1
|
Karthikeyan A, Tabassum N, Mani AK, Javaid A, Kim TH, Oh DK, Kim YM, Jung WK, Khan F. Targeting motility of Listeria monocytogenes: Alternative strategies to control foodborne illness. Microb Pathog 2025; 202:107427. [PMID: 40021026 DOI: 10.1016/j.micpath.2025.107427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Listeria monocytogenes, a gram-positive facultative anaerobic bacterium, demonstrates remarkable adaptability to various environmental stressors in food processing environments. It can survive and grow under extremely challenging environmental conditions such as low pH and temperatures, high salinity, and UV radiation. Its ability to generate biofilms at multiple stages of the food processing chain poses significant food safety issues. This bacterium is known for causing severe listeriosis, making it a major problem in microbiology and food safety. L. monocytogenes relies on motility to explore surfaces, attach, and build biofilms. It comprises actin-based motility, which is used for cell-to-cell propagation inside host tissues, and flagellar-driven motility, which assists in surface colonization and infection spread. Flagellar motility also plays an important function in increasing virulence throughout infection cycles. L. monocytogenes motility is regulated by a complex network of regulatory proteins that govern the expression of motility-associated genes. These proteins directly impact pathogenicity by influencing motility and biofilm formation, as well as an indirect impact via regulatory pathways. Efforts to control L. monocytogenes infections and decrease food safety impact include a variety of procedures. Natural compounds, synthetic agents, nanomaterials, and conjugates have emerged as intriguing options for inhibiting motility, disrupting biofilm formation, and reducing virulence. These strategies focus on vital elements of the L. monocytogenes life cycle and pathophysiology to improve food safety and public health. This review provides a comprehensive discussion of the regulatory mechanisms governing L. monocytogenes motility, emphasizing their role in pathogenicity, and explores potential strategies for attenuating the motility and virulence properties. Understanding these mechanisms is essential for developing targeted therapeutic approaches against L. monocytogenes infections and improving food safety practices.
Collapse
Affiliation(s)
- Abirami Karthikeyan
- Department of Chemistry and Biosciences, Srinivasa Ramanujan Centre, SASTRA Deemed University, Kumbakonam, 612001, Tamil Nadu, India; Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, 48513, Republic of Korea
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Arun Kumar Mani
- Department of Chemistry and Biosciences, Srinivasa Ramanujan Centre, SASTRA Deemed University, Kumbakonam, 612001, Tamil Nadu, India
| | - Aqib Javaid
- Interdisciplinary Program of Marine and Fisheries Sciences and Convergent Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Tae-Hee Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Do Kyung Oh
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Won-Kyo Jung
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Ocean and Fisheries Development International Cooperation Institute, Pukyong National University, Busan, 48513, Republic of Korea; International Graduate Program of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
2
|
Luciani M, Krasteva I, Schirone M, D'Onofrio F, Iannetti L, Torresi M, Di Pancrazio C, Perletta F, Valentinuzzi S, Tittarelli M, Pomilio F, D'Alterio N, Paparella A, Del Boccio P. Adaptive strategies of Listeria monocytogenes: An in-depth analysis of the virulent strain involved in an outbreak in Italy through quantitative proteomics. Int J Food Microbiol 2025; 427:110951. [PMID: 39486093 DOI: 10.1016/j.ijfoodmicro.2024.110951] [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: 08/10/2024] [Revised: 09/28/2024] [Accepted: 10/18/2024] [Indexed: 11/04/2024]
Abstract
Despite the general classification of L. monocytogenes strains as equally virulent by global safety authorities, molecular epidemiology reveals diverse subtypes in food, processing environments, and clinical cases. This study focuses on a highly virulent strain associated with a listeriosis outbreak in Italy in 2022, providing insights through comprehensive foodomics approaches, with a specific emphasis on quantitative proteomics. In particular, the ST155 strain of L. monocytogenes strain was subjected in vitro to growth stress conditions (NaCl 2.4 %, pH 6.2, T 12 °C), mimicking the conditions present in the frankfurter, its original source. Then, the protein expression patterns were compared with those obtained in optimal growth conditions. Through quantitative proteomic analysis and bioinformatic assessment, different proteins associated with virulence during the exponential growth phase were identified. This study unveils unique proteins specific to each environment, providing insights into how L. monocytogenes adapts to conditions that are similar to those encountered in frankfurters. This investigation contributes valuable insights into the adaptive strategies of L. monocytogenes under stressful conditions, with implications for enhancing food safety practices.
Collapse
Affiliation(s)
- Mirella Luciani
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", 64100 Teramo, Italy; Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Ivanka Krasteva
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", 64100 Teramo, Italy
| | - Maria Schirone
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy.
| | - Federica D'Onofrio
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", 64100 Teramo, Italy
| | - Luigi Iannetti
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", 64100 Teramo, Italy
| | - Marina Torresi
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", 64100 Teramo, Italy
| | - Chiara Di Pancrazio
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", 64100 Teramo, Italy
| | - Fabrizia Perletta
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", 64100 Teramo, Italy
| | - Silvia Valentinuzzi
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy; Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
| | - Manuela Tittarelli
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", 64100 Teramo, Italy
| | - Francesco Pomilio
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", 64100 Teramo, Italy
| | - Nicola D'Alterio
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", 64100 Teramo, Italy
| | - Antonello Paparella
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Piero Del Boccio
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy; Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
3
|
Hanes R, Liu Y, Huang Z. Druggability Analysis of Protein Targets for Drug Discovery to Combat Listeria monocytogenes. Microorganisms 2024; 12:1073. [PMID: 38930455 PMCID: PMC11205737 DOI: 10.3390/microorganisms12061073] [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: 04/15/2024] [Revised: 05/19/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Extensive research has been conducted to identify key proteins governing stress responses, virulence, and antimicrobial resistance, as well as to elucidate their interactions within Listeria monocytogenes. While these proteins hold promise as potential targets for novel strategies to control L. monocytogenes, given their critical roles in regulating the pathogen's metabolism, additional analysis is needed to further assess their druggability-the chance of being effectively bound by small-molecule inhibitors. In this work, 535 binding pockets of 46 protein targets for known drugs (mainly antimicrobials) were first analyzed to extract 13 structural features (e.g., hydrophobicity) in a ligand-protein docking platform called Molsoft ICM Pro. The extracted features were used as inputs to develop a logistic regression model to assess the druggability of protein binding pockets, with a value of one if ligands can bind to the protein pocket. The developed druggability model was then used to evaluate 23 key proteins from L. monocytogenes that have been identified in the literature. The following proteins are predicted to be high-potential druggable targets: GroEL, FliH/FliI complex, FliG, FlhB, FlgL, FlgK, InlA, MogR, and PrfA. These findings serve as an initial point for future research to identify specific compounds that can inhibit druggable target proteins and to design experimental work to confirm their effectiveness as drug targets.
Collapse
Affiliation(s)
- Robert Hanes
- Department of Chemical and Biological Engineering, Villanova University, Villanova, PA 19085, USA;
| | - Yanhong Liu
- Eastern Regional Research Center, U.S. Department of Agriculture, Wyndmoor, PA 19038, USA
| | - Zuyi Huang
- Department of Chemical and Biological Engineering, Villanova University, Villanova, PA 19085, USA;
| |
Collapse
|
4
|
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
|
5
|
Abdulkadieva MM, Sysolyatina EV, Vasilieva EV, Litvinenko VV, Kalinin EV, Zhukhovitsky VG, Shevlyagina NV, Andreevskaya SG, Stanishevskyi YM, Vasiliev MM, Petrov OF, Ermolaeva SA. Motility provides specific adhesion patterns and improves Listeria monocytogenes invasion into human HEp-2 cells. PLoS One 2023; 18:e0290842. [PMID: 37651463 PMCID: PMC10470941 DOI: 10.1371/journal.pone.0290842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/17/2023] [Indexed: 09/02/2023] Open
Abstract
Listeria monocytogenes is motile at 22°C and non-motile at 37°C. In contrast, expression of L. monocytogenes virulence factors is low at 22°C and up-regulated at 37°C. Here, we studied a character of L. monocytogenes near surface swimming (NSS) motility and its effects on adhesion patterns and invasion into epithelial cells. L. monocytogenes and its saprophytic counterpart L. innocua both grown at 22°C showed similar NSS characteristics including individual velocities, trajectory lengths, residence times, and an asymmetric distribution of velocity directions. Similar NSS patterns correlated with similar adhesion patterns. Motile bacteria, including both pathogenic and saprophytic species, showed a preference for adhering to the periphery of epithelial HEp-2 cells. In contrast, non-motile bacteria were evenly distributed across the cell surface, including areas over the nucleus. However, the uneven distribution of motile bacteria did not enhance the invasion into HEp-2 cells unless virulence factor production was up-regulated by the transient shift of the culture to 37°C. Motile L. monocytogenes grown overnight at 22°C and then shifted to 37°C for 2 h expressed invasion factors at the same level and invaded human cells up to five times more efficiently comparatively with non-motile bacteria grown overnight at 37°C. Taken together, obtained results demonstrated that (i) NSS motility and correspondent peripheral location over the cell surface did not depend on L. monocytogenes virulence traits; (ii) motility improved L. monocytogenes invasion into human HEp-2 cells within a few hours after the transition from the ambient temperature to the human body temperature.
