolyzed water (TNEW) and lactic acid (LA) against the foodborne pathogens Escherichia coli O157:H7, Salmonella Typhimurium, and Listeria monocytogenes on stainless steel. The results revealed that simultaneous treatment with TNEW (ACC of 4.60 mg/L) and 0.1% LA (TNEW-LA) for 5 min resulted in 4.99-, 4.34-, and >5.4- log CFU/cm2 reductions in E. coli O157:H7, S. Typhimurium, and L. monocytogenes on stainless steel, respectively. Of these, 4.00-, 3.57-, and >4.76-log CFU/cm2 reductions in E. coli O157:H7, S. Typhimurium, and L. monocytogenes, respectively were exclusively attributed to the synergistic action of the combined treatments after factoring out the reductions due to individual treatments. Furthermore, five mechanistic investigations revealed that the key mechanisms underlying the synergistic antibacterial effect of TNEW-LA were reactive oxygen species (ROS) production, cell membrane damage resulting from membrane lipid oxidation, DNA damage, and inactivation of intracellular enzymes. Overall, our findings suggest that the TNEW-LA combination treatment could be effectively used in the sanitization of food processing environments, especially the food contact surfaces, to control major pathogens and enhance food safety.
Collapse
|
8
|
Fernández-Gómez P, Cobo-Díaz JF, Oliveira M, González-Raurich M, Alvarez-Ordóñez A, Prieto M, Walsh JL, Sivertsvik M, Noriega-Fernández E, López M. Susceptibility and transcriptomic response to plasma-activated water of Listeria monocytogenes planktonic and sessile cells. Food Microbiol 2023; 113:104252. [PMID: 37098419 DOI: 10.1016/j.fm.2023.104252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 03/09/2023]
Abstract
Plasma-Activated Water (PAW) was generated from tap water using a surface dielectric barrier discharge at different discharge power (26 and 36 W) and activation time (5 and 30 min). The inactivation of a three-strain Listeria monocytogenes cocktail in planktonic and biofilm state was evaluated. PAW generated at 36 W-30 min showed the lowest pH and the highest hydrogen peroxide, nitrates, nitrites contents and effectiveness against cells on planktonic state, resulting in 4.6 log reductions after a 15-min treatment. Although the antimicrobial activity in biofilms formed on stainless steel and on polystyrene was lower, increasing the exposure time to 30 min allowed an inactivation >4.5 log cycles. The mechanisms of action of PAW were investigated using chemical solutions that mimic its physico-chemical characteristics and also RNA-seq analysis. The main transcriptomic changes affected carbon metabolism, virulence and general stress response genes, with several overexpressed genes belonging to the cobalamin-dependent gene cluster.
Collapse
|
9
|
Lima-Noronha MA, Fonseca DLH, Oliveira RS, Freitas RR, Park JH, Galhardo RS. Sending out an SOS - the bacterial DNA damage response. Genet Mol Biol 2022; 45:e20220107. [PMID: 36288458 PMCID: PMC9578287 DOI: 10.1590/1678-4685-gmb-2022-0107] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 07/15/2022] [Indexed: 11/04/2022] Open
Abstract
The term “SOS response” was first coined by Radman in 1974, in an intellectual effort to put together the data suggestive of a concerted gene expression program in cells undergoing DNA damage. A large amount of information about this cellular response has been collected over the following decades. In this review, we will focus on a few of the relevant aspects about the SOS response: its mechanism of control and the stressors which activate it, the diversity of regulated genes in different species, its role in mutagenesis and evolution including the development of antimicrobial resistance, and its relationship with mobile genetic elements.
Collapse
Affiliation(s)
- Marco A. Lima-Noronha
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brazil
| | - Douglas L. H. Fonseca
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brazil
| | - Renatta S. Oliveira
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brazil
| | - Rúbia R. Freitas
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brazil
| | - Jung H. Park
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brazil
| | - Rodrigo S. Galhardo
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brazil
| |
Collapse
|
10
|
Bar N, Nikparvar B, Jayavelu ND, Roessler FK. Constrained Fourier estimation of short-term time-series gene expression data reduces noise and improves clustering and gene regulatory network predictions. BMC Bioinformatics 2022; 23:330. [PMID: 35945515 PMCID: PMC9364503 DOI: 10.1186/s12859-022-04839-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 07/12/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Biological data suffers from noise that is inherent in the measurements. This is particularly true for time-series gene expression measurements. Nevertheless, in order to to explore cellular dynamics, scientists employ such noisy measurements in predictive and clustering tools. However, noisy data can not only obscure the genes temporal patterns, but applying predictive and clustering tools on noisy data may yield inconsistent, and potentially incorrect, results. RESULTS To reduce the noise of short-term (< 48 h) time-series expression data, we relied on the three basic temporal patterns of gene expression: waves, impulses and sustained responses. We constrained the estimation of the true signals to these patterns by estimating the parameters of first and second-order Fourier functions and using the nonlinear least-squares trust-region optimization technique. Our approach lowered the noise in at least 85% of synthetic time-series expression data, significantly more than the spline method ([Formula: see text]). When the data contained a higher signal-to-noise ratio, our method allowed downstream network component analyses to calculate consistent and accurate predictions, particularly when the noise variance was high. Conversely, these tools led to erroneous results from untreated noisy data. Our results suggest that at least 5-7 time points are required to efficiently de-noise logarithmic scaled time-series expression data. Investing in sampling additional time points provides little benefit to clustering and prediction accuracy. CONCLUSIONS Our constrained Fourier de-noising method helps to cluster noisy gene expression and interpret dynamic gene networks more accurately. The benefit of noise reduction is large and can constitute the difference between a successful application and a failing one.
Collapse
Affiliation(s)
- Nadav Bar
- grid.5947.f0000 0001 1516 2393Department of Chemical Engineering, Norwegian University of Science and Technology (NTNU), Sem Sælandsvei 4, Trondheim, NO-7491 Norway
| | - Bahareh Nikparvar
- grid.5947.f0000 0001 1516 2393Department of Chemical Engineering, Norwegian University of Science and Technology (NTNU), Sem Sælandsvei 4, Trondheim, NO-7491 Norway
| | - Naresh Doni Jayavelu
- grid.34477.330000000122986657Division of Medical Genetics, Department of Medicine, University of Washington Seattle, Seattle, WA 98195-7720 USA
| | - Fabienne Krystin Roessler
- grid.5947.f0000 0001 1516 2393Department of Chemical Engineering, Norwegian University of Science and Technology (NTNU), Sem Sælandsvei 4, Trondheim, NO-7491 Norway
| |
Collapse
|
11
|
Myintzaw P, Pennone V, McAuliffe O, Begley M, Callanan M. Correlation of organic acid tolerance and genotypic characteristics of Listeria monocytogenes food and clinical isolates. Food Microbiol 2022; 104:104004. [DOI: 10.1016/j.fm.2022.104004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 01/05/2022] [Accepted: 02/11/2022] [Indexed: 11/29/2022]
|
12
|
Balogh D, Eckel K, Fetzer C, Sieber SA. Listeria monocytogenes utilizes the ClpP1/2 proteolytic machinery for fine-tuned substrate degradation at elevated temperatures. RSC Chem Biol 2022; 3:955-971. [PMID: 35866172 PMCID: PMC9257651 DOI: 10.1039/d2cb00077f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/30/2022] [Indexed: 11/21/2022] Open
Abstract
Listeria monocytogenes exhibits two ClpP isoforms (ClpP1/ClpP2) which assemble into a heterooligomeric complex with enhanced proteolytic activity. Herein, we demonstrate that the formation of this complex depends on temperature and reaches a maximum ratio of about 1 : 1 at 30 °C, while almost no complex formation occurred below 4 °C. In order to decipher the role of the two isoforms at elevated temperatures, we constructed L. monocytogenes ClpP1, ClpP2 and ClpP1/2 knockout strains and analyzed their protein regulation in comparison to the wild type (WT) strain via whole proteome mass-spectrometry (MS) at 37 °C and 42 °C. While the ΔclpP1 strain only altered the expression of very few proteins, the ΔclpP2 and ΔclpP1/2 strains revealed the dysregulation of many proteins at both temperatures. These effects were corroborated by crosslinking co-immunoprecipitation MS analysis. Thus, while ClpP1 serves as a mere enhancer of protein degradation in the heterocomplex, ClpP2 is essential for ClpX binding and functions as a gatekeeper for substrate entry. Applying an integrated proteomic approach combining whole proteome and co-immunoprecipitation datasets, several putative ClpP2 substrates were identified in the context of different temperatures and discussed with regards to their function in cellular pathways such as the SOS response. Unlike most bacteria, L. monocytogenes encodes 2 isoforms of Caseinolytic Protease P. Balogh et al. show that both proteins form a heterocomplex temperature-dependently and find protein substrate candidates with an integrated proteomic approach.![]()
Collapse
Affiliation(s)
- Dóra Balogh
- Center for Functional Protein Assemblies (CPA), Department of Chemistry, Chair of Organic Chemistry II, Technische Universität München 85748 Garching Germany
| | - Konstantin Eckel
- Center for Functional Protein Assemblies (CPA), Department of Chemistry, Chair of Organic Chemistry II, Technische Universität München 85748 Garching Germany
| | - Christian Fetzer
- Center for Functional Protein Assemblies (CPA), Department of Chemistry, Chair of Organic Chemistry II, Technische Universität München 85748 Garching Germany
| | - Stephan A Sieber
- Center for Functional Protein Assemblies (CPA), Department of Chemistry, Chair of Organic Chemistry II, Technische Universität München 85748 Garching Germany
| |
Collapse
|
13
|
Chmielowska C, Korsak D, Chapkauskaitse E, Decewicz P, Lasek R, Szuplewska M, Bartosik D. Plasmidome of Listeria spp.-The repA-Family Business. Int J Mol Sci 2021; 22:ijms221910320. [PMID: 34638661 PMCID: PMC8508797 DOI: 10.3390/ijms221910320] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022] Open
Abstract
Bacteria of the genus Listeria (phylum Firmicutes) include both human and animal pathogens, as well as saprophytic strains. A common component of Listeria spp. genomes are plasmids, i.e., extrachromosomal replicons that contribute to gene flux in bacteria. This study provides an in-depth insight into the structure, diversity and evolution of plasmids occurring in Listeria strains inhabiting various environments under different anthropogenic pressures. Apart from the components of the conserved plasmid backbone (providing replication, stable maintenance and conjugational transfer functions), these replicons contain numerous adaptive genes possibly involved in: (i) resistance to antibiotics, heavy metals, metalloids and sanitizers, and (ii) responses to heat, oxidative, acid and high salinity stressors. Their genomes are also enriched by numerous transposable elements, which have influenced the plasmid architecture. The plasmidome of Listeria is dominated by a group of related replicons encoding the RepA replication initiation protein. Detailed comparative analyses provide valuable data on the level of conservation of these replicons and their role in shaping the structure of the Listeria pangenome, as well as their relationship to plasmids of other genera of Firmicutes, which demonstrates the range and direction of flow of genetic information in this important group of bacteria.
Collapse
Affiliation(s)
- Cora Chmielowska
- Department of Bacterial Genetics, Institute of Microbiology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland; (E.C.); (R.L.); (M.S.)
- Correspondence: (C.C.); (D.B.)
| | - Dorota Korsak
- Department of Molecular Microbiology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland;
| | - Elvira Chapkauskaitse
- Department of Bacterial Genetics, Institute of Microbiology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland; (E.C.); (R.L.); (M.S.)
| | - Przemysław Decewicz
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland;
| | - Robert Lasek
- Department of Bacterial Genetics, Institute of Microbiology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland; (E.C.); (R.L.); (M.S.)
| | - Magdalena Szuplewska
- Department of Bacterial Genetics, Institute of Microbiology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland; (E.C.); (R.L.); (M.S.)
| | - Dariusz Bartosik
- Department of Bacterial Genetics, Institute of Microbiology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland; (E.C.); (R.L.); (M.S.)
- Correspondence: (C.C.); (D.B.)
| |
Collapse
|
14
|
Role of the SOS Response in the Generation of Antibiotic Resistance In Vivo. Antimicrob Agents Chemother 2021; 65:e0001321. [PMID: 33875437 DOI: 10.1128/aac.00013-21] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The SOS response to DNA damage is a conserved stress response in Gram-negative and Gram-positive bacteria. Although this pathway has been studied for years, its relevance is still not familiar to many working in the fields of clinical antibiotic resistance and stewardship. Under some conditions, the SOS response favors DNA repair and preserves the genetic integrity of the organism. On the other hand, the SOS response also includes induction of error-prone DNA polymerases, which can increase the rate of mutation, called the mutator phenotype or "hypermutation." As a result, mutations can occur in genes conferring antibiotic resistance, increasing the acquisition of resistance to antibiotics. Almost all of the work on the SOS response has been on bacteria exposed to stressors in vitro. In this study, we sought to quantitate the effects of SOS-inducing drugs in vivo, in comparison with the same drugs in vitro. We used a rabbit model of intestinal infection with enteropathogenic Escherichia coli strain E22. SOS-inducing drugs triggered the mutator phenotype response in vivo as well as in vitro. Exposure of E. coli strain E22 to ciprofloxacin or zidovudine, both of which induce the SOS response in vitro, resulted in increased antibiotic resistance to 3 antibiotics: rifampin, minocycline, and fosfomycin. Zinc was able to inhibit the SOS-induced emergence of antibiotic resistance in vivo, as previously observed in vitro. Our findings may have relevance in reducing the emergence of resistance to new antimicrobial drugs.
Collapse
|
15
|
Nowak J, Visnovsky SB, Pitman AR, Cruz CD, Palmer J, Fletcher GC, Flint S. Biofilm Formation by Listeria monocytogenes 15G01, a Persistent Isolate from a Seafood-Processing Plant, Is Influenced by Inactivation of Multiple Genes Belonging to Different Functional Groups. Appl Environ Microbiol 2021; 87:e02349-20. [PMID: 33741610 PMCID: PMC8117777 DOI: 10.1128/aem.02349-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 02/25/2021] [Indexed: 01/13/2023] Open
Abstract
Listeria monocytogenes is a ubiquitous foodborne pathogen that results in a high rate of mortality in sensitive and immunocompromised people. Contamination of food with L. monocytogenes is thought to occur during food processing, most often as a result of the pathogen producing a biofilm that persists in the environment and acting as the source for subsequent dispersal of cells onto food. A survey of seafood-processing plants in New Zealand identified the persistent strain 15G01, which has a high capacity to form biofilms. In this study, a transposon library of L. monocytogenes 15G01 was screened for mutants with altered biofilm formation, assessed by a crystal violet assay, to identify genes involved in biofilm formation. This screen identified 36 transposants that showed a significant change in biofilm formation compared to the wild type. The insertion sites were in 27 genes, 20 of which led to decreased biofilm formation and seven to an increase. Two insertions were in intergenic regions. Annotation of the genes suggested that they are involved in diverse cellular processes, including stress response, autolysis, transporter systems, and cell wall/membrane synthesis. Analysis of the biofilms produced by the transposants using scanning electron microscopy and fluorescence microscopy showed notable differences in the structure of the biofilms compared to the wild type. In particular, inactivation of uvrB and mltD produced coccoid-shaped cells and elongated cells in long chains, respectively, and the mgtB mutant produced a unique biofilm with a sandwich structure which was reversed to the wild-type level upon magnesium addition. The mltD transposant was successfully complemented with the wild-type gene, whereas the phenotypes were not or only partially restored for the remaining mutants.IMPORTANCE The major source of contamination of food with Listeria monocytogenes is thought to be due to biofilm formation and/or persistence in food-processing plants. By establishing as a biofilm, L. monocytogenes cells become harder to eradicate due to their increased resistance to environmental threats. Understanding the genes involved in biofilm formation and their influence on biofilm structure will help identify new ways to eliminate harmful biofilms in food processing environments. To date, multiple genes have been identified as being involved in biofilm formation by L. monocytogenes; however, the exact mechanism remains unclear. This study identified four genes associated with biofilm formation by a persistent strain. Extensive microscopic analysis illustrated the effect of the disruption of mgtB, clsA, uvrB, and mltD and the influence of magnesium on the biofilm structure. The results strongly suggest an involvement in biofilm formation for the four genes and provide a basis for further studies to analyze gene regulation to assess the specific role of these biofilm-associated genes.
Collapse
Affiliation(s)
- Jessika Nowak
- The New Zealand Institute for Plant and Food Research Limited, Auckland, New Zealand
- Institute of Food, Nutrition and Human Health, Massey University, Palmerston North, New Zealand
| | - Sandra B Visnovsky
- The New Zealand Institute for Plant and Food Research Limited, Lincoln, New Zealand
| | - Andrew R Pitman
- The Foundation for Arable Research, Christchurch, New Zealand
| | - Cristina D Cruz
- The New Zealand Institute for Plant and Food Research Limited, Auckland, New Zealand
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jon Palmer
- Institute of Food, Nutrition and Human Health, Massey University, Palmerston North, New Zealand
| | - Graham C Fletcher
- The New Zealand Institute for Plant and Food Research Limited, Auckland, New Zealand
| | - Steve Flint
- Institute of Food, Nutrition and Human Health, Massey University, Palmerston North, New Zealand
| |
Collapse
|
16
|
Nikparvar B, Andreevskaya M, Duru IC, Bucur FI, Grigore-Gurgu L, Borda D, Nicolau AI, Riedel CU, Auvinen P, Bar N. Analysis of temporal gene regulation of Listeria monocytogenes revealed distinct regulatory response modes after exposure to high pressure processing. BMC Genomics 2021; 22:266. [PMID: 33853520 PMCID: PMC8045354 DOI: 10.1186/s12864-021-07461-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/10/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The pathogen Listeria (L.) monocytogenes is known to survive heat, cold, high pressure, and other extreme conditions. Although the response of this pathogen to pH, osmotic, temperature, and oxidative stress has been studied extensively, its reaction to the stress produced by high pressure processing HPP (which is a preservation method in the food industry), and the activated gene regulatory network (GRN) in response to this stress is still largely unknown. RESULTS We used RNA sequencing transcriptome data of L. monocytogenes (ScottA) treated at 400 MPa and 8∘C, for 8 min and combined it with current information in the literature to create a transcriptional regulation database, depicting the relationship between transcription factors (TFs) and their target genes (TGs) in L. monocytogenes. We then applied network component analysis (NCA), a matrix decomposition method, to reconstruct the activities of the TFs over time. According to our findings, L. monocytogenes responded to the stress applied during HPP by three statistically different gene regulation modes: survival mode during the first 10 min post-treatment, repair mode during 1 h post-treatment, and re-growth mode beyond 6 h after HPP. We identified the TFs and their TGs that were responsible for each of the modes. We developed a plausible model that could explain the regulatory mechanism that L. monocytogenes activated through the well-studied CIRCE operon via the regulator HrcA during the survival mode. CONCLUSIONS Our findings suggest that the timely activation of TFs associated with an immediate stress response, followed by the expression of genes for repair purposes, and then re-growth and metabolism, could be a strategy of L. monocytogenes to survive and recover extreme HPP conditions. We believe that our results give a better understanding of L. monocytogenes behavior after exposure to high pressure that may lead to the design of a specific knock-out process to target the genes or mechanisms. The results can help the food industry select appropriate HPP conditions to prevent L. monocytogenes recovery during food storage.