Collapse
Affiliation(s)
- Mariam M. Abdulkadieva
- Department of Infections with Natural Foci, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
- Department of Dusty Plasmas, Joint Institute of High Temperatures RAS, Moscow, Russia
- Institute of Biochemical Technology and Nanotechnology, People’s Friendship University RUDN, Moscow, Russia
| | - Elena V. Sysolyatina
- Department of Infections with Natural Foci, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
| | - Elena V. Vasilieva
- Department of Infections with Natural Foci, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
- Department of Dusty Plasmas, Joint Institute of High Temperatures RAS, Moscow, Russia
| | - Veronika V. Litvinenko
- Department of Infections with Natural Foci, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
- Department of Dusty Plasmas, Joint Institute of High Temperatures RAS, Moscow, Russia
| | - Egor V. Kalinin
- Department of Infections with Natural Foci, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
- Institute of Biochemical Technology and Nanotechnology, People’s Friendship University RUDN, Moscow, Russia
| | - Vladimir G. Zhukhovitsky
- Department of Bacterial Infections, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
- Russian Medical Academy of Continuing Professional Education (RMANPO), Ministry of Public Health, Moscow, Russia
| | - Natalia V. Shevlyagina
- Department of Bacterial Infections, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
| | - Svetlana G. Andreevskaya
- Department of Bacterial Infections, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
| | - Yaroslav M. Stanishevskyi
- Institute of Biochemical Technology and Nanotechnology, People’s Friendship University RUDN, Moscow, Russia
| | - Mikhail M. Vasiliev
- Department of Dusty Plasmas, Joint Institute of High Temperatures RAS, Moscow, Russia
| | - Oleg F. Petrov
- Department of Dusty Plasmas, Joint Institute of High Temperatures RAS, Moscow, Russia
| | - Svetlana A. Ermolaeva
- Department of Infections with Natural Foci, Gamaleya National Research Centre of Epidemiology and Microbiology, Moscow, Russia
| |
Collapse
|
6
|
Hanes R, Zhang F, Huang Z. Protein Interaction Network Analysis to Investigate Stress Response, Virulence, and Antibiotic Resistance Mechanisms in Listeria monocytogenes. Microorganisms 2023; 11:microorganisms11040930. [PMID: 37110353 PMCID: PMC10144942 DOI: 10.3390/microorganisms11040930] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Listeria monocytogenes is a deadly and costly foodborne pathogen that has a high fatality rate in the elderly, pregnant women, and people with weakened immunity. It can survive under various stress conditions and is a significant concern for the food industry. In this work, a data analysis approach was developed with existing tools and databases and used to create individual and combined protein interaction networks to study stress response, virulence, and antimicrobial resistance and their interaction with L. monocytogenes. The networks were analyzed, and 28 key proteins were identified that may serve as potential targets for new strategies to combat L. monocytogenes. Five of the twenty-eight proteins (i.e., sigB, flaA, cheA, cheY, and lmo0693) represent the most promising targets because they are highly interconnected within the combined network. The results of this study provide a new set of targets for future work to identify new strategies to improve food preservation methods and treatments for L. monocytogenes.
Collapse
Affiliation(s)
- Robert Hanes
- Department of Chemical Engineering, Villanova University, Villanova, PA 19085, USA
| | - Fangyuan Zhang
- Department of Chemical Engineering, Villanova University, Villanova, PA 19085, USA
| | - Zuyi Huang
- Department of Chemical Engineering, Villanova University, Villanova, PA 19085, USA
| |
Collapse
|
7
|
Li P, Zong W, Zhang Z, Lv W, Ji X, Zhu D, Du X, Wang S. Effects and molecular mechanism of flagellar gene flgK on the motility, adhesion/invasion, and desiccation resistance of Cronobacter sakazakii. Food Res Int 2023; 164:112418. [PMID: 36738023 DOI: 10.1016/j.foodres.2022.112418] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 12/21/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022]
Abstract
Cronobacter sakazakii (C. sakazakii), a food-borne pathogen, can infect neonates, elderly and immunocompromised populations with a high infection and mortality rate. However, the specific molecular mechanism of its motility, biofilm formation, cell adhesion, and desiccation resistance remains unclear, and flagellum hook associated protein (FlgK), a main component of the flagellar complex, may be an important determinant of its virulence and desiccation resistance. In this study, the flgK mutant strain (ΔflgK) was constructed using the homologous recombination method, and the cpflgK complementary strain was obtained by gene complementation, followed by analysis of the difference between the wild type (WT), mutant, and complementary strains in mobility, biofilm formation, cell adhesion, and desiccation resistance. Results indicated that flgK gene played a positive role in motility and invasion, with no significant effect on biofilm formation. Interestingly, flagellar assembly gene deletion showed increased resistance of C. sakazakii to dehydration. The mechanism underlying the negative correlation of flgK gene with dehydration resistance was further investigated by using the high-throughput sequencing technology to compare the gene expression between WT and ΔflgK strains after drying. The results revealed up-regulation in the expression of 54 genes, including genes involved in osmosis and formate dehydrogenase, while down-regulation in the expression of 50 genes, including genes involved in flagellum hook and nitrate reductase. qRT-PCR analysis of the RNA-seq data further indicated that the flgK gene played an important role in the environmental stress resistance of C. sakazakii by up-regulating the formate dehydrogenase, betaine synthesis, and arginine deiminase pathways, due to dynamic proton imbalance caused by lack of flagella. This study facilitates our understanding of the roles of flgK in motion-related functions and the molecular mechanism of desiccation resistance in C. sakazakii.
Collapse
Affiliation(s)
- Ping Li
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Wenyue Zong
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zhengyang Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Wen Lv
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xuemeng Ji
- Tianjin Key Laboratory of Food Science and Health, College of Medicine, Nankai University, Tianjin 300071, China
| | - Dongdong Zhu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xinjun Du
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China.
| | - Shuo Wang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China; Tianjin Key Laboratory of Food Science and Health, College of Medicine, Nankai University, Tianjin 300071, China.
| |
Collapse
|
8
|
The Transcriptional Regulator SpxA1 Influences the Morphology and Virulence of Listeria monocytogenes. Infect Immun 2022; 90:e0021122. [PMID: 36102657 PMCID: PMC9584327 DOI: 10.1128/iai.00211-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Listeria monocytogenes is a Gram-positive facultative anaerobe and an excellent model pathogen for investigating regulatory changes that occur during infection of a mammalian host. SpxA1 is a widely conserved transcriptional regulator that induces expression of peroxide-detoxifying genes in L. monocytogenes and is thus required for aerobic growth. SpxA1 is also required for L. monocytogenes virulence, although the SpxA1-dependent genes important in this context remain to be identified. Here, we sought to investigate the role of SpxA1 in a tissue culture model of infection and made the surprising discovery that ΔspxA1 cells are dramatically elongated during growth in the host cytosol. Quantitative microscopy revealed that ΔspxA1 cells also form elongated filaments extracellularly during early exponential phase in rich medium. Scanning and transmission electron microscopy analysis found that the likely cause of this morphological phenotype is aberrantly placed division septa localized outside cell midpoints. Quantitative mass spectrometry of whole-cell lysates identified SpxA1-dependent changes in protein abundance, including a significant number of motility and flagellar proteins that were depleted in the ΔspxA1 mutant. Accordingly, we found that both the filamentation and the lack of motility contributed to decreased phagocytosis of ΔspxA1 cells by macrophages. Overall, we identify a novel role for SpxA1 in regulating cell elongation and motility, both of which impact L. monocytogenes virulence.
Collapse
|
9
|
Birk MS, Ahmed-Begrich R, Tran S, Elsholz AKW, Frese CK, Charpentier E. Time-Resolved Proteome Analysis of Listeria monocytogenes during Infection Reveals the Role of the AAA+ Chaperone ClpC for Host Cell Adaptation. mSystems 2021; 6:e0021521. [PMID: 34342529 PMCID: PMC8407217 DOI: 10.1128/msystems.00215-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/14/2021] [Indexed: 12/13/2022] Open
Abstract
The cellular proteome comprises all proteins expressed at a given time and defines an organism's phenotype under specific growth conditions. The proteome is shaped and remodeled by both protein synthesis and protein degradation. Here, we developed a new method which combines metabolic and chemical isobaric peptide labeling to simultaneously determine the time-resolved protein decay and de novo synthesis in an intracellular human pathogen. We showcase this method by investigating the Listeria monocytogenes proteome in the presence and absence of the AAA+ chaperone protein ClpC. ClpC associates with the peptidase ClpP to form an ATP-dependent protease complex and has been shown to play a role in virulence development in L. monocytogenes. However, the mechanism by which ClpC is involved in the survival and proliferation of intracellular L. monocytogenes remains elusive. Employing this new method, we observed extensive proteome remodeling in L. monocytogenes upon interaction with the host, supporting the hypothesis that ClpC-dependent protein degradation is required to initiate bacterial adaptation mechanisms. We identified more than 100 putative ClpC target proteins through their stabilization in a clpC deletion strain. Beyond the identification of direct targets, we also observed indirect effects of the clpC deletion on the protein abundance in diverse cellular and metabolic pathways, such as iron acquisition and flagellar assembly. Overall, our data highlight the crucial role of ClpC for L. monocytogenes adaptation to the host environment through proteome remodeling. IMPORTANCE Survival and proliferation of pathogenic bacteria inside the host depend on their ability to adapt to the changing environment. Profiling the underlying changes on the bacterial proteome level during the infection process is important to gain a better understanding of the pathogenesis and the host-dependent adaptation processes. The cellular protein abundance is governed by the interplay between protein synthesis and decay. The direct readout of these events during infection can be accomplished using pulsed stable-isotope labeling by amino acids in cell culture (SILAC). Combining this approach with tandem-mass-tag (TMT) labeling enabled multiplexed and time-resolved bacterial proteome quantification during infection. Here, we applied this integrated approach to investigate protein turnover during the temporal progression of adaptation of the human pathogen L. monocytogenes to its host on a system-wide scale. Our experimental approach can easily be transferred to probe the proteome remodeling in other bacteria under a variety of perturbations.
Collapse
Affiliation(s)
- Marlène S. Birk
- Max Planck Unit for the Science of Pathogens, Berlin, Germany
| | | | - Stefan Tran
- Max Planck Unit for the Science of Pathogens, Berlin, Germany
| | | | | | | |
Collapse
|
10
|
Janež N, Škrlj B, Sterniša M, Klančnik A, Sabotič J. The role of the Listeria monocytogenes surfactome in biofilm formation. Microb Biotechnol 2021; 14:1269-1281. [PMID: 34106516 PMCID: PMC8313260 DOI: 10.1111/1751-7915.13847] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
Listeria monocytogenes is a highly pathogenic foodborne bacterium that is ubiquitous in the natural environment and capable of forming persistent biofilms in food processing environments. This species has a rich repertoire of surface structures that enable it to survive, adapt and persist in various environments and promote biofilm formation. We review current understanding and advances on how L. monocytogenes organizes its surface for biofilm formation on surfaces associated with food processing settings, because they may be an important target for development of novel antibiofilm compounds. A synthesis of the current knowledge on the role of Listeria surfactome, comprising peptidoglycan, teichoic acids and cell wall proteins, during biofilm formation on abiotic surfaces is provided. We consider indications gained from genome-wide studies and discuss surfactome structures with established mechanistic aspects in biofilm formation. Additionally, we look at the analogies to the species L. innocua, which is closely related to L. monocytogenes and often used as its model (surrogate) organism.
Collapse
Affiliation(s)
- Nika Janež
- Department of BiotechnologyJožef Stefan InstituteLjubljanaSlovenia
| | - Blaž Škrlj
- Department of Knowledge TechnologiesJožef Stefan InstituteLjubljanaSlovenia
- Jožef Stefan International Postgraduate SchoolLjubljanaSlovenia
| | - Meta Sterniša
- Department of Food Science and TechnologyBiotechnical FacultyUniversity of LjubljanaLjubljanaSlovenia
| | - Anja Klančnik
- Department of Food Science and TechnologyBiotechnical FacultyUniversity of LjubljanaLjubljanaSlovenia
| | - Jerica Sabotič
- Department of BiotechnologyJožef Stefan InstituteLjubljanaSlovenia
| |
Collapse
|
11
|
Listeria monocytogenes Cold Shock Proteins: Small Proteins with A Huge Impact. Microorganisms 2021; 9:microorganisms9051061. [PMID: 34068949 PMCID: PMC8155936 DOI: 10.3390/microorganisms9051061] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 01/26/2023] Open
Abstract
Listeria monocytogenes has evolved an extensive array of mechanisms for coping with stress and adapting to changing environmental conditions, ensuring its virulence phenotype expression. For this reason, L. monocytogenes has been identified as a significant food safety and public health concern. Among these adaptation systems are cold shock proteins (Csps), which facilitate rapid response to stress exposure. L. monocytogenes has three highly conserved csp genes, namely, cspA, cspB, and cspD. Using a series of csp deletion mutants, it has been shown that L. monocytogenes Csps are important for biofilm formation, motility, cold, osmotic, desiccation, and oxidative stress tolerance. Moreover, they are involved in overall virulence by impacting the expression of virulence-associated phenotypes, such as hemolysis and cell invasion. It is postulated that during stress exposure, Csps function to counteract harmful effects of stress, thereby preserving cell functions, such as DNA replication, transcription and translation, ensuring survival and growth of the cell. Interestingly, it seems that Csps might suppress tolerance to some stresses as their removal resulted in increased tolerance to stresses, such as desiccation for some strains. Differences in csp roles among strains from different genetic backgrounds are apparent for desiccation tolerance and biofilm production. Additionally, hierarchical trends for the different Csps and functional redundancies were observed on their influences on stress tolerance and virulence. Overall current data suggest that Csps have a wider role in bacteria physiology than previously assumed.
Collapse
|
12
|
Veronica EK, Sara A O, Everardo CQ, Héctor Q, Oscar MC, Elizabeth FR, Irma RP, José AG, Bulmaro C, Rigoberto HC, Juan XC, Ariadnna CC. Proteomics profiles of Cronobacter sakazakii and a fliF mutant: Adherence and invasion in mouse neuroblastoma cells. Microb Pathog 2020; 149:104595. [PMID: 33157215 DOI: 10.1016/j.micpath.2020.104595] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/17/2023]
Abstract
Cronobacter sakazakii is an opportunistic foodborne pathogen associated with necrotizing enterocolitis, bacteremia, and meningitis in infants. A comparative proteomic study of C. sakazakii ATCC BAA-894 (CS WT) and a fliF::Tn5 mutant was performed, including the ability of both strains to adhere to and invade N1E-115 cells. To achieve this goal, a nonmotile C. sakazakii ATCC BAA-894 fliF::Tn5 (CS fliF::Tn5) strain was generated using an EZ-Tn5 <KAN-2>Tnp Transposome kit. Analysis of differential protein expression showed that 81.49% (361/443) of the proteins were expressed in both strains, 8.35% (37/443) were exclusively expressed in the CS WT strain, and 10.16% (45/443) were exclusively expressed in the CS fliF::Tn5 strain. The main exclusively expressed proteins in the CS WT strain were classified into the "cell motility" and "signal transduction mechanisms" subcategories. The proteins exclusively expressed in the CS fliF::Tn5 strain were classified into the following subcategories: "intracellular trafficking, secretion, and vesicular transport", "replication, recombination, and repair", "nucleotide transport and metabolism", "carbohydrate transport and metabolism", "coenzyme transport and metabolism", and "lipid transport and metabolism". Expression of the Cpa protein was detected in both strains, but Cpa was more abundant in the CS WT strain than in the CS fliF::Tn5 strain. A significant increase (p = 0.0001) in adherence to N1E-115 cells was observed in the nonmotile CS fliF::Tn5 strain (31.3 × 106 CFU/mL) compared to the CS WT strain (14.5 × 106 CFU/mL). Additionally, the CS WT strain showed a 0.17% invasion frequency in N1E-115 cells, which was significantly higher (p = 0.01) than that of the nonmotile CS fliF::Tn5 strain. In conclusion, the proteins involved in the motility were mainly identified by proteomic analysis in the CS WT strain compared to the CS fliF::Tn5 strain. Our data indicate that flagella are required to promote the invasion of N1E-115 cells and that the absence of flagella significantly increases the adherence to N1E-115 cells.
Collapse
Affiliation(s)
- Esteban-Kenel Veronica
- Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México Federico Gómez, Ciudad de México, Mexico; Laboratorio de Ingeniería Genética, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Ochoa Sara A
- Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México Federico Gómez, Ciudad de México, Mexico
| | - Curiel-Quesada Everardo
- Laboratorio de Ingeniería Genética, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Quezada Héctor
- Unidad de Investigación Epidemiológica en Endocrinología y Nutrición. Hospital Infantil de México Federico Gómez, Ciudad de México, Mexico
| | - Medina-Contreras Oscar
- Unidad de Investigación Epidemiológica en Endocrinología y Nutrición. Hospital Infantil de México Federico Gómez, Ciudad de México, Mexico
| | - Fernández-Rendón Elizabeth
- Laboratorio de Microbiología Sanitaria, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Rosas-Pérez Irma
- Laboratorio de Aerobiología, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Arellano-Galindo José
- Área de Virología, Laboratorio de Infectología, Hospital Infantil de México Federico Gómez, Ciudad de México, Mexico
| | - Cisneros Bulmaro
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Hernandez-Castro Rigoberto
- Departamento de Ecología de Agentes Patógenos. Hospital General "Dr. Manuel Gea González", Delegación Tlalpan, México D., 14080, Mexico
| | - Xicohtencatl-Cortes Juan
- Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México Federico Gómez, Ciudad de México, Mexico.
| | - Cruz-Córdova Ariadnna
- Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México Federico Gómez, Ciudad de México, Mexico.
| |
Collapse
|
13
|
Ghosh P, Zhou Y, Richardson Q, Higgins DE. Characterization of the pathogenesis and immune response to Listeria monocytogenes strains isolated from a sustained national outbreak. Sci Rep 2019; 9:19587. [PMID: 31862952 PMCID: PMC6925182 DOI: 10.1038/s41598-019-56028-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 12/05/2019] [Indexed: 12/16/2022] Open
Abstract
Listeria monocytogenes is an intracellular pathogen responsible for listeriosis, a foodborne disease that can lead to life-threatening meningitis. The 2011 L. monocytogenes cantaloupe outbreak was among the deadliest foodborne outbreaks in the United States. We conducted in vitro and in vivo infection analyses to determine whether strains LS741 and LS743, two clinical isolates from the cantaloupe outbreak, differ significantly from the common laboratory strain 10403S. We showed that LS741 and LS743 exhibited increased virulence, characterized by higher colonization of the brain and other organs in mice. Assessment of cellular immune responses to known CD8+ T cell antigens was comparable between all strains. However, pre-existing immunity to 10403S did not confer protection in the brain against challenge with LS741. These studies provide insights into the pathogenesis of clinical isolates linked to the 2011 cantaloupe outbreak and also indicate that currently utilized laboratory strains are imperfect models for studying L. monocytogenes pathogenesis.
Collapse
Affiliation(s)
- Pallab Ghosh
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Yan Zhou
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Quentin Richardson
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Darren E Higgins
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Validation of Predicted Virulence Factors in Listeria monocytogenes Identified Using Comparative Genomics. Toxins (Basel) 2019; 11:toxins11090508. [PMID: 31480280 PMCID: PMC6783856 DOI: 10.3390/toxins11090508] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/14/2019] [Accepted: 08/24/2019] [Indexed: 12/19/2022] Open
Abstract
Listeria monocytogenes is an intracellular facultative pathogen that causes listeriosis, a foodborne zoonotic infection. There are differences in the pathogenic potential of L. monocytogenes subtypes and strains. Comparison of the genome sequences among L. monocytogenes pathogenic strains EGD-e and F2365 with nonpathogenic L. innocua CLIP1182 and L. monocytogenes strain HCC23 revealed a set of proteins that were present in pathogenic strains and had no orthologs among the nonpathogenic strains. Among the candidate virulence factors are five proteins: putrescine carbamoyltransferase; InlH/InlC2 family class 1 internalin; phosphotransferase system (PTS) fructose transporter subunit EIIC; putative transketolase; and transcription antiterminator BglG family. To determine if these proteins have a role in adherence and invasion of intestinal epithelial Caco-2 cells and/or contribute to virulence, five mutant strains were constructed. F2365ΔinlC2, F2365Δeiic, and F2365Δtkt exhibited a significant (p < 0.05) reduction in adhesion to Caco-2 cells compared to parent F2365 strain. The invasion of F2365ΔaguB, F2365ΔinlC2, and F2365ΔbglG decreased significantly (p < 0.05) compared with the parent strain. Bacterial loads in mouse liver and spleen infected by F2365 was significantly (p < 0.05) higher than it was for F2365ΔaguB, F2365ΔinlC2, F2365Δeiic, F2365Δtkt, and F2365ΔbglG strains. This study demonstrates that aguB, inlC2, eiic, tkt, and bglG play a role in L. monocytogenes pathogenicity.
Collapse
|
15
|
Gandra TKV, Volcan D, Kroning IS, Marini N, de Oliveira AC, Bastos CP, da Silva WP. Expression levels of the agr locus and prfA gene during biofilm formation by Listeria monocytogenes on stainless steel and polystyrene during 8 to 48 h of incubation 10 to 37 °C. Int J Food Microbiol 2019; 300:1-7. [PMID: 30981136 DOI: 10.1016/j.ijfoodmicro.2019.03.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 02/25/2019] [Accepted: 03/31/2019] [Indexed: 01/25/2023]
Abstract
The objective of this study was to compare the gene expression levels of the agr locus and prfA gene during adhesion and biofilm formation by four L. monocytogenes isolates (2 biofilm-forming and 2 non-forming) on stainless steel and polystyrene surfaces at different temperatures (10 °C, 20 °C and 37 °C), and times (8 h, 12 h, 24 h and 48 h). The agrA and prfA genes were expressed at higher levels than the agrBCD genes. The levels of agr locus expression were higher in the biofilm-forming strains, and the greatest difference between biofilm-forming and non-forming isolates was observed for the agrB, agrC and agrD genes. However, no difference in the expression of the prfA gene was seen among the isolates, independent of the biofilm-forming ability. Maximum expression of the agr locus and prfA gene was observed at 37 °C, whereas expression was lowest at 10 °C. The agr locus, and particularly the agrB, agrC and agrD genes, is important in the initial adhesion phase of biofilm production by L. monocytogenes, with this expression independent of prfA. In addition, the agr locus and prfA gene expression levels were strongly influenced by time and temperature.