Collapse
Affiliation(s)
- Bahareh Nikparvar
- Department of Chemical Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Ilhan C Duru
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Florentina I Bucur
- Faculty of Food Science and Engineering, Dunarea de Jos University of Galati, Galati, Romania
| | - Leontina Grigore-Gurgu
- Faculty of Food Science and Engineering, Dunarea de Jos University of Galati, Galati, Romania
| | - Daniela Borda
- Faculty of Food Science and Engineering, Dunarea de Jos University of Galati, Galati, Romania
| | - Anca I Nicolau
- Faculty of Food Science and Engineering, Dunarea de Jos University of Galati, Galati, Romania
| | - Christian U Riedel
- Institute of Microbiology and Biotechnology, Ulm University, Ulm, Germany
| | - Petri Auvinen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Nadav Bar
- Department of Chemical Engineering, Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
17
|
Alternative σ Factors Regulate Overlapping as Well as Distinct Stress Response and Metabolic Functions in Listeria monocytogenes under Stationary Phase Stress Condition. Pathogens 2021; 10:pathogens10040411. [PMID: 33915780 PMCID: PMC8066629 DOI: 10.3390/pathogens10040411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/13/2022] Open
Abstract
Listeria monocytogenes can regulate and fine-tune gene expression, to adapt to diverse stress conditions encountered during foodborne transmission. To further understand the contributions of alternative sigma (σ) factors to the regulation of L. monocytogenes gene expression, RNA-Seq was performed on L. monocytogenes strain 10403S and five isogenic mutants (four strains bearing in-frame null mutations in three out of four alternative σ factor genes, ΔCHL, ΔBHL, ΔBCL, and ΔBCH, and one strain bearing null mutations in all four genes, ΔBCHL), grown to stationary phase. Our data showed that 184, 35, 34, and 20 genes were positively regulated by σB, σL, σH, and σC (posterior probability > 0.9 and Fold Change (FC) > 5.0), respectively. Moreover, σB-dependent genes showed the highest FC (based on comparisons between the ΔCHL and the ΔBCHL strain), with 44 genes showing an FC > 100; only four σL-dependent, and no σH- or σC-dependent genes showed FC >100. While σB-regulated genes identified in this study are involved in stress-associated functions and metabolic pathways, σL appears to largely regulate genes involved in a few specific metabolic pathways, including positive regulation of operons encoding phosphoenolpyruvate (PEP)-dependent phosphotransferase systems (PTSs). Overall, our data show that (i) σB and σL directly and indirectly regulate genes involved in several energy metabolism-related functions; (ii) alternative σ factors are involved in complex regulatory networks and appear to have epistatic effects in stationary phase cells; and (iii) σB regulates multiple stress response pathways, while σL and σH positively regulate a smaller number of specific pathways.
Collapse
|
18
|
Kılıç S, Sánchez-Osuna M, Collado-Padilla A, Barbé J, Erill I. Flexible comparative genomics of prokaryotic transcriptional regulatory networks. BMC Genomics 2020; 21:466. [PMID: 33327941 PMCID: PMC7739468 DOI: 10.1186/s12864-020-06838-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/16/2020] [Indexed: 11/25/2022] Open
Abstract
Background Comparative genomics methods enable the reconstruction of bacterial regulatory networks using available experimental data. In spite of their potential for accelerating research into the composition and evolution of bacterial regulons, few comparative genomics suites have been developed for the automated analysis of these regulatory systems. Available solutions typically rely on precomputed databases for operon and ortholog predictions, limiting the scope of analyses to processed complete genomes, and several key issues such as the transfer of experimental information or the integration of regulatory information in a probabilistic setting remain largely unaddressed. Results Here we introduce CGB, a flexible platform for comparative genomics of prokaryotic regulons. CGB has few external dependencies and enables fully customized analyses of newly available genome data. The platform automates the merging of experimental information and uses a gene-centered, Bayesian framework to generate and integrate easily interpretable results. We demonstrate its flexibility and power by analyzing the evolution of type III secretion system regulation in pathogenic Proteobacteria and by characterizing the SOS regulon of a new bacterial phylum, the Balneolaeota. Conclusions Our results demonstrate the applicability of the CGB pipeline in multiple settings. CGB’s ability to automatically integrate experimental information from multiple sources and use complete and draft genomic data, coupled with its non-reliance on precomputed databases and its easily interpretable display of gene-centered posterior probabilities of regulation provide users with an unprecedented level of flexibility in launching comparative genomics analyses of prokaryotic transcriptional regulatory networks. The analyses of type III secretion and SOS response regulatory networks illustrate instances of convergent and divergent evolution of these regulatory systems, showcasing the power of formal ancestral state reconstruction at inferring the evolutionary history of regulatory networks.
Collapse
Affiliation(s)
- Sefa Kılıç
- University of Maryland Baltimore County, Baltimore, MD, 21250, USA
| | | | | | - Jordi Barbé
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Ivan Erill
- University of Maryland Baltimore County, Baltimore, MD, 21250, USA.
| |
Collapse
|
19
|
Arcari T, Feger ML, Guerreiro DN, Wu J, O’Byrne CP. Comparative Review of the Responses of Listeria monocytogenes and Escherichia coli to Low pH Stress. Genes (Basel) 2020; 11:genes11111330. [PMID: 33187233 PMCID: PMC7698193 DOI: 10.3390/genes11111330] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/03/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
Acidity is one of the principal physicochemical factors that influence the behavior of microorganisms in any environment, and their response to it often determines their ability to grow and survive. Preventing the growth and survival of pathogenic bacteria or, conversely, promoting the growth of bacteria that are useful (in biotechnology and food production, for example), might be improved considerably by a deeper understanding of the protective responses that these microorganisms deploy in the face of acid stress. In this review, we survey the molecular mechanisms used by two unrelated bacterial species in their response to low pH stress. We chose to focus on two well-studied bacteria, Escherichia coli (phylum Proteobacteria) and Listeria monocytogenes (phylum Firmicutes), that have both evolved to be able to survive in the mammalian gastrointestinal tract. We review the mechanisms that these species use to maintain a functional intracellular pH as well as the protective mechanisms that they deploy to prevent acid damage to macromolecules in the cells. We discuss the mechanisms used to sense acid in the environment and the regulatory processes that are activated when acid is encountered. We also highlight the specific challenges presented by organic acids. Common themes emerge from this comparison as well as unique strategies that each species uses to cope with acid stress. We highlight some of the important research questions that still need to be addressed in this fascinating field.
Collapse
|
20
|
Memar MY, Yekani M, Celenza G, Poortahmasebi V, Naghili B, Bellio P, Baghi HB. The central role of the SOS DNA repair system in antibiotics resistance: A new target for a new infectious treatment strategy. Life Sci 2020; 262:118562. [PMID: 33038378 DOI: 10.1016/j.lfs.2020.118562] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/15/2020] [Accepted: 10/01/2020] [Indexed: 01/19/2023]
Abstract
Bacteria have a considerable ability and potential to acquire resistance against antimicrobial agents by acting diverse mechanisms such as target modification or overexpression, multidrug transporter systems, and acquisition of drug hydrolyzing enzymes. Studying the mechanisms of bacterial cell physiology is mandatory for the development of novel strategies to control the antimicrobial resistance phenomenon, as well as for the control of infections in clinics. The SOS response is a cellular DNA repair mechanism that has an essential role in the bacterial biologic process involved in resistance to antibiotics. The activation of the SOS network increases the resistance and tolerance of bacteria to stress and, as a consequence, to antimicrobial agents. Therefore, SOS can be an applicable target for the discovery of new antimicrobial drugs. In the present review, we focus on the central role of SOS response in bacterial resistance mechanisms and its potential as a new target for control of resistant pathogens.
Collapse
Affiliation(s)
- Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mina Yekani
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Giuseppe Celenza
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Vahdat Poortahmasebi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrooz Naghili
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pierangelo Bellio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
The recA gene is crucial to mediate colonization of Bacillus cereus 905 on wheat roots. Appl Microbiol Biotechnol 2020; 104:9251-9265. [PMID: 32970180 DOI: 10.1007/s00253-020-10915-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/07/2020] [Accepted: 09/17/2020] [Indexed: 10/23/2022]
Abstract
Bacillus cereus 905, one of the plant growth-promoting rhizobacteria (PGPRs), is capable of colonizing wheat roots in a large population size. From previous studies, we learned that the sodA2-encoding manganese-containing superoxide dismutase (MnSOD2) is important for B. cereus 905 to survive in wheat rhizosphere. In this investigation, we demonstrated that deletion of the recA gene, which codes for the recombinase A, significantly reduced MnSOD2 expression at both the mRNA and the protein levels. Through comparison with the wild-type, the ∆recA showed a dramatic decrease in cell survival after exposure to 50 μM paraquat or 15 mM H2O2. Evidence indicated that the recA gene of B. cereus 905 also notably regulated nutrition utilization efficiency, biofilm formation, and swarming motility. The root colonization examination showed that the ∆recA had a 1000- to 2500-fold reduction in colonization on wheat roots, suggesting that RecA plays an indispensable role in effective colonization on wheat roots by B. cereus 905. Taken together, the recA gene positively regulates MnSOD2 production and nutrition utilization and protects B. cereus 905 cells against paraquat and H2O2. Besides, biofilm formation and swarming motility of B. cereus 905 are promoted by RecA. Finally, RecA significantly contributes to wheat root colonization of B. cereus 905. Our results showed the important role of RecA during physiological processes in B. cereus 905, especially for colonization on wheat roots. Our findings will point out a research direction to study the colonization mechanisms of B. cereus 905 in the future and provide potential effective strategy to enhance the biocontrol efficacy of PGPR strains. KEY POINTS : • RecA plays an indispensable role in root colonization of B. cereus.