Collapse
Affiliation(s)
- Tatiane Kuka Valente Gandra
- Postgraduate Program in Agroindustrial Science and Technology, Agroindustrial Technology and Science Department, Eliseu Maciel Faculty of Agronomy, Federal University of Pelotas (UFPel), 96010-900, P.O. Box 354, Pelotas, RS, Brazil.
| | - Darla Volcan
- Postgraduate Program in Agroindustrial Science and Technology, Agroindustrial Technology and Science Department, Eliseu Maciel Faculty of Agronomy, Federal University of Pelotas (UFPel), 96010-900, P.O. Box 354, Pelotas, RS, Brazil
| | - Isabela Schneid Kroning
- Postgraduate Program in Agroindustrial Science and Technology, Agroindustrial Technology and Science Department, Eliseu Maciel Faculty of Agronomy, Federal University of Pelotas (UFPel), 96010-900, P.O. Box 354, Pelotas, RS, Brazil
| | - Naciele Marini
- Catarinense Federal Institute of Education, Science and Technology, Campus Santa Rosa do Sul, P.O. Box 04, 88965-000, Santa Rosa do Sul, SC, Brazil
| | - Antônio Costa de Oliveira
- Eliseu Maciel Faculty of Agronomy, Federal University of Pelotas (UFPel), 96010-900, P.O. Box 354, Pelotas, RS, Brazil
| | - Caroline Peixoto Bastos
- Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), 96010-900, P.O. Box 354, Pelotas, RS, Brazil
| | - Wladimir Padilha da Silva
- Postgraduate Program in Agroindustrial Science and Technology, Agroindustrial Technology and Science Department, Eliseu Maciel Faculty of Agronomy, Federal University of Pelotas (UFPel), 96010-900, P.O. Box 354, Pelotas, RS, Brazil.
| |
Collapse
|
16
|
Srisanga K, Suthapot P, Permsirivisarn P, Govitrapong P, Tungpradabkul S, Wongtrakoongate P. Polyphosphate kinase 1 of Burkholderia pseudomallei controls quorum sensing, RpoS and host cell invasion. J Proteomics 2019; 194:14-24. [DOI: 10.1016/j.jprot.2018.12.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 12/16/2018] [Accepted: 12/25/2018] [Indexed: 12/18/2022]
|
17
|
Guariglia-Oropeza V, Orsi RH, Guldimann C, Wiedmann M, Boor KJ. The Listeria monocytogenes Bile Stimulon under Acidic Conditions Is Characterized by Strain-Specific Patterns and the Upregulation of Motility, Cell Wall Modification Functions, and the PrfA Regulon. Front Microbiol 2018; 9:120. [PMID: 29467736 PMCID: PMC5808219 DOI: 10.3389/fmicb.2018.00120] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/18/2018] [Indexed: 11/16/2022] Open
Abstract
Listeria monocytogenes uses a variety of transcriptional regulation strategies to adapt to the extra-host environment, the gastrointestinal tract, and the intracellular host environment. While the alternative sigma factor SigB has been proposed to be a key transcriptional regulator that facilitates L. monocytogenes adaptation to the gastrointestinal environment, the L. monocytogenes' transcriptional response to bile exposure is not well-understood. RNA-seq characterization of the bile stimulon was performed in two L. monocytogenes strains representing lineages I and II. Exposure to bile at pH 5.5 elicited a large transcriptomic response with ~16 and 23% of genes showing differential transcription in 10403S and H7858, respectively. The bile stimulon includes genes involved in motility and cell wall modification mechanisms, as well as genes in the PrfA regulon, which likely facilitate survival during the gastrointestinal stages of infection that follow bile exposure. The fact that bile exposure induced the PrfA regulon, but did not induce further upregulation of the SigB regulon (beyond that expected by exposure to pH 5.5), suggests a model where at the earlier stages of gastrointestinal infection (e.g., acid exposure in the stomach), SigB-dependent gene expression plays an important role. Subsequent exposure to bile induces the PrfA regulon, potentially priming L. monocytogenes for subsequent intracellular infection stages. Some members of the bile stimulon showed lineage- or strain-specific distribution when 27 Listeria genomes were analyzed. Even though sigB null mutants showed increased sensitivity to bile, the SigB regulon was not found to be upregulated in response to bile beyond levels expected by exposure to pH 5.5. Comparison of wildtype and corresponding ΔsigB strains newly identified 26 SigB-dependent genes, all with upstream putative SigB-dependent promoters.
Collapse
Affiliation(s)
| | - Renato H Orsi
- Food Safety Laboratory, Department of Food Science, Cornell University, Ithaca, NY, United States
| | - Claudia Guldimann
- Food Safety Laboratory, Department of Food Science, Cornell University, Ithaca, NY, United States
| | - Martin Wiedmann
- Food Safety Laboratory, Department of Food Science, Cornell University, Ithaca, NY, United States
| | - Kathryn J Boor
- Food Safety Laboratory, Department of Food Science, Cornell University, Ithaca, NY, United States
| |
Collapse
|
18
|
Schardt J, Jones G, Müller-Herbst S, Schauer K, D'Orazio SEF, Fuchs TM. Comparison between Listeria sensu stricto and Listeria sensu lato strains identifies novel determinants involved in infection. Sci Rep 2017; 7:17821. [PMID: 29259308 PMCID: PMC5736727 DOI: 10.1038/s41598-017-17570-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/28/2017] [Indexed: 01/01/2023] Open
Abstract
The human pathogen L. monocytogenes and the animal pathogen L. ivanovii, together with four other species isolated from symptom-free animals, form the “Listeria sensu stricto” clade. The members of the second clade, “Listeria sensu lato”, are believed to be solely environmental bacteria without the ability to colonize mammalian hosts. To identify novel determinants that contribute to infection by L. monocytogenes, the causative agent of the foodborne disease listeriosis, we performed a genome comparison of the two clades and found 151 candidate genes that are conserved in the Listeria sensu stricto species. Two factors were investigated further in vitro and in vivo. A mutant lacking an ATP-binding cassette transporter exhibited defective adhesion and invasion of human Caco-2 cells. Using a mouse model of foodborne L. monocytogenes infection, a reduced number of the mutant strain compared to the parental strain was observed in the small intestine and the liver. Another mutant with a defective 1,2-propanediol degradation pathway showed reduced persistence in the stool of infected mice, suggesting a role of 1,2-propanediol as a carbon and energy source of listeriae during infection. These findings reveal the relevance of novel factors for the colonization process of L. monocytogenes.
Collapse
Affiliation(s)
- Jakob Schardt
- ZIEL-Institute for Food & Health, and Lehrstuhl für Mikrobielle Ökologie, Wissenschaftszentrum Weihenstephan, Technische Universität München, Weihenstephaner Berg 3, 85354, Freising, Germany
| | - Grant Jones
- Department of Microbiology, Immunology, & Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - Stefanie Müller-Herbst
- ZIEL-Institute for Food & Health, and Lehrstuhl für Mikrobielle Ökologie, Wissenschaftszentrum Weihenstephan, Technische Universität München, Weihenstephaner Berg 3, 85354, Freising, Germany
| | - Kristina Schauer
- Lehrstuhl für Hygiene und Technologie der Milch, Tiermedizinische Fakultät, Ludwig-Maximilians-Universität München, Schönleutner Str. 8, 85764, Oberschleißheim, Germany
| | - Sarah E F D'Orazio
- Department of Microbiology, Immunology, & Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - Thilo M Fuchs
- ZIEL-Institute for Food & Health, and Lehrstuhl für Mikrobielle Ökologie, Wissenschaftszentrum Weihenstephan, Technische Universität München, Weihenstephaner Berg 3, 85354, Freising, Germany. .,Friedrich-Loeffler-Institut, Institut für Molekulare Pathogenese, Naumburger Str. 96a, 07743, Jena, Germany.
| |
Collapse
|
19
|
Beurmann S, Ushijima B, Videau P, Svoboda CM, Smith AM, Rivers OS, Aeby GS, Callahan SM. Pseudoalteromonas piratica strain OCN003 is a coral pathogen that causes a switch from chronic to acute Montipora white syndrome in Montipora capitata. PLoS One 2017; 12:e0188319. [PMID: 29145488 PMCID: PMC5690655 DOI: 10.1371/journal.pone.0188319] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/03/2017] [Indexed: 12/31/2022] Open
Abstract
Reports of mass coral mortality from disease have increased over the last two decades. Montipora white syndrome (MWS) is a tissue loss disease that has negatively impacted populations of the coral Montipora capitata in Kāne'ohe Bay, Hawai'i. Two types of MWS have been documented; a progressive disease termed chronic MWS (cMWS), that can be caused by Vibrio owensii strain OCN002, and a comparatively faster disease termed acute MWS (aMWS), that can be caused by Vibrio coralliilyticus strain OCN008. M. capitata colonies exhibiting cMWS can spontaneously switch to aMWS in the field. In this study, a novel Pseudoalteromonas species, P. piratica strain OCN003, fulfilled Koch's postulates of disease causation as another etiological agent of aMWS. Additionally, OCN003 induced a switch from cMWS to aMWS on M. capitata in laboratory infection trials. A comparison of OCN003 and Vibrio coralliilyticus strain OCN008, showed that OCN003 was more effective at inducing the cMWS to aMWS switch in M. capitata than OCN008. This study is the first to demonstrate that similar disease signs on one coral species (aMWS on M. capitata) can be caused by multiple pathogens, and describes the first Pseudoalteromonas species that infects coral.
Collapse
Affiliation(s)
- Silvia Beurmann
- Universtiy of Hawaiʻi at Mānoa, Department of Microbiology, Honolulu, HI, United States of America
- Hawaiʻi Institute of Marine Biology, Kāneʻohe, HI, United States of America
| | - Blake Ushijima
- Oregon State University, College of Veterinary Medicine, Corvallis, OR, United States of America
| | - Patrick Videau
- Dakota State University, College of Arts and Sciences, Madison, SD, United States of America
| | - Christina Marie Svoboda
- Universtiy of Hawaiʻi at Mānoa, Department of Microbiology, Honolulu, HI, United States of America
- Hawaiʻi Institute of Marine Biology, Kāneʻohe, HI, United States of America
| | - Ashley Marie Smith
- Universtiy of Hawaiʻi at Mānoa, Department of Microbiology, Honolulu, HI, United States of America
- Hawaiʻi Institute of Marine Biology, Kāneʻohe, HI, United States of America
| | - Orion Silverstar Rivers
- Universtiy of Hawaiʻi at Mānoa, Department of Microbiology, Honolulu, HI, United States of America
| | - Greta Smith Aeby
- Hawaiʻi Institute of Marine Biology, Kāneʻohe, HI, United States of America
| | - Sean Michael Callahan
- Universtiy of Hawaiʻi at Mānoa, Department of Microbiology, Honolulu, HI, United States of America
- Hawaiʻi Institute of Marine Biology, Kāneʻohe, HI, United States of America
| |
Collapse
|
20
|
Eshwar AK, Guldimann C, Oevermann A, Tasara T. Cold-Shock Domain Family Proteins (Csps) Are Involved in Regulation of Virulence, Cellular Aggregation, and Flagella-Based Motility in Listeria monocytogenes. Front Cell Infect Microbiol 2017; 7:453. [PMID: 29124040 PMCID: PMC5662587 DOI: 10.3389/fcimb.2017.00453] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/04/2017] [Indexed: 11/13/2022] Open
Abstract
Cold shock-domain family proteins (Csps) are highly conserved nucleic acid binding proteins regulating the expression of various genes including those involved in stress resistance and virulence in bacteria. We show here that Csps are involved in virulence, cell aggregation and flagella-based extracellular motility of Listeria monocytogenes. A L. monocytogenes mutant deleted in all three csp genes (ΔcspABD) is attenuated with respect to human macrophage infection as well as virulence in a zebrafish infection model. Moreover, this mutant is incapable of aggregation and fails to express surface flagella or exhibit swarming motility. An evaluation of double csp gene deletion mutant (ΔcspBD, ΔcspAD and ΔcspAB) strains that produce single csp genes showed that there is redundancy as well as functional differences among the three L. monocytogenes Csps in their contributions to virulence, cellular aggregation, flagella production, and swarming motility. Protein and mRNA expression analysis further showed impaired expression of key virulence and motility genes in the csp mutants. Our observations at protein and mRNA level suggest Csp-dependent expression regulation of these genes at transcriptional and post-transcriptional levels. In a mutant lacking all csp genes (ΔcspABD) as well as those possessing single csp genes (ΔcspBD, ΔcspAD, and ΔcspAB) we detected reduced levels of proteins or activity as well as transcripts from the prfA, hly, mpl, and plcA genes suggesting a Csp-dependent transcriptional regulation of these genes. These csp mutants also had reduced or completely lacked ActA proteins and cell surface flagella but contained elevated actA and flaA mRNA levels compared to the parental wild type strain suggesting Csp involvement in post-transcriptional regulation of these genes. Overall, our results suggest that Csps contribute to the expression regulation of virulence and flagella-associated genes thereby promoting host pathogenicity, cell aggregation and flagella-based motility processes in L. monocytogenes.
Collapse
Affiliation(s)
- Athmanya K. Eshwar
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Claudia Guldimann
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Anna Oevermann
- Neuropathology—Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Taurai Tasara
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
21
|
InlP, a New Virulence Factor with Strong Placental Tropism. Infect Immun 2016; 84:3584-3596. [PMID: 27736782 DOI: 10.1128/iai.00625-16] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/02/2016] [Indexed: 11/20/2022] Open
Abstract
Intrauterine infection is a major detriment for maternal-child health and occurs despite local mechanisms that protect the maternal-fetal interface from a wide variety of pathogens. The bacterial pathogen Listeria monocytogenes causes spontaneous abortion, stillbirth, and preterm labor in humans and serves as a model for placental pathogenesis. Given the unique immunological environment of the maternal-fetal interface, we hypothesized that virulence determinants with placental tropism are required for infection of this tissue. We performed a genomic screen in pregnant guinea pigs that led to the identification of 201 listerial genes important for infection of the placenta but not maternal liver. Among these genes was lmrg1778 (lmo2470), here named inlP, predicted to encode a secreted protein that belongs to the internalin family. InlP is conserved in virulent L. monocytogenes strains but absent in Listeria species that are nonpathogenic for humans. The intracellular life cycle of L. monocytogenes deficient in inlP (ΔinlP) was not impaired. In guinea pigs and mice, InlP increased the placental bacterial burden by a factor of 3 log10 while having only a minor role in other maternal organs. Furthermore, the ΔinlP strain was attenuated in intracellular growth in primary human placental organ cultures and trophoblasts. InlP is a novel virulence factor for listeriosis with a strong tropism for the placenta. This virulence factor represents a tool for the development of new modalities to prevent and treat infection-related pregnancy complications.
Collapse
|
22
|
Elucidation of the functional role of flagella in virulence and ecological traits of Pseudomonas cichorii using flagella absence (Δ fliJ ) and deficiency (Δ fliI ) mutants. Res Microbiol 2016; 167:262-271. [DOI: 10.1016/j.resmic.2016.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 01/20/2016] [Accepted: 01/20/2016] [Indexed: 02/02/2023]
|
23
|
Upadhyay A, Venkitanarayanan K. In vivo efficacy of trans-cinnamaldehyde, carvacrol, and thymol in attenuating Listeria monocytogenes infection in a Galleria mellonella model. J Nat Med 2016; 70:667-72. [DOI: 10.1007/s11418-016-0990-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 03/25/2016] [Indexed: 11/28/2022]
|
24
|
Gowrishankar S, Sivaranjani M, Kamaladevi A, Ravi AV, Balamurugan K, Karutha Pandian S. Cyclic dipeptide cyclo(l-leucyl-l-prolyl) from marineBacillus amyloliquefaciensmitigates biofilm formation and virulence inListeria monocytogenes. Pathog Dis 2016; 74:ftw017. [DOI: 10.1093/femspd/ftw017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/29/2016] [Indexed: 01/18/2023] Open
|
25
|
Cordero N, Maza F, Navea-Perez H, Aravena A, Marquez-Fontt B, Navarrete P, Figueroa G, González M, Latorre M, Reyes-Jara A. Different Transcriptional Responses from Slow and Fast Growth Rate Strains of Listeria monocytogenes Adapted to Low Temperature. Front Microbiol 2016; 7:229. [PMID: 26973610 PMCID: PMC4772535 DOI: 10.3389/fmicb.2016.00229] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 02/12/2016] [Indexed: 01/12/2023] Open
Abstract
Listeria monocytogenes has become one of the principal foodborne pathogens worldwide. The capacity of this bacterium to grow at low temperatures has opened an interesting field of study in terms of the identification and classification of new strains of L. monocytogenes with different growth capacities at low temperatures. We determined the growth rate at 8°C of 110 strains of L. monocytogenes isolated from different food matrices. We identified a group of slow and fast strains according to their growth rate at 8°C and performed a global transcriptomic assay in strains previously adapted to low temperature. We then identified shared and specific transcriptional mechanisms, metabolic and cellular processes of both groups; bacterial motility was the principal process capable of differentiating the adaptation capacity of L. monocytogenes strains with different ranges of tolerance to low temperatures. Strains belonging to the fast group were less motile, which may allow these strains to achieve a greater rate of proliferation at low temperature.
Collapse
Affiliation(s)
- Ninoska Cordero
- Laboratorio de Microbiología y Probióticos, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile Santiago, Chile
| | - Felipe Maza
- Laboratorio de Microbiología y Probióticos, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile Santiago, Chile
| | - Helen Navea-Perez
- Laboratorio de Microbiología y Probióticos, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile Santiago, Chile
| | - Andrés Aravena
- Department of Molecular Biology and Genetics, Istanbul University Istanbul, Turkey
| | - Bárbara Marquez-Fontt
- Laboratorio de Microbiología y Probióticos, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile Santiago, Chile
| | - Paola Navarrete
- Laboratorio de Microbiología y Probióticos, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile Santiago, Chile
| | - Guillermo Figueroa
- Laboratorio de Microbiología y Probióticos, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile Santiago, Chile
| | - Mauricio González
- Laboratorio de Bioinformática y Expresión Génica, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de ChileSantiago, Chile; Center for Genome Regulation (Fondap 15090007), Universidad de ChileSantiago, Chile
| | - Mauricio Latorre
- Laboratorio de Bioinformática y Expresión Génica, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de ChileSantiago, Chile; Center for Genome Regulation (Fondap 15090007), Universidad de ChileSantiago, Chile; Mathomics, Center for Mathematical Modeling, Universidad de ChileSantiago, Chile
| | - Angélica Reyes-Jara
- Laboratorio de Microbiología y Probióticos, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile Santiago, Chile
| |
Collapse
|
26
|
Lobel L, Herskovits AA. Systems Level Analyses Reveal Multiple Regulatory Activities of CodY Controlling Metabolism, Motility and Virulence in Listeria monocytogenes. PLoS Genet 2016; 12:e1005870. [PMID: 26895237 PMCID: PMC4760761 DOI: 10.1371/journal.pgen.1005870] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/22/2016] [Indexed: 02/05/2023] Open
Abstract
Bacteria sense and respond to many environmental cues, rewiring their regulatory network to facilitate adaptation to new conditions/niches. Global transcription factors that co-regulate multiple pathways simultaneously are essential to this regulatory rewiring. CodY is one such global regulator, controlling expression of both metabolic and virulence genes in Gram-positive bacteria. Branch chained amino acids (BCAAs) serve as a ligand for CodY and modulate its activity. Classically, CodY was considered to function primarily as a repressor under rich growth conditions. However, our previous studies of the bacterial pathogen Listeria monocytogenes revealed that CodY is active also when the bacteria are starved for BCAAs. Under these conditions, CodY loses the ability to repress genes (e.g., metabolic genes) and functions as a direct activator of the master virulence regulator gene, prfA. This observation raised the possibility that CodY possesses multiple functions that allow it to coordinate gene expression across a wide spectrum of metabolic growth conditions, and thus better adapt bacteria to the mammalian niche. To gain a deeper understanding of CodY's regulatory repertoire and identify direct target genes, we performed a genome wide analysis of the CodY regulon and DNA binding under both rich and minimal growth conditions, using RNA-Seq and ChIP-Seq techniques. We demonstrate here that CodY is indeed active (i.e., binds DNA) under both conditions, serving as a repressor and activator of different genes. Further, we identified new genes and pathways that are directly regulated by CodY (e.g., sigB, arg, his, actA, glpF, gadG, gdhA, poxB, glnR and fla genes), integrating metabolism, stress responses, motility and virulence in L. monocytogenes. This study establishes CodY as a multifaceted factor regulating L. monocytogenes physiology in a highly versatile manner.