Collapse
|
22
|
Zhang J, Wang S, Abee T, van der Veen S. Role of Base Excision Repair in Listeria monocytogenes DNA Stress Survival During Infections. J Infect Dis 2020; 223:721-732. [PMID: 32644146 DOI: 10.1093/infdis/jiaa412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/07/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Base excision repair (BER), consisting mostly of lesion-specific DNA glycosylases and apurinic/apyrimidinic (AP) endonucleases, is one of the most important DNA repair mechanisms for repair of single nucleobase lesions generated by reactive oxygen and nitrogen species as part of an immune response against bacterial infections. However, few studies have addressed the contribution of BER to bacterial virulence and Listeria monocytogenes BER has thus far remained completely uncharacterized. METHODS Analysis of the L. monocytogenes EGDe genome identified 7 DNA glycosylases (MutM, MutY, Nth, Tag, Mpg, Ung, and Ung2) and 2 apurinic/apyrimidinic endonucleases (Xth and Nfo) as part of BER. Markerless in-frame deletion mutants were generated for all 9 genes, and mutants were tested for DNA damage survival, mutagenesis, and the ability to colonize a mouse model of infection. RESULTS Distinct lesion-specific phenotypes were identified for all deletion mutants. Importantly, the Δnth, ΔmutY, and Δnfo mutants were significantly attenuated for virulence in the mouse model and showed much lower colonization of the liver and spleen or were unable to compete with the wild-type strain during in vivo competition assays. CONCLUSIONS Our results highlight the importance of BER for L. monocytogenes virulence and survival of DNA-damaging insults during host colonization.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Microbiology and Parasitology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuyi Wang
- Department of Microbiology and Parasitology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tjakko Abee
- Food Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Stijn van der Veen
- Department of Microbiology and Parasitology, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
23
|
Khan F, Yu H, Kim YM. Bactericidal Activity of Usnic Acid-Chitosan Nanoparticles against Persister Cells of Biofilm-Forming Pathogenic Bacteria. Mar Drugs 2020; 18:E270. [PMID: 32443816 PMCID: PMC7281555 DOI: 10.3390/md18050270] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to prepare usnic acid (UA)-loaded chitosan (CS) nanoparticles (UA-CS NPs) and evaluate its antibacterial activity against biofilm-forming pathogenic bacteria. UA-CS NPs were prepared through simple ionic gelification of UA with CS, and further characterized using Fourier transform infrared spectroscopy, X-ray diffraction, and field-emission transmission electron microscopy. The UA-CS NPs presented a loading capacity (LC) of 5.2%, encapsulation efficiency (EE) of 24%, and a spherical shape and rough surface. The maximum release of UA was higher in pH 1.2 buffer solution as compared to that in pH 6.8 and 7.4 buffer solution. The average size and zeta potential of the UA-CS NPs was 311.5 ± 49.9 nm in diameter and +27.3 ± 0.8 mV, respectively. The newly prepared UA-CS NPs exhibited antibacterial activity against persister cells obtained from the stationary phase in batch culture, mature biofilms, and antibiotic-induced gram-positive and gram-negative pathogenic bacteria. Exposure of sub-inhibitory concentrations of UA-CS NPs to the bacterial cells resulted in a change in morphology. The present study suggests an alternative method for the application of UA into nanoparticles. Furthermore, the anti-persister activity of UA-CS NPs may be another possible strategy for the treatment of infections caused by biofilm-forming pathogenic bacteria.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Institute of Food Science, Pukyong National University, Busan 48513, Korea;
| | - Hongsik Yu
- Food Safety and Processing Research Division, National Institute of Fisheries Science, Busan 46083, Korea;
| | - Young-Mog Kim
- Institute of Food Science, Pukyong National University, Busan 48513, Korea;
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Korea
| |
Collapse
|
24
|
Ojha D, Patil KN. Molecular and functional characterization of the Listeria monocytogenes RecA protein: Insights into the homologous recombination process. Int J Biochem Cell Biol 2019; 119:105642. [PMID: 31698090 DOI: 10.1016/j.biocel.2019.105642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 10/20/2019] [Accepted: 10/31/2019] [Indexed: 12/28/2022]
Abstract
The recombinases present in the all kingdoms in nature play a crucial role in DNA metabolism processes such as replication, repair, recombination and transcription. However, till date, the role of RecA in the deadly foodborne pathogen Listeria monocytogenes remains unknown. In this study, the authors show that L. monocytogenes expresses recA more than two-fold in vivo upon exposure to the DNA damaging agents, methyl methanesulfonate and ultraviolet radiation. The purified L. monocytogenes RecA protein show robust binding to single stranded DNA. The RecA is capable of forming displacement loop and hydrolyzes ATP, whereas the mutant LmRecAK70A fails to hydrolyze ATP, showing conserved walker A and B motifs. Interestingly, L. monocytogenes RecA and LmRecAK70A perform the DNA strand transfer activity, which is the hallmark feature of RecA protein with an oligonucleotide-based substrate. Notably, L. monocytogenes RecA readily cleaves L. monocytogenes LexA, the SOS regulon and protects the presynaptic filament from the exonuclease I activity. Altogether, this study provides the first detailed characterization of L. monocytogenes RecA and presents important insights into the process of homologous recombination in the gram-positive foodborne bacteria L. monocytogenes.
Collapse
Affiliation(s)
- Debika Ojha
- Department of Protein Chemistry and Technology, Council of Scientific & Industrial Research-Central Food Technological Research Institute (CSIR-CFTRI), Mysuru, 570 020, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - K Neelakanteshwar Patil
- Department of Protein Chemistry and Technology, Council of Scientific & Industrial Research-Central Food Technological Research Institute (CSIR-CFTRI), Mysuru, 570 020, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India.
| |
Collapse
|
25
|
Horlbog JA, Stevens MJA, Stephan R, Guldimann C. Global Transcriptional Response of Three Highly Acid-Tolerant Field Strains of Listeria monocytogenes to HCl Stress. Microorganisms 2019; 7:microorganisms7100455. [PMID: 31623206 PMCID: PMC6843411 DOI: 10.3390/microorganisms7100455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/08/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
Tolerance to acid is of dual importance for the food-borne pathogen Listeria monocytogenes: acids are used as a preservative, and gastric acid is one of the first defenses within the host. There are considerable differences in the acid tolerance of strains. Here we present the transcriptomic response of acid-tolerant field strains of L. monocytogenes to HCl at pH 3.0. RNAseq revealed significant differential expression of genes involved in phosphotransferase systems, oxidative phosphorylation, cell morphology, motility, and biofilm formation. Genes in the acetoin biosynthesis pathway were upregulated, suggesting that L. monocytogenes shifts to metabolizing pyruvate to acetoin under organic acid stress. We also identified the formation of cell aggregates in microcolonies as a potential relief strategy. A motif search within the first 150 bp upstream of differentially expressed genes identified a novel potential regulatory sequence that may have a function in the regulation of virulence gene expression. Our data support a model where an excess of intracellular H+ ions is counteracted by pumping H+ out of the cytosol via cytochrome C under reduced activity of the ATP synthase. The observed morphological changes suggest that acid stress may cause cells to aggregate in biofilm microcolonies to create a more favorable microenvironment. Additionally, HCl stress in the host stomach may serve as (i) a signal to downregulate highly immunogenic flagella, and (ii) as an indicator for the imminent contact with host cells which triggers early stage virulence genes.
Collapse
Affiliation(s)
- Jule Anna Horlbog
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, 8006 Zürich, Switzerland.
| | - Marc J A Stevens
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, 8006 Zürich, Switzerland.
| | - Roger Stephan
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, 8006 Zürich, Switzerland.
| | - Claudia Guldimann
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, 8006 Zürich, Switzerland.
| |
Collapse
|
26
|
Expression, purification and biochemical characterization of Listeria monocytogenes single stranded DNA binding protein 1. Protein Expr Purif 2019; 161:63-69. [DOI: 10.1016/j.pep.2019.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/22/2019] [Accepted: 04/22/2019] [Indexed: 11/22/2022]
|
27
|
Bojer MS, Wacnik K, Kjelgaard P, Gallay C, Bottomley AL, Cohn MT, Lindahl G, Frees D, Veening JW, Foster SJ, Ingmer H. SosA inhibits cell division in Staphylococcus aureus in response to DNA damage. Mol Microbiol 2019; 112:1116-1130. [PMID: 31290194 PMCID: PMC6851548 DOI: 10.1111/mmi.14350] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2019] [Indexed: 01/10/2023]
Abstract
Inhibition of cell division is critical for viability under DNA‐damaging conditions. DNA damage induces the SOS response that in bacteria inhibits cell division while repairs are being made. In coccoids, such as the human pathogen, Staphylococcus aureus, this process remains poorly studied. Here, we identify SosA as the staphylococcal SOS‐induced cell division inhibitor. Overproduction of SosA inhibits cell division, while sosA inactivation sensitizes cells to genotoxic stress. SosA is a small, predicted membrane protein with an extracellular C‐terminal domain in which point mutation of residues that are conserved in staphylococci and major truncations abolished the inhibitory activity. In contrast, a minor truncation led to SosA accumulation and a strong cell division inhibitory activity, phenotypically similar to expression of wild‐type SosA in a CtpA membrane protease mutant. This suggests that the extracellular C‐terminus of SosA is required both for cell division inhibition and for turnover of the protein. Microscopy analysis revealed that SosA halts cell division and synchronizes the cell population at a point where division proteins such as FtsZ and EzrA are localized at midcell, and the septum formation is initiated but unable to progress to closure. Thus, our findings show that SosA is central in cell division regulation in staphylococci.