Collapse
Affiliation(s)
- Lior Lobel
- The Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Anat A. Herskovits
- The Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| |
Collapse
|
27
|
Halbedel S, Reiss S, Hahn B, Albrecht D, Mannala GK, Chakraborty T, Hain T, Engelmann S, Flieger A. A systematic proteomic analysis of Listeria monocytogenes house-keeping protein secretion systems. Mol Cell Proteomics 2014; 13:3063-81. [PMID: 25056936 DOI: 10.1074/mcp.m114.041327] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Listeria monocytogenes is a firmicute bacterium causing serious infections in humans upon consumption of contaminated food. Most of its virulence factors are secretory proteins either released to the medium or attached to the bacterial surface. L. monocytogenes encodes at least six different protein secretion pathways. Although great efforts have been made in the past to predict secretory proteins and their secretion routes using bioinformatics, experimental evidence is lacking for most secretion systems. Therefore, we constructed mutants in the main housekeeping protein secretion systems, which are the Sec-dependent transport, the YidC membrane insertases SpoIIIJ and YqjG, as well as the twin-arginine pathway, and analyzed their secretion and virulence defects. Our results demonstrate that Sec-dependent secretion and membrane insertion of proteins via YidC proteins are essential for viability of L. monocytogenes. Depletion of SecA or YidC activity severely affected protein secretion, whereas loss of the Tat-pathway was without any effect on secretion, viability, and virulence. Two-dimensional gel electrophoresis combined with protein identification by mass spectrometry revealed that secretion of many virulence factors and of enzymes synthesizing and degrading the cell wall depends on the SecA route. This finding was confirmed by SecA inhibition experiments using sodium azide. Analysis of secretion of substrates typically dependent on the accessory SecA2 ATPase in wild type and azide resistant mutants of L. monocytogenes revealed for the first time that SecA2-dependent protein secretion also requires the ATPase activity of the house-keeping SecA protein.
Collapse
Affiliation(s)
- Sven Halbedel
- From the ‡Robert Koch Institute, FG11 - Division of Enteropathogenic Bacteria and Legionella, Burgstrasse 37, 38855 Wernigerode, Germany;
| | - Swantje Reiss
- §Institute of Microbiology, University of Greifswald, F.-L.-Jahn-Strasse 15, 17487 Greifswald, Germany
| | - Birgit Hahn
- From the ‡Robert Koch Institute, FG11 - Division of Enteropathogenic Bacteria and Legionella, Burgstrasse 37, 38855 Wernigerode, Germany
| | - Dirk Albrecht
- §Institute of Microbiology, University of Greifswald, F.-L.-Jahn-Strasse 15, 17487 Greifswald, Germany
| | - Gopala Krishna Mannala
- ¶Institute of Medical Microbiology, University of Gießen, Schubertstrasse 81, 35392 Gießen, Germany
| | - Trinad Chakraborty
- ¶Institute of Medical Microbiology, University of Gießen, Schubertstrasse 81, 35392 Gießen, Germany
| | - Torsten Hain
- ¶Institute of Medical Microbiology, University of Gießen, Schubertstrasse 81, 35392 Gießen, Germany
| | - Susanne Engelmann
- §Institute of Microbiology, University of Greifswald, F.-L.-Jahn-Strasse 15, 17487 Greifswald, Germany; ‖Institute of Microbiology, Technical University of Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany; **Helmholtz Centre for Infection Research, Microbial Proteomics, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Antje Flieger
- From the ‡Robert Koch Institute, FG11 - Division of Enteropathogenic Bacteria and Legionella, Burgstrasse 37, 38855 Wernigerode, Germany;
| |
Collapse
|
28
|
Carvalho F, Sousa S, Cabanes D. How Listeria monocytogenes organizes its surface for virulence. Front Cell Infect Microbiol 2014; 4:48. [PMID: 24809022 PMCID: PMC4010754 DOI: 10.3389/fcimb.2014.00048] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/02/2014] [Indexed: 02/04/2023] Open
Abstract
Listeria monocytogenes is a Gram-positive pathogen responsible for the manifestation of human listeriosis, an opportunistic foodborne disease with an associated high mortality rate. The key to the pathogenesis of listeriosis is the capacity of this bacterium to trigger its internalization by non-phagocytic cells and to survive and even replicate within phagocytes. The arsenal of virulence proteins deployed by L. monocytogenes to successfully promote the invasion and infection of host cells has been progressively unveiled over the past decades. A large majority of them is located at the cell envelope, which provides an interface for the establishment of close interactions between these bacterial factors and their host targets. Along the multistep pathways carrying these virulence proteins from the inner side of the cytoplasmic membrane to their cell envelope destination, a multiplicity of auxiliary proteins must act on the immature polypeptides to ensure that they not only maturate into fully functional effectors but also are placed or guided to their correct position in the bacterial surface. As the major scaffold for surface proteins, the cell wall and its metabolism are critical elements in listerial virulence. Conversely, the crucial physical support and protection provided by this structure make it an ideal target for the host immune system. Therefore, mechanisms involving fine modifications of cell envelope components are activated by L. monocytogenes to render it less recognizable by the innate immunity sensors or more resistant to the activity of antimicrobial effectors. This review provides a state-of-the-art compilation of the mechanisms used by L. monocytogenes to organize its surface for virulence, with special focus on those proteins that work “behind the frontline”, either supporting virulence effectors or ensuring the survival of the bacterium within its host.
Collapse
Affiliation(s)
- Filipe Carvalho
- Group of Molecular Microbiology, Unit of Infection and Immunity, Instituto de Biologia Molecular e Celular, University of Porto Porto, Portugal
| | - Sandra Sousa
- Group of Molecular Microbiology, Unit of Infection and Immunity, Instituto de Biologia Molecular e Celular, University of Porto Porto, Portugal
| | - Didier Cabanes
- Group of Molecular Microbiology, Unit of Infection and Immunity, Instituto de Biologia Molecular e Celular, University of Porto Porto, Portugal
| |
Collapse
|
29
|
Wright E, Neethirajan S, Warriner K, Retterer S, Srijanto B. Single cell swimming dynamics of Listeria monocytogenes using a nanoporous microfluidic platform. LAB ON A CHIP 2014; 14:938-946. [PMID: 24399451 DOI: 10.1039/c3lc51138c] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Listeria monocytogenes remains a significant foodborne pathogen due to its virulence and ability to become established in food processing facilities. The pathogen is characterized by its ability to grow over a wide temperature range and withstand a broad range of stresses. The following reports on the chemotaxis and motility of the L. monocytogenes when exposed to relatively small concentrations of acetic acid. Using the developed nanoporous microfluidic device to precisely modulate the cellular environment, we exposed the individual Listeria cells to acetic acid and, in real time and with high resolution, observed how the cells reacted to the change in their surroundings. Our results showed that concentrations of acetic acid below 10 mM had very little, if any, effect on the motility. However, when exposed to 100 mM acetic acid, the cells exhibited a sharp drop in velocity and displayed a more random pattern of motion. These results indicate that at appropriate concentrations, acetic acid has the ability to disable the flagellum of the cells, thus impairing their motility. This drop in motility has numerous effects on the cell; its main effects being the obstruction of the cell's ability to properly form biofilms and a reduction in the overall infectivity of the cells. Since these characteristics are especially useful in controlling the proliferation of L. monocytogenes, acetic acid shows potential for application in the food industry as an active compound in designing a food packaging environment and as an antimicrobial agent.
Collapse
Affiliation(s)
- Evan Wright
- BioNano Lab, School of Engineering, University of Guelph, Ontario, Guelph, N1G 2W1, Canada.
| | | | | | | | | |
Collapse
|
30
|
Oliver HF, Orsi RH, Wiedmann M, Boor KJ. σ(B) plays a limited role in the ability of Listeria monocytogenes strain F2365 to survive oxidative and acid stress and in its virulence characteristics. J Food Prot 2013; 76:2079-86. [PMID: 24290686 DOI: 10.4315/0362-028x.jfp-12-542] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Listeria monocytogenes strain F2365 was the first strain representative of serotype 4b (lineage I) to be sequenced in 2004, suggesting it could become the model organism for this serotype, which is associated with most human outbreaks of listeriosis worldwide to date. F2365 itself is an outbreak strain that was involved in the listeriosis outbreak associated with Mexican-style soft cheese in California in 1985. In this study, we show through phenotypic and transcriptomic analysis that L. monocytogenes strain F2365 has reduced ability to respond to acid and oxidative stress. F2365 has neither the σ(B)-dependent ability to survive acid or oxidative stress nor the σ(B)-dependent ability to infect Caco-2 epithelial cells in vitro or guinea pigs in vivo. More studies are needed to determine whether the atypical σ(B)-independent response to stress observed in F2365 is strain specific, serotype specific, or even lineage specific.
Collapse
Affiliation(s)
- H F Oliver
- Department of Food Science, Purdue University, West Lafayette, Indiana 47907, USA.
| | | | | | | |
Collapse
|
31
|
Bergmann S, Rohde M, Schughart K, Lengeling A. The bioluminescent Listeria monocytogenes strain Xen32 is defective in flagella expression and highly attenuated in orally infected BALB/cJ mice. Gut Pathog 2013; 5:19. [PMID: 23856386 PMCID: PMC3720536 DOI: 10.1186/1757-4749-5-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 06/28/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In vivo bioluminescence imaging (BLI) is a powerful method for the analysis of host-pathogen interactions in small animal models. The commercially available bioluminescent Listeria monocytogenes strain Xen32 is commonly used to analyse immune functions in knockout mice and pathomechanisms of listeriosis. FINDINGS To analyse and image listerial dissemination after oral infection we have generated a murinised Xen32 strain (Xen32-mur) which expresses a previously described mouse-adapted internalin A. This strain was used alongside the Xen32 wild type strain and the bioluminescent L. monocytogenes strains EGDe-lux and murinised EGDe-mur-lux to characterise bacterial dissemination in orally inoculated BALB/cJ mice. After four days of infection, Xen32 and Xen32-mur infected mice displayed consistently higher rates of bioluminescence compared to EGDe-lux and EGDe-mur-lux infected animals. However, surprisingly both Xen32 strains showed attenuated virulence in orally infected BALB/c mice that correlated with lower bacterial burden in internal organs at day 5 post infection, smaller losses in body weights and increased survival compared to EGDe-lux or EGDe-mur-lux inoculated animals. The Xen32 strain was made bioluminescent by integration of a lux-kan transposon cassette into the listerial flaA locus. We show here that this integration results in Xen32 in a flaA frameshift mutation which makes this strain flagella deficient. CONCLUSIONS The bioluminescent L. monocytogenes strain Xen32 is deficient in flagella expression and highly attenuated in orally infected BALB/c mice. As this listerial strain has been used in many BLI studies of murine listeriosis, it is important that the scientific community is aware of its reduced virulence in vivo.
Collapse
Affiliation(s)
- Silke Bergmann
- Department of Infection Genetics, Helmholtz Centre for Infection Research & University of Veterinary Medicine Hannover, Braunschweig D-38124, Germany
| | - Manfred Rohde
- Department of Medical Microbiology, Helmholtz Centre for Infection Research, Braunschweig D-38124, Germany
| | - Klaus Schughart
- Department of Infection Genetics, Helmholtz Centre for Infection Research & University of Veterinary Medicine Hannover, Braunschweig D-38124, Germany
- University of Tennessee Health Science Center, Memphis, TN, USA
| | - Andreas Lengeling
- Infection and Immunity Division, The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Veterinary Campus, Edinburgh EH25 9RG, UK
| |
Collapse
|
32
|
Protein export according to schedule: architecture, assembly, and regulation of type III secretion systems from plant- and animal-pathogenic bacteria. Microbiol Mol Biol Rev 2012; 76:262-310. [PMID: 22688814 DOI: 10.1128/mmbr.05017-11] [Citation(s) in RCA: 312] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Flagellar and translocation-associated type III secretion (T3S) systems are present in most gram-negative plant- and animal-pathogenic bacteria and are often essential for bacterial motility or pathogenicity. The architectures of the complex membrane-spanning secretion apparatuses of both systems are similar, but they are associated with different extracellular appendages, including the flagellar hook and filament or the needle/pilus structures of translocation-associated T3S systems. The needle/pilus is connected to a bacterial translocon that is inserted into the host plasma membrane and mediates the transkingdom transport of bacterial effector proteins into eukaryotic cells. During the last 3 to 5 years, significant progress has been made in the characterization of membrane-associated core components and extracellular structures of T3S systems. Furthermore, transcriptional and posttranscriptional regulators that control T3S gene expression and substrate specificity have been described. Given the architecture of the T3S system, it is assumed that extracellular components of the secretion apparatus are secreted prior to effector proteins, suggesting that there is a hierarchy in T3S. The aim of this review is to summarize our current knowledge of T3S system components and associated control proteins from both plant- and animal-pathogenic bacteria.
Collapse
|
33
|
Upadhyay A, Johny AK, Amalaradjou MAR, Ananda Baskaran S, Kim KS, Venkitanarayanan K. Plant-derived antimicrobials reduce Listeria monocytogenes virulence factors in vitro, and down-regulate expression of virulence genes. Int J Food Microbiol 2012; 157:88-94. [DOI: 10.1016/j.ijfoodmicro.2012.04.018] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 02/28/2012] [Accepted: 04/21/2012] [Indexed: 11/26/2022]
|
34
|
Hain T, Ghai R, Billion A, Kuenne CT, Steinweg C, Izar B, Mohamed W, Mraheil MA, Domann E, Schaffrath S, Kärst U, Goesmann A, Oehm S, Pühler A, Merkl R, Vorwerk S, Glaser P, Garrido P, Rusniok C, Buchrieser C, Goebel W, Chakraborty T. Comparative genomics and transcriptomics of lineages I, II, and III strains of Listeria monocytogenes. BMC Genomics 2012; 13:144. [PMID: 22530965 PMCID: PMC3464598 DOI: 10.1186/1471-2164-13-144] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 04/12/2012] [Indexed: 12/13/2022] Open
Abstract
Background Listeria monocytogenes is a food-borne pathogen that causes infections with a high-mortality rate and has served as an invaluable model for intracellular parasitism. Here, we report complete genome sequences for two L. monocytogenes strains belonging to serotype 4a (L99) and 4b (CLIP80459), and transcriptomes of representative strains from lineages I, II, and III, thereby permitting in-depth comparison of genome- and transcriptome -based data from three lineages of L. monocytogenes. Lineage III, represented by the 4a L99 genome is known to contain strains less virulent for humans. Results The genome analysis of the weakly pathogenic L99 serotype 4a provides extensive evidence of virulence gene decay, including loss of several important surface proteins. The 4b CLIP80459 genome, unlike the previously sequenced 4b F2365 genome harbours an intact inlB invasion gene. These lineage I strains are characterized by the lack of prophage genes, as they share only a single prophage locus with other L. monocytogenes genomes 1/2a EGD-e and 4a L99. Comparative transcriptome analysis during intracellular growth uncovered adaptive expression level differences in lineages I, II and III of Listeria, notable amongst which was a strong intracellular induction of flagellar genes in strain 4a L99 compared to the other lineages. Furthermore, extensive differences between strains are manifest at levels of metabolic flux control and phosphorylated sugar uptake. Intriguingly, prophage gene expression was found to be a hallmark of intracellular gene expression. Deletion mutants in the single shared prophage locus of lineage II strain EGD-e 1/2a, the lma operon, revealed severe attenuation of virulence in a murine infection model. Conclusion Comparative genomics and transcriptome analysis of L. monocytogenes strains from three lineages implicate prophage genes in intracellular adaptation and indicate that gene loss and decay may have led to the emergence of attenuated lineages.
Collapse
Affiliation(s)
- Torsten Hain
- Institute of Medical Microbiology, Justus-Liebig-University, Schubertstrasse 81, Giessen, D-35392, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Halbedel S, Hahn B, Daniel RA, Flieger A. DivIVA affects secretion of virulence-related autolysins in Listeria monocytogenes. Mol Microbiol 2012; 83:821-39. [PMID: 22353466 DOI: 10.1111/j.1365-2958.2012.07969.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
DivIVA is a well-conserved coiled-coil protein present in most Gram-positive bacteria and has been implicated in division site selection, peptidoglycan biosynthesis and sporulation. DivIVA proteins bind lipid membranes and characteristically accumulate at curved membrane areas, i.e. the cell poles and the division site, to which they recruit various interaction partners. We have studied the role of this morphogen in the human pathogen Listeria monocytogenes and our results suggest a novel mechanism by which DivIVA contributes to cell division. Contrary to expectation a ΔdivIVA mutant exhibited a pronounced chaining phenotype rather than a defect in cell division which we attributed to reduced extracellular levels of the autolytic enzymes p60 and MurA. We demonstrate that this is due to a malfunction in secretion of these autolysins and phenotypic comparison of the ΔdivIVA strain with a ΔsecA2 mutant suggests that DivIVA influences the activity of the SecA2 secretion route in L. monocytogenes. Also from the phenotypic analysis it was clear that divIVA affected swarming motility, biofilm formation, invasiveness and cell-to-cell spread in cell culture infection models. Thus, our experiments show that DivIVA is an important factor for various listerial traits that are essential for the pathogenicity of this organism.
Collapse
Affiliation(s)
- Sven Halbedel
- Robert Koch Institute, FG11 - Division of Bacterial Infections, Burgstrasse 37, 38855 Wernigerode, Germany.
| | | | | | | |
Collapse
|
36
|
Xiao Q, Jiang X, Moore KJ, Shao Y, Pi H, Dubail I, Charbit A, Newton SM, Klebba PE. Sortase independent and dependent systems for acquisition of haem and haemoglobin in Listeria monocytogenes. Mol Microbiol 2011; 80:1581-97. [PMID: 21545655 PMCID: PMC3115469 DOI: 10.1111/j.1365-2958.2011.07667.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We studied three Fur-regulated systems of Listeria monocytogenes: the srtB region, that encodes sortase-anchored proteins and a putative ABC transporter, and the fhu and hup operons, that produce putative ABC transporters for ferric hydroxamates and haemin (Hn)/haemoglobin (Hb) respectively. Deletion of lmo2185 in the srtB region reduced listerial [(59) Fe]-Hn transport, and purified Lmo2185 bound [(59) Fe]-Hn (K(D) = 12 nM), leading to its designation as a Hn/Hb binding protein (hbp2). Purified Hbp2 also acted as a haemophore, capturing and supplying Hn from the environment. Nevertheless, Hbp2 only functioned in [(59) Fe]-Hn transport at external concentrations less than 50 nM: at higher Hn levels its uptake occurred with equivalent affinity and rate without Hbp2. Similarly, deletion of sortase A had no effect on ferric siderophore or Hn/Hb transport at any concentration, and the srtA-independence of listerial Hn/Hb uptake distinguished it from comparable systems of Staphylococcus aureus. In the cytoplasmic membrane, the Hup transporter was specific for Hn: its lipoprotein (HupD) only showed high affinity for the iron porphyrin (K(D) = 26 nM). Conversely, the FhuD lipoprotein encoded by the fhu operon had broad specificity: it bound both ferric siderophores and Hn, with the highest affinity for ferrioxamine B (K(D) = 123 nM). Deletions of Hup permease components hupD, hupG or hupDGC reduced Hn/Hb uptake, and complementation of ΔhupC and ΔhupG by chromosomal integration of hupC(+) and hupG(+) alleles on pPL2 restored growth promotion by Hn/Hb. However, ΔhupDGC did not completely eliminate [(59) Fe]-Hn transport, implying the existence of another cytoplasmic membrane Hn transporter. The overall K(M) of Hn uptake by wild-type strain EGD-e was 1 nM, and it occurred at similar rates (V(max) = 23 pmol 10(9) cells(-1) min(-1)) to those of ferric siderophore transporters. In the ΔhupDGC strain uptake occurred at a threefold lower rate (V(max) = 7 pmol 10(9) cells(-1) min(-1)). The results show that at low (< 50 nM) levels of Hn, SrtB-dependent peptidoglycan-anchored proteins (e.g. Hbp2) bind the porphyrin, and HupDGC or another transporter completes its uptake into the cytoplasm. However, at higher concentrations Hn uptake is SrtB-independent: peptidoglycan-anchored binding proteins are dispensable because HupDGC directly absorbs and internalizes Hn. Finally, ΔhupDGC increased the LD(50) of L. monocytogenes 100-fold in the mouse infection model, reiterating the importance of this system in listerial virulence.