Collapse
Affiliation(s)
- Martin S Bojer
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Centre for Bacterial Stress Response and Persistence, University of Copenhagen, Copenhagen, Denmark
| | - Katarzyna Wacnik
- Department of Molecular Biology and Biotechnology, The Krebs Institute, University of Sheffield, Sheffield, UK
| | - Peter Kjelgaard
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Clement Gallay
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Amy L Bottomley
- Department of Molecular Biology and Biotechnology, The Krebs Institute, University of Sheffield, Sheffield, UK
| | - Marianne T Cohn
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gunnar Lindahl
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dorte Frees
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jan-Willem Veening
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Simon J Foster
- Department of Molecular Biology and Biotechnology, The Krebs Institute, University of Sheffield, Sheffield, UK
| | - Hanne Ingmer
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Centre for Bacterial Stress Response and Persistence, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Establishment of Listeria monocytogenes in the Gastrointestinal Tract. Microorganisms 2019; 7:microorganisms7030075. [PMID: 30857361 PMCID: PMC6463042 DOI: 10.3390/microorganisms7030075] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/05/2019] [Accepted: 03/05/2019] [Indexed: 12/15/2022] Open
Abstract
Listeria monocytogenes is a Gram positive foodborne pathogen that can colonize the gastrointestinal tract of a number of hosts, including humans. These environments contain numerous stressors such as bile, low oxygen and acidic pH, which may impact the level of colonization and persistence of this organism within the GI tract. The ability of L. monocytogenes to establish infections and colonize the gastrointestinal tract is directly related to its ability to overcome these stressors, which is mediated by the efficient expression of several stress response mechanisms during its passage. This review will focus upon how and when this occurs and how this impacts the outcome of foodborne disease.
Collapse
|
29
|
Ji X, Lu P, van der Veen S. Development of a dual-antimicrobial counterselection method for markerless genetic engineering of bacterial genomes. Appl Microbiol Biotechnol 2019; 103:1465-1474. [PMID: 30607491 DOI: 10.1007/s00253-018-9565-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 11/28/2022]
Abstract
Markerless genetic engineering of bacterial genomes is commonly performed by two-step homologous recombination methods using vectors carrying flanking regions of the target gene for site-specific vector integration and counterselection markers to provide positive selection pressure on the second recombination step resulting in vector excision. Here, we provide the proof-of-principle of a novel counterselection method that exploits antagonistic activities between bactericidal and bacteriostatic antibiotics and which can provide selection pressure on the second recombination step by selective killing of bacteria retaining the antibiotic selection marker. This method was optimized for the bacterial pathogens Listeria monocytogenes and Neisseria meningitidis by screening for antagonistic activities between the bactericidal aminoglycosides kanamycin, streptomycin, and gentamicin in combination with the bacteriostatic antibiotics chloramphenicol and erythromycin. The largest difference in selective killing of both L. monocytogenes and N. meningitidis containing an antibiotic selection marker versus wild-type bacteria was observed for the combination of erythromycin, gentamicin, and ermC as antibiotic selection marker. Therefore, this combination was used to generate two markerless deletion mutants for both L. monocytogenes and N. meningitidis. After applying the dual-antimicrobial selection pressure on cultures during the second recombination step, surviving colonies were replica plated on agar with and without erythromycin. On average, 12-13% of the randomly selected bacterial colonies had lost the selection marker due to a second recombination event and approximately half of these colonies were the desired markerless in-frame deletion mutants. Therefore, this method proved to be easy and fast and should be applicable to a wide variety of bacterial species.
Collapse
Affiliation(s)
- Xuemeng Ji
- Department of Microbiology and Parasitology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ping Lu
- Department of Microbiology and Parasitology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Stijn van der Veen
- Department of Microbiology and Parasitology, School of Medicine, Zhejiang University, Hangzhou, China. .,Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China. .,State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
30
|
Dos Santos PT, Larsen PT, Menendez-Gil P, Lillebæk EMS, Kallipolitis BH. Listeria monocytogenes Relies on the Heme-Regulated Transporter hrtAB to Resist Heme Toxicity and Uses Heme as a Signal to Induce Transcription of lmo1634, Encoding Listeria Adhesion Protein. Front Microbiol 2018; 9:3090. [PMID: 30619169 PMCID: PMC6305404 DOI: 10.3389/fmicb.2018.03090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/29/2018] [Indexed: 01/29/2023] Open
Abstract
For pathogenic bacteria, host-derived heme represents an important metabolic cofactor and a source for iron. However, high levels of heme are toxic to bacteria. We have previously shown that excess heme has a growth-inhibitory effect on the Gram-positive foodborne pathogen Listeria monocytogenes, and we have learned that the LhrC1-5 family of small RNAs, together with the two-component system (TCS) LisRK, play a role in the adaptation of L. monocytogenes to heme stress conditions. However, a broader knowledge on how this pathogen responds to heme toxicity is still lacking. Here, we analyzed the global transcriptomic response of L. monocytogenes to heme stress. We found that the response of L. monocytogenes to excess heme is multifaceted, involving various strategies acting to minimize the toxic effects of heme. For example, heme exposure triggers the SOS response that deals with DNA damage. In parallel, L. monocytogenes shuts down the transcription of genes involved in heme/iron uptake and utilization. Furthermore, heme stress resulted in a massive increase in the transcription of a putative heme detoxification system, hrtAB, which is highly conserved in Gram-positive bacteria. As expected, we found that the TCS HssRS is required for heme-mediated induction of hrtAB and that a functional heme efflux system is essential for L. monocytogenes to resist heme toxicity. Curiously, the most highly up-regulated gene upon heme stress was lmo1634, encoding the Listeria adhesion protein, LAP, which acts to promote the translocation of L. monocytogenes across the intestinal barrier. Additionally, LAP is predicted to act as a bifunctional acetaldehyde-CoA/alcohol dehydrogenase. Surprisingly, a mutant lacking lmo1634 grows well under heme stress conditions, showing that LAP is not required for L. monocytogenes to resist heme toxicity. Likewise, a functional ResDE TCS, which contributes to heme-mediated expression of lmo1634, is not required for the adaptation of L. monocytogenes to heme stress conditions. Collectively, this study provides novel insights into the strategies employed by L. monocytogenes to resist heme toxicity. Our findings indicate that L. monocytogenes is using heme as a host-derived signaling molecule to control the expression of its virulence genes, as exemplified by lmo1634.
Collapse
Affiliation(s)
| | - Pernille Tholund Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Pilar Menendez-Gil
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | | | |
Collapse
|
31
|
Comparison of oxidative stress response and biofilm formation of Listeria monocytogenes serotypes 4b and 1/2a. Food Control 2018. [DOI: 10.1016/j.foodcont.2017.10.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
32
|
Du W, Zhou M, Liu Z, Chen Y, Li R. Inhibition effects of low concentrations of epigallocatechin gallate on the biofilm formation and hemolytic activity of Listeria monocytogenes. Food Control 2018. [DOI: 10.1016/j.foodcont.2017.09.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
33
|
Enhancing the Adaptability of the Deep-Sea Bacterium Shewanella piezotolerans WP3 to High Pressure and Low Temperature by Experimental Evolution under H 2O 2 Stress. Appl Environ Microbiol 2018; 84:AEM.02342-17. [PMID: 29269502 DOI: 10.1128/aem.02342-17] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/10/2017] [Indexed: 11/20/2022] Open
Abstract
Oxidative stresses commonly exist in natural environments, and microbes have developed a variety of defensive systems to counteract such events. Although increasing evidence has shown that high hydrostatic pressure (HHP) and low temperature (LT) induce antioxidant defense responses in cells, there is no direct evidence to prove the connection between antioxidant defense mechanisms and the adaptation of bacteria to HHP and LT. In this study, using the wild-type (WT) strain of a deep-sea bacterium, Shewanella piezotolerans WP3, as an ancestor, we obtained a mutant, OE100, with an enhanced antioxidant defense capacity by experimental evolution under H2O2 stress. Notably, OE100 exhibited better tolerance not only to H2O2 stress but also to HHP and LT (20 MPa and 4°C, respectively). Whole-genome sequencing identified a deletion mutation in the oxyR gene, which encodes the transcription factor that controls the oxidative stress response. Comparative transcriptome analysis showed that the genes associated with oxidative stress defense, anaerobic respiration, DNA repair, and the synthesis of flagella and bacteriophage were differentially expressed in OE100 compared with the WT at 20 MPa and 4°C. Genetic analysis of oxyR and ccpA2 indicated that the OxyR-regulated cytochrome c peroxidase CcpA2 significantly contributed to the adaptation of WP3 to HHP and LT. Taken together, these results confirmed the inherent relationship between antioxidant defense mechanisms and the adaptation of a benthic microorganism to HHP and LT.IMPORTANCE Oxidative stress exists in various niches, including the deep-sea ecosystem, which is an extreme environment with conditions of HHP and predominantly LT. Although previous studies have shown that HHP and LT induce antioxidant defense responses in cells, direct evidence to prove the connection between antioxidant defense mechanisms and the adaptation of bacteria to HHP and LT is lacking. In this work, using the deep-sea bacterium Shewanella piezotolerans WP3 as a model, we proved that enhancement of the adaptability of WP3 to HHP and LT can benefit from its antioxidant defense mechanism, which provided useful insight into the ecological roles of antioxidant genes in a benthic microorganism and contributed to an improved understanding of microbial adaptation strategies in deep-sea environments.
Collapse
|
34
|
Dellus-Gur E, Ram Y, Hadany L. Errors in mutagenesis and the benefit of cell-to-cell signalling in the evolution of stress-induced mutagenesis. ROYAL SOCIETY OPEN SCIENCE 2017; 4:170529. [PMID: 29291054 PMCID: PMC5717628 DOI: 10.1098/rsos.170529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 10/02/2017] [Indexed: 06/07/2023]
Abstract
Stress-induced mutagenesis is a widely observed phenomenon. Theoretical models have shown that stress-induced mutagenesis can be favoured by natural selection due to the beneficial mutations it generates. These models, however, assumed an error-free regulation of mutation rate in response to stress. Here, we explore the effects of errors in the regulation of mutagenesis on the evolution of stress-induced mutagenesis, and consider the role of cell-to-cell signalling. Using theoretical models, we show (i) that stress-induced mutagenesis can be disadvantageous if errors are common; and (ii) that cell-to-cell signalling can allow stress-induced mutagenesis to be favoured by selection even when error rates are high. We conclude that cell-to-cell signalling can facilitate the evolution of stress-induced mutagenesis in microbes through second-order selection.
Collapse
|
35
|
Krajewski SS, Isoz I, Johansson J. Antibacterial and antivirulence effect of 6-N-hydroxylaminopurine in Listeria monocytogenes. Nucleic Acids Res 2017; 45:1914-1924. [PMID: 28062853 PMCID: PMC5389569 DOI: 10.1093/nar/gkw1308] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 12/20/2016] [Indexed: 01/07/2023] Open
Abstract
The emerging development of antibiotic resistant bacteria calls for novel types of antibacterial agents. In this work we examined the putative antibacterial effect of purine analogs in Listeria monocytogenes. We show that, among several tested purine analogs, only 6-N-hydroxylaminopurine (6-N-HAP) reduces the viability of the Gram-positive pathogen Listeria monocytogenes. As in Bacillus subtilis, 6-N-HAP terminates expression at guanine riboswitches in L. monocytogenes hence preventing expression of their downstream genes. However, we show that the bacteriocidal effect of the compound was unlinked to the terminated expression at the guanine riboswitches. When further examining the antimicrobial effect, we observed that 6-N-HAP acts as a potent mutagen in L. monocytogenes, by increasing the mutation rate and inducing the SOS-response. Also, addition of 6-N-HAP decreased virulence gene expression by reducing both the levels and activity of the virulence regulator PrfA.
Collapse
Affiliation(s)
- Stefanie Sandra Krajewski
- Department of Molecular Biology, Umeå University, 90187 Umeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187 Umeå, Sweden.,Umeå Centre for Microbial Research (UCMR), Umeå University, 90187 Umeå, Sweden
| | - Isabelle Isoz
- Department of Molecular Biology, Umeå University, 90187 Umeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187 Umeå, Sweden.,Umeå Centre for Microbial Research (UCMR), Umeå University, 90187 Umeå, Sweden
| | - Jörgen Johansson
- Department of Molecular Biology, Umeå University, 90187 Umeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187 Umeå, Sweden.,Umeå Centre for Microbial Research (UCMR), Umeå University, 90187 Umeå, Sweden
| |
Collapse
|
36
|
Ochiai Y, Yamada F, Yoshikawa Y, Mochizuki M, Takano T, Hondo R, Ueda F. Sequential transition of the injury phenotype, temperature-dependent survival and transcriptional response in Listeria monocytogenes following lethal H 2 O 2 exposure. Int J Food Microbiol 2017; 259:52-58. [DOI: 10.1016/j.ijfoodmicro.2017.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 07/29/2017] [Accepted: 08/01/2017] [Indexed: 11/29/2022]
|
37
|
Omori Y, Miake K, Nakamura H, Kage-Nakadai E, Nishikawa Y. Influence of lactic acid and post-treatment recovery time on the heat resistance of Listeria monocytogenes. Int J Food Microbiol 2017. [DOI: 10.1016/j.ijfoodmicro.2017.06.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
38
|
Akashi M, Harada S, Moki S, Okouji Y, Takahashi K, Kada S, Yamagami K, Sekine Y, Watanabe S, Chibazakura T, Yoshikawa H. Transposition of insertion sequence IS256Bsu1 in Bacillus subtilis 168 is strictly dependent on recA. Genes Genet Syst 2017; 92:59-71. [PMID: 28344191 DOI: 10.1266/ggs.16-00071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
We developed an insertion sequence transposition detection system called the "jumping cat assay" and applied it to the Bacillus subtilis chromosome using IS256Bsu1 derived from B. subtilis natto. The high frequency of transposition enabled us to explore host factors; combining the assay and genetic analyses revealed that recA is essential for the transposition of IS256Bsu1. Detailed analyses using various domain mutants of recA demonstrated that this essentiality is not related to the function of recA in homologous recombination. Instead, the ATP binding and hydrolysis function seemed to be crucial for IS transposition. To elucidate the role of recA, we focused on the muB gene of the enterobacteriophage Mu. Based on information from the NCBI Conserved Domain Database, both MuB and RecA belong to the P-loop dNTPase superfamily. Further experiments revealed that muB complements the transposition-defective phenotype of a recA deletant, although it could not rescue UV sensitivity. These results suggest that recA shares a common function with muB that helps the transposition of IS256Bsu1 in B. subtilis.
Collapse
Affiliation(s)
| | - Shota Harada
- Department of Bioscience, Tokyo University of Agriculture
| | - Syunsuke Moki
- Department of Bioscience, Tokyo University of Agriculture
| | - Yuki Okouji
- Department of Bioscience, Tokyo University of Agriculture
| | | | - Shigeki Kada
- Central Research Institute, Mitsukan Group Co., Ltd
| | | | - Yasuhiko Sekine
- Department of Life Science, College of Science, Rikkyo (St Paul's) University
| | | | | | | |
Collapse
|
39
|
Nale JY, Chutia M, Carr P, Hickenbotham PT, Clokie MRJ. 'Get in Early'; Biofilm and Wax Moth (Galleria mellonella) Models Reveal New Insights into the Therapeutic Potential of Clostridium difficile Bacteriophages. Front Microbiol 2016; 7:1383. [PMID: 27630633 PMCID: PMC5005339 DOI: 10.3389/fmicb.2016.01383] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/22/2016] [Indexed: 12/19/2022] Open
Abstract
Clostridium difficile infection (CDI) is a global health threat associated with high rates of morbidity and mortality. Conventional antibiotic CDI therapy can result in treatment failure and recurrent infection. C. difficile produces biofilms which contribute to its virulence and impair antimicrobial activity. Some bacteriophages (phages) can penetrate biofilms and thus could be developed to either replace or supplement antibiotics. Here, we determined the impact of a previously optimized 4-phage cocktail on C. difficile ribotype 014/020 biofilms, and additionally as adjunct to vancomycin treatment in Galleria mellonella larva CDI model. The phages were applied before or after biofilm establishment in vitro, and the impact was analyzed according to turbidity, viability counts and topography as observed using scanning electron and confocal microscopy. The infectivity profiles and efficacies of orally administered phages and/or vancomycin were ascertained by monitoring colonization levels and larval survival rates. Phages prevented biofilm formation, and penetrated established biofilms. A single phage application reduced colonization causing extended longevity in the remedial treatment and prevented disease in the prophylaxis group. Multiple phage doses significantly improved the larval remedial regimen, and this treatment is comparable to vancomycin and the combined treatments. Taken together, our data suggest that the phages significantly reduce C. difficile biofilms, and prevent colonization in the G. mellonella model when used alone or in combination with vancomycin. The phages appear to be highly promising therapeutics in the targeted eradication of CDI and the use of these models has revealed that prophylactic use could be a propitious therapeutic option.
Collapse
Affiliation(s)
- Janet Y Nale
- Department of Infection, Immunity and Inflammation, University of Leicester Leicester, UK
| | - Mahananda Chutia
- Pathology and Microbiology Division, Central Muga Eri Research and Training Institute Assam, India
| | - Philippa Carr
- Department of Infection, Immunity and Inflammation, University of Leicester Leicester, UK
| | - Peter T Hickenbotham
- Department of Infection, Immunity and Inflammation, University of Leicester Leicester, UK
| | - Martha R J Clokie
- Department of Infection, Immunity and Inflammation, University of Leicester Leicester, UK
| |
Collapse
|
40
|
Kothary MH, Franco AA, Tall BD, Gopinath GR, Datta AR. Purification and Characterization of a Rabbit Serum Factor That Kills Listeria Species and Other Foodborne Bacterial Pathogens. Foodborne Pathog Dis 2016; 13:441-7. [PMID: 27455064 DOI: 10.1089/fpd.2015.2109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In an in-vitro assay, rabbit serum, but not human serum, killed Listeria monocytogenes, a foodborne pathogen. The aim of our study was to purify and partially characterize this killing factor. Listericidin was purified from rabbit serum by a single-step ion-exchange chromatography with DEAE-Sephadex A-50 and its antimicrobial activity was assessed by a microdilution method. Listericidin is a protein with a molecular weight of 9 kDa and an isoelectric point of 8.1. It kills L. monocytogenes at 4°C, 25°C, and 37°C, and its activity is resistant to heat (boiling) and acidic conditions (pH <2). Listericidin's activity is inhibited by sodium chloride and various growth media, is sensitive to proteolytic enzymes and is enhanced by calcium chloride, and is neutralized by monoclonal antibodies to human complement C3a. However, the listericidin reacts weakly with these antibodies in an ELISA. The first 33 N-terminal residues of listericidin (SVQLTEKRMDKVGQYTNKELRKXXEDGMRDNPM) have homology to various complement C3a components. Listericidin also kills other Listeria spp., Vibrio spp., Salmonella spp., Escherichia spp., Cronobacter spp., and Bacillus spp. The listericidin peptide purified in a single-step chromatography is pH and heat stable, and has a broad antimicrobial spectrum against major foodborne pathogens in addition to L. monocytogenes.
Collapse
Affiliation(s)
- Mahendra H Kothary
- Division of Virulence Assessment, Food and Drug Administration , Laurel, Maryland
| | - Augusto A Franco
- Division of Virulence Assessment, Food and Drug Administration , Laurel, Maryland
| | - Ben D Tall
- Division of Virulence Assessment, Food and Drug Administration , Laurel, Maryland
| | - Gopal R Gopinath
- Division of Virulence Assessment, Food and Drug Administration , Laurel, Maryland
| | - Atin R Datta
- Division of Virulence Assessment, Food and Drug Administration , Laurel, Maryland
| |
Collapse
|
41
|
Fornelos N, Browning DF, Butala M. The Use and Abuse of LexA by Mobile Genetic Elements. Trends Microbiol 2016; 24:391-401. [PMID: 26970840 DOI: 10.1016/j.tim.2016.02.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 02/12/2016] [Accepted: 02/18/2016] [Indexed: 11/15/2022]
Abstract
The SOS response is an essential process for responding to DNA damage in bacteria. The expression of SOS genes is under the control of LexA, a global transcription factor that undergoes self-cleavage during stress to allow the expression of DNA repair functions and delay cell division until the damage is rectified. LexA also regulates genes that are not part of this cell rescue program, and the induction of bacteriophages, the movement of pathogenicity islands, and the expression of virulence factors and bacteriocins are all controlled by this important transcription factor. Recently it has emerged that when regulating the expression of genes from mobile genetic elements (MGEs), LexA often does so in concert with a corepressor. This accessory regulator can either be a host-encoded global transcription factor, which responds to various metabolic changes, or a factor that is encoded for by the MGE itself. Thus, the coupling of LexA-mediated regulation to a secondary transcription factor not only detaches LexA from its primary SOS role, but also fine-tunes gene expression from the MGE, enabling it to respond to multiple stresses. Here we discuss the mechanisms of such coordinated regulation and its implications for cells carrying such MGEs.
Collapse
Affiliation(s)
- Nadine Fornelos
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyvaskyla, PO Box 35, F-40014 Jyvaskyla, Finland.
| | - Douglas F Browning
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Matej Butala
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, 1000 Ljubljana, Slovenia.
| |
Collapse
|
42
|
Abee T, Koomen J, Metselaar K, Zwietering M, den Besten H. Impact of Pathogen Population Heterogeneity and Stress-Resistant Variants on Food Safety. Annu Rev Food Sci Technol 2016; 7:439-56. [DOI: 10.1146/annurev-food-041715-033128] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- T. Abee
- Laboratory of Food Microbiology, Wageningen University, 6700 AA Wageningen, The Netherlands;
| | - J. Koomen
- Laboratory of Food Microbiology, Wageningen University, 6700 AA Wageningen, The Netherlands;
| | - K.I. Metselaar
- Laboratory of Food Microbiology, Wageningen University, 6700 AA Wageningen, The Netherlands;
| | - M.H. Zwietering
- Laboratory of Food Microbiology, Wageningen University, 6700 AA Wageningen, The Netherlands;
| | - H.M.W. den Besten
- Laboratory of Food Microbiology, Wageningen University, 6700 AA Wageningen, The Netherlands;
| |
Collapse
|
43
|
Schons-Fonseca L, da Silva JB, Milanez JS, Domingos RH, Smith JL, Nakaya HI, Grossman AD, Ho PL, da Costa RMA. Analysis of LexA binding sites and transcriptomics in response to genotoxic stress in Leptospira interrogans. Nucleic Acids Res 2016; 44:1179-91. [PMID: 26762976 PMCID: PMC4756842 DOI: 10.1093/nar/gkv1536] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 12/15/2015] [Accepted: 12/25/2015] [Indexed: 11/13/2022] Open
Abstract
We determined the effects of DNA damage caused by ultraviolet radiation on gene expression in Leptospira interrogans using DNA microarrays. These data were integrated with DNA binding in vivo of LexA1, a regulator of the DNA damage response, assessed by chromatin immunoprecipitation and massively parallel DNA sequencing (ChIP-seq). In response to DNA damage, Leptospira induced expression of genes involved in DNA metabolism, in mobile genetic elements and defective prophages. The DNA repair genes involved in removal of photo-damage (e.g. nucleotide excision repair uvrABC, recombinases recBCD and resolvases ruvABC) were not induced. Genes involved in various metabolic pathways were down regulated, including genes involved in cell growth, RNA metabolism and the tricarboxylic acid cycle. From ChIP-seq data, we observed 24 LexA1 binding sites located throughout chromosome 1 and one binding site in chromosome 2. Expression of many, but not all, genes near those sites was increased following DNA damage. Binding sites were found as far as 550 bp upstream from the start codon, or 1 kb into the coding sequence. Our findings indicate that there is a shift in gene expression following DNA damage that represses genes involved in cell growth and virulence, and induces genes involved in mutagenesis and recombination.
Collapse
Affiliation(s)
- Luciane Schons-Fonseca
- Centro de Biotecnologia, Instituto Butantan, São Paulo, São Paulo 05503-900, Brazil Instituto de Química, Universidade de São Paulo, São Paulo, São Paulo 05508-000, Brazil Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, MA 02139, USA
| | - Josefa B da Silva
- Centro de Biotecnologia, Instituto Butantan, São Paulo, São Paulo 05503-900, Brazil
| | - Juliana S Milanez
- Centro de Biotecnologia, Instituto Butantan, São Paulo, São Paulo 05503-900, Brazil Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, São Paulo 05508-000, Brazil
| | - Renan H Domingos
- Centro de Biotecnologia, Instituto Butantan, São Paulo, São Paulo 05503-900, Brazil
| | - Janet L Smith
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, MA 02139, USA
| | - Helder I Nakaya
- Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, São Paulo 05508-000, Brazil
| | - Alan D Grossman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, MA 02139, USA
| | - Paulo L Ho
- Centro de Biotecnologia, Instituto Butantan, São Paulo, São Paulo 05503-900, Brazil
| | - Renata M A da Costa
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, São Paulo 09210580, Brazil
| |
Collapse
|
44
|
Jian H, Xiong L, He Y, Xiao X. The regulatory function of LexA is temperature-dependent in the deep-sea bacterium Shewanella piezotolerans WP3. Front Microbiol 2015; 6:627. [PMID: 26150814 PMCID: PMC4471891 DOI: 10.3389/fmicb.2015.00627] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/08/2015] [Indexed: 01/01/2023] Open
Abstract
The SOS response addresses DNA lesions and is conserved in the bacterial domain. The response is governed by the DNA binding protein LexA, which has been characterized in model microorganisms such as Escherichia coli. However, our understanding of its roles in deep-sea bacteria is limited. Here, the influence of LexA on the phenotype and gene transcription of Shewanella piezotolerans WP3 (WP3) was investigated by constructing a lexA deletion strain (WP3ΔlexA), which was compared with the wild-type strain. No growth defect was observed for WP3ΔlexA. A total of 481 and 108 genes were differentially expressed at 20 and 4°C, respectively, as demonstrated by comparative whole genome microarray analysis. Furthermore, the swarming motility and dimethylsulfoxide reduction assay demonstrated that the function of LexA was related to temperature. The transcription of the lexA gene was up-regulated during cold acclimatization and after cold shock, indicating that the higher expression level of LexA at low temperatures may be responsible for its temperature-dependent functions. The deep-sea microorganism S. piezotolerans WP3 is the only bacterial species whose SOS regulator has been demonstrated to be significantly influenced by environmental temperatures to date. Our data support the hypothesis that SOS is a formidable strategy used by bacteria against various environmental stresses.
Collapse
Affiliation(s)
- Huahua Jian
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University Shanghai, China
| | - Lei Xiong
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University Shanghai, China
| | - Ying He
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University Shanghai, China
| | - Xiang Xiao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University Shanghai, China ; State Key Laboratory of Ocean Engineering, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University Shanghai, China
| |
Collapse
|
45
|
Genes Associated with Desiccation and Osmotic Stress in Listeria monocytogenes as Revealed by Insertional Mutagenesis. Appl Environ Microbiol 2015; 81:5350-62. [PMID: 26025900 DOI: 10.1128/aem.01134-15] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 05/22/2015] [Indexed: 12/29/2022] Open
Abstract
Listeria monocytogenes is a foodborne pathogen whose survival in food processing environments may be associated with its tolerance to desiccation. To probe the molecular mechanisms used by this bacterium to adapt to desiccation stress, a transposon library of 11,700 L. monocytogenes mutants was screened, using a microplate assay, for strains displaying increased or decreased desiccation survival (43% relative humidity, 15°C) in tryptic soy broth (TSB). The desiccation phenotypes of selected mutants were subsequently assessed on food-grade stainless steel (SS) coupons in TSB plus 1% glucose (TSB-glu). Single transposon insertions in mutants exhibiting a change in desiccation survival of >0.5 log CFU/cm(2) relative to that of the wild type were determined by sequencing arbitrary PCR products. Strain morphology, motility, and osmotic stress survival (in TSB-glu plus 20% NaCl) were also analyzed. The initial screen selected 129 desiccation-sensitive (DS) and 61 desiccation-tolerant (DT) mutants, out of which secondary screening on SS confirmed 15 DT and 15 DS mutants. Among the DT mutants, seven immotile and flagellum-less strains contained transposons in genes involved in flagellum biosynthesis (fliP, flhB, flgD, flgL) and motor control (motB, fliM, fliY), while others harbored transposons in genes involved in membrane lipid biosynthesis, energy production, potassium uptake, and virulence. The genes that were interrupted in the 15 DS mutants included those involved in energy production, membrane transport, protein metabolism, lipid biosynthesis, oxidative damage control, and putative virulence. Five DT and 14 DS mutants also demonstrated similar significantly (P < 0.05) different survival relative to that of the wild type when exposed to osmotic stress, demonstrating that some genes likely have similar roles in allowing the organism to survive the two water stresses.
Collapse
|
46
|
An SOS Regulon under Control of a Noncanonical LexA-Binding Motif in the Betaproteobacteria. J Bacteriol 2015; 197:2622-30. [PMID: 25986903 DOI: 10.1128/jb.00035-15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 05/09/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The SOS response is a transcriptional regulatory network governed by the LexA repressor that activates in response to DNA damage. In the Betaproteobacteria, LexA is known to target a palindromic sequence with the consensus sequence CTGT-N8-ACAG. We report the characterization of a LexA regulon in the iron-oxidizing betaproteobacterium Sideroxydans lithotrophicus. In silico and in vitro analyses show that LexA targets six genes by recognizing a binding motif with the consensus sequence GAACGaaCGTTC, which is strongly reminiscent of the Bacillus subtilis LexA-binding motif. We confirm that the closely related Gallionella capsiferriformans shares the same LexA-binding motif, and in silico analyses indicate that this motif is also conserved in the Nitrosomonadales and the Methylophilales. Phylogenetic analysis of LexA and the alpha subunit of DNA polymerase III (DnaE) reveal that the organisms harboring this noncanonical LexA form a compact taxonomic cluster within the Betaproteobacteria. However, their lexA gene is unrelated to the standard Betaproteobacteria lexA, and there is evidence of its spread through lateral gene transfer. In contrast to other reported cases of noncanonical LexA-binding motifs, the regulon of S. lithotrophicus is comparable in size and function to that of many other Betaproteobacteria, suggesting that a convergent SOS regulon has reevolved under the control of a new LexA protein. Analysis of the DNA-binding domain of S. lithotrophicus LexA reveals little sequence similarity with that of other LexA proteins targeting similar binding motifs, suggesting that network structure may limit site evolution or that structural constrains make the B. subtilis-type motif an optimal interface for multiple LexA sequences. IMPORTANCE Understanding the evolution of transcriptional systems enables us to address important questions in microbiology, such as the emergence and transfer potential of different regulatory systems to regulate virulence or mediate responses to stress. The results reported here constitute the first characterization of a noncanonical LexA protein regulating a standard SOS regulon. This is significant because it illustrates how a complex transcriptional program can be put under the control of a novel transcriptional regulator. Our results also reveal a substantial degree of plasticity in the LexA recognition domain, raising intriguing questions about the space of protein-DNA interfaces and the specific evolutionary constrains faced by these elements.
Collapse
|
47
|
Metselaar KI, den Besten HMW, Boekhorst J, van Hijum SAFT, Zwietering MH, Abee T. Diversity of acid stress resistant variants of Listeria monocytogenes and the potential role of ribosomal protein S21 encoded by rpsU. Front Microbiol 2015; 6:422. [PMID: 26005439 PMCID: PMC4424878 DOI: 10.3389/fmicb.2015.00422] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 04/21/2015] [Indexed: 12/02/2022] Open
Abstract
The dynamic response of microorganisms to environmental conditions depends on the behavior of individual cells within the population. Adverse environments can select for stable stress resistant subpopulations. In this study, we aimed to get more insight in the diversity within Listeria monocytogenes LO28 populations, and the genetic basis for the increased resistance of stable resistant fractions isolated after acid exposure. Phenotypic cluster analysis of 23 variants resulted in three clusters and four individual variants and revealed multiple-stress resistance, with both unique and overlapping features related to stress resistance, growth, motility, biofilm formation, and virulence indicators. A higher glutamate decarboxylase activity correlated with increased acid resistance. Whole genome sequencing revealed mutations in rpsU, encoding ribosomal protein S21 in the largest phenotypic cluster, while mutations in ctsR, which were previously shown to be responsible for increased resistance of heat and high hydrostatic pressure resistant variants, were not found in the acid resistant variants. This underlined that large population diversity exists within one L. monocytogenes strain and that different adverse conditions drive selection for different variants. The finding that acid stress selects for rpsU variants provides potential insights in the mechanisms underlying population diversity of L. monocytogenes.
Collapse
Affiliation(s)
- Karin I Metselaar
- Top Institute Food and Nutrition Wageningen, Netherlands ; Laboratory of Food Microbiology, Wageningen University Wageningen, Netherlands
| | | | - Jos Boekhorst
- Top Institute Food and Nutrition Wageningen, Netherlands ; NIZO Food Research Ede, Netherlands
| | - Sacha A F T van Hijum
- Top Institute Food and Nutrition Wageningen, Netherlands ; NIZO Food Research Ede, Netherlands ; Bacterial Genomics Group, CMBI Centre for Molecular and Biomolecular Informatics, Radboud University Medical Center Nijmegen, Netherlands
| | - Marcel H Zwietering
- Top Institute Food and Nutrition Wageningen, Netherlands ; Laboratory of Food Microbiology, Wageningen University Wageningen, Netherlands
| | - Tjakko Abee
- Top Institute Food and Nutrition Wageningen, Netherlands ; Laboratory of Food Microbiology, Wageningen University Wageningen, Netherlands
| |
Collapse
|
48
|
Phosphate Limitation Induces Drastic Physiological Changes, Virulence-Related Gene Expression, and Secondary Metabolite Production in Pseudovibrio sp. Strain FO-BEG1. Appl Environ Microbiol 2015; 81:3518-28. [PMID: 25769826 DOI: 10.1128/aem.04167-14] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/09/2015] [Indexed: 12/11/2022] Open
Abstract
Phosphorus is a vital nutrient for living organisms and is obtained by bacteria primarily via phosphate uptake. However, phosphate is often scarcely accessible in nature, and there is evidence that in many areas of the ocean, its concentration limits bacterial growth. Surprisingly, the phosphate starvation response has been extensively investigated in different model organisms (e.g., Escherichia coli), but there is a dearth of studies on heterotrophic marine bacteria. In this work, we describe the response of Pseudovibrio sp. strain FO-BEG1, a metabolically versatile alphaproteobacterium and potential symbiont of marine sponges, to phosphate limitation. We compared the physiology, protein expression, and secondary metabolite production under phosphate-limited conditions to those under phosphate surplus conditions. We observed that phosphate limitation had a pleiotropic effect on the physiology of the strain, triggering cell elongation, the accumulation of polyhydroxyalkanoate, the degradation of polyphosphate, and the exchange of membrane lipids in favor of phosphorus-free lipids such as sulfoquinovosyl diacylglycerols. Many proteins involved in the uptake and degradation of phospho-organic compounds were upregulated, together with subunits of the ABC transport system for phosphate. Under conditions of phosphate limitation, FO-BEG1 secreted compounds into the medium that conferred an intense yellow coloration to the cultures. Among these compounds, we identified the potent antibiotic tropodithietic acid. Finally, toxin-like proteins and other proteins likely involved in the interaction with the eukaryotic host were also upregulated. Altogether, our data suggest that phosphate limitation leads to a pronounced reorganization of FO-BEG1 physiology, involving phosphorus, carbon, and sulfur metabolism; cell morphology; secondary metabolite production; and the expression of virulence-related genes.
Collapse
|
49
|
Suo Y, Liu Y, Zhou X, Huang Y, Shi C, Matthews K, Shi X. Impact of Sod on the expression of stress-related genes in Listeria monocytogenes 4b G with/without paraquat treatment. J Food Sci 2014; 79:M1745-9. [PMID: 25146690 DOI: 10.1111/1750-3841.12545] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 05/30/2014] [Indexed: 11/28/2022]
Abstract
Listeria monocytogenes is a foodborne pathogen that causes listeriosis. Paraquat can generate reactive oxygen species (ROS) in cells, which results in oxidative stress. It was first shown that 1 mM of paraquat inhibited the growth rate of a superoxide dismutase (sod)-deletion mutant (∆sod) generated from L. monocytogenes 4b G but not in the wild-type, and induced the expression of other resistance genes (kat, fri, perR, sigB, and recA) as well as sod in the wild type. Interestingly, without paraquat treatment the expression of all the 5 genes were repressed in ∆sod compared to the wild type, while the expression of recA triggering SOS response, a global response to DNA damage, was increased in ∆sod in the presence of 1 mM paraquat. Taken together, these results suggest that SOD plays a central role in oxidant defense of L. monocytogenes 4b G, and SOS probably significantly impacts ∆sod survival under oxidative stress.
Collapse
Affiliation(s)
- Yujuan Suo
- MOST-USDA Joint Research Center for Food Safety, School of Agriculture and Biology, and State Key Lab of Microbial Metabolism, Shanghai Jiao Tong Univ, Shanghai, 200240, China
| | | | | | | | | | | | | |
Collapse
|
50
|
Antibiotic-induced replication stress triggers bacterial competence by increasing gene dosage near the origin. Cell 2014; 157:395-406. [PMID: 24725406 DOI: 10.1016/j.cell.2014.01.068] [Citation(s) in RCA: 172] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 12/01/2013] [Accepted: 01/29/2014] [Indexed: 12/12/2022]
Abstract
Streptococcus pneumoniae (pneumococcus) kills nearly 1 million children annually, and the emergence of antibiotic-resistant strains poses a serious threat to human health. Because pneumococci can take up DNA from their environment by a process called competence, genes associated with antibiotic resistance can rapidly spread. Remarkably, competence is activated in response to several antibiotics. Here, we demonstrate that antibiotics targeting DNA replication cause an increase in the copy number of genes proximal to the origin of replication (oriC). As the genes required for competence initiation are located near oriC, competence is thereby activated. Transcriptome analyses show that antibiotics targeting DNA replication also upregulate origin-proximal gene expression in other bacteria. This mechanism is a direct, intrinsic consequence of replication fork stalling. Our data suggest that evolution has conserved the oriC-proximal location of important genes in bacteria to allow for a robust response to replication stress without the need for complex gene-regulatory pathways. PAPERCLIP:
Collapse
|