Collapse
Affiliation(s)
- Qiaobin Xiao
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Xiaoxu Jiang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Kyle J. Moore
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Yi Shao
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Hualiang Pi
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Iharilalao Dubail
- Université Paris Descartes, Faculté de Médecine, Necker-Enfants Malades, Paris, FRANCE
- INSERM, U1002, Unité de Pathogénie des Infections Systémiques, Paris, FRANCE
| | - Alain Charbit
- Université Paris Descartes, Faculté de Médecine, Necker-Enfants Malades, Paris, FRANCE
- INSERM, U1002, Unité de Pathogénie des Infections Systémiques, Paris, FRANCE
| | - Salete M. Newton
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Phillip E. Klebba
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| |
Collapse
|
37
|
Diverse geno- and phenotypes of persistent Listeria monocytogenes isolates from fermented meat sausage production facilities in Portugal. Appl Environ Microbiol 2011; 77:2701-15. [PMID: 21378045 DOI: 10.1128/aem.02553-10] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The persistence of Listeria monocytogenes in food-associated environments represents a key factor in transmission of this pathogen. To identify persistent and transient strains associated with production of fermented meat sausages in northern Portugal, 1,723 L. monocytogenes isolates from raw material and finished products from 11 processors were initially characterized by random amplification of polymorphic DNA (RAPD), PCR-based molecular serotyping, and epidemic clone characterization, as well as cadmium, arsenic, and tetracycline resistance typing. Pulsed-field gel electrophoresis (PFGE) typing of 240 representative isolates provided evidence for persistence of L. monocytogenes for periods of time ranging from 10 to 32 months for all seven processors for which isolates from different production dates were available. Among 50 L. monocytogenes isolates that included one representative for each PFGE pattern obtained from a given sample, 12 isolates showed reduced invasion efficiency in Caco-2 cells, including 8 isolates with premature stop codons in inlA. Among 41 isolates representing sporadic and persistent PFGE types, 22 isolates represented lysogens. Neither strains with reduced invasion nor lysogens were overrepresented among persistent isolates. While the susceptibility of isolates to lysogenic phages also did not correlate with persistence, it appeared to be associated with molecular serotype. Our data show the following. (i) RAPD may not be suitable for analysis of large sets of L. monocytogenes isolates. (ii) While a large diversity of L. monocytogenes subtypes is found in Portuguese fermented meat sausages, persistence of L. monocytogenes in this food chain is common. (iii) Persistent L. monocytogenes strains are diverse and do not appear to be characterized by unique genetic or phenotypic characteristics.
Collapse
|
38
|
Waltman P, Kacmarczyk T, Bate AR, Kearns DB, Reiss DJ, Eichenberger P, Bonneau R. Multi-species integrative biclustering. Genome Biol 2010; 11:R96. [PMID: 20920250 PMCID: PMC2965388 DOI: 10.1186/gb-2010-11-9-r96] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 09/19/2010] [Accepted: 09/29/2010] [Indexed: 12/22/2022] Open
Abstract
We describe an algorithm, multi-species cMonkey, for the simultaneous biclustering of heterogeneous multiple-species data collections and apply the algorithm to a group of bacteria containing Bacillus subtilis, Bacillus anthracis, and Listeria monocytogenes. The algorithm reveals evolutionary insights into the surprisingly high degree of conservation of regulatory modules across these three species and allows data and insights from well-studied organisms to complement the analysis of related but less well studied organisms.
Collapse
Affiliation(s)
- Peter Waltman
- Computer Science Department, Warren Weaver Hall (Room 305), 251 Mercer Street, New York, NY 10012, USA.
| | | | | | | | | | | | | |
Collapse
|
39
|
Schuppler M, Loessner MJ. The Opportunistic Pathogen Listeria monocytogenes: Pathogenicity and Interaction with the Mucosal Immune System. Int J Inflam 2010; 2010:704321. [PMID: 21188219 PMCID: PMC3003996 DOI: 10.4061/2010/704321] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Accepted: 06/01/2010] [Indexed: 12/22/2022] Open
Abstract
Listeria monocytogenes is an opportunistic foodborne pathogen causing listeriosis, an often fatal infection leading to meningitis, sepsis, or infection of the fetus and abortion in susceptible individuals. It was recently found that the bacterium can also cause acute, self-limiting febrile gastroenteritis in healthy individuals. In the intestinal tract, L. monocytogenes penetrates the mucosa directly via enterocytes, or indirectly via invasion of Peyer's patches. Animal models for L. monocytogenes infection have provided many insights into the mechanisms of pathogenesis, and the development of new model systems has allowed the investigation of factors that influence adaptation to the gastrointestinal environment as well as adhesion to and invasion of the intestinal mucosa. The mucosal surfaces of the gastrointestinal tract are permanently exposed to an enormous antigenic load derived from the gastrointestinal microbiota present in the human bowel. The integrity of the important epithelial barrier is maintained by the mucosal immune system and its interaction with the commensal flora via pattern recognition receptors (PRRs). Here, we discuss recent advances in our understanding of the interaction of L. monocytogenes with the host immune system that triggers the antibacterial immune responses on the mucosal surfaces of the human gastrointestinal tract.
Collapse
Affiliation(s)
- Markus Schuppler
- Institute of Food, Nutrition and Health, ETH Zurich, Schmelzbergstraße 7, 8092 Zurich, Switzerland
| | | |
Collapse
|
40
|
Duodu S, Holst-Jensen A, Skjerdal T, Cappelier JM, Pilet MF, Loncarevic S. Influence of storage temperature on gene expression and virulence potential of Listeria monocytogenes strains grown in a salmon matrix. Food Microbiol 2010; 27:795-801. [PMID: 20630322 DOI: 10.1016/j.fm.2010.04.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Revised: 03/29/2010] [Accepted: 04/20/2010] [Indexed: 11/27/2022]
Abstract
Little is understood about the impact of environmental conditions on the virulence plasticity of Listeria monocytogenes strains grown in food. In this report, we monitored changes in the virulence properties of one high virulent (CCUG 3998) and one low virulent (442) L. monocytogenes strains grown on raw salmon (Salmo salar L.). The effect of temperature exposures (0 degrees C, 4 degrees C and 20 degrees C) on the expression levels of virulence genes (hlyA, actA, inlA and prfA), invasion into Caco-2 cells and in vivo mouse infection was analysed. Our results showed that L. monocytogenes virulence genes are differentially expressed when salmon is stored at different temperatures. Of the four virulence genes, the transcript levels for inlA were strongly affected, which correlated with the strain's virulence capacity as assessed by Caco-2 cells. In contrast to CCUG 3998, the virulence of strain 442 was altered with tested conditions. This strain maintains its low virulence status as far as salmon is stored at lower temperatures, but increases its virulence at higher temperatures. These results lead to the indication that exposure to abuse temperature conditions might influence the virulence potential of low pathogenic L. monocytogenes strains in salmon.
Collapse
Affiliation(s)
- Samuel Duodu
- National Veterinary Institute, Ullevaalsveien 68, Sentrum N-0106, Oslo, Norway.
| | | | | | | | | | | |
Collapse
|
41
|
Comparison of invasiveness among surface-adherent variants of Listeria monocytogenes in Caco-2 cell culture assays. Int J Food Microbiol 2010; 138:166-71. [DOI: 10.1016/j.ijfoodmicro.2009.11.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 11/19/2009] [Accepted: 11/25/2009] [Indexed: 11/20/2022]
|
42
|
Stone CB, Bulir DC, Gilchrist JD, Toor RK, Mahony JB. Interactions between flagellar and type III secretion proteins in Chlamydia pneumoniae. BMC Microbiol 2010; 10:18. [PMID: 20096108 PMCID: PMC2830194 DOI: 10.1186/1471-2180-10-18] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Accepted: 01/22/2010] [Indexed: 11/30/2022] Open
Abstract
Background Flagellar secretion systems are utilized by a wide variety of bacteria to construct the flagellum, a conserved apparatus that allows for migration towards non-hostile, nutrient rich environments. Chlamydia pneumoniae is an obligate, intracellular pathogen whose genome contains at least three orthologs of flagellar proteins, namely FliI, FlhA and FliF, but the role of these proteins remains unknown. Results Full length FliI, and fragments of FlhA, FliF, and FliI, were cloned and expressed as either GST or His tagged proteins in E. coli. The GST-tagged full length FliI protein was shown to possess ATPase activity, hydrolyzing ATP at a rate of 0.15 ± .02 μmol min-1 mg-1 in a time- and dose-dependant manner. Using bacterial-2-hybrid and GST pull-down assays, the N-terminal domain of FliI was shown to interact with the cytoplasmic domain of FlhA, but not with FliF, and the cytoplasmic domain of FlhA was shown to interact with the C-terminus of FliF. The absence of other flagellar orthologs led us to explore cross-reaction of flagellar proteins with type III secretion proteins, and we found that FliI interacted with CdsL and CopN, while FlhA interacted with CdsL and Cpn0322 (YscU ortholog CdsU). Conclusions The specific interaction of the four orthologous flagellar proteins in C. pneumoniae suggests that they interact in vivo and, taken together with their conservation across members of the chlamydiae sps., and their interaction with T3S components, suggests a role in bacterial replication and/or intracellular survival.
Collapse
Affiliation(s)
- Chris B Stone
- M,G, DeGroote Institute for Infectious Disease Research, Faculty of Health Sciences and the Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | |
Collapse
|
43
|
Oliver HF, Orsi RH, Ponnala L, Keich U, Wang W, Sun Q, Cartinhour SW, Filiatrault MJ, Wiedmann M, Boor KJ. Deep RNA sequencing of L. monocytogenes reveals overlapping and extensive stationary phase and sigma B-dependent transcriptomes, including multiple highly transcribed noncoding RNAs. BMC Genomics 2009; 10:641. [PMID: 20042087 PMCID: PMC2813243 DOI: 10.1186/1471-2164-10-641] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Accepted: 12/30/2009] [Indexed: 11/30/2022] Open
Abstract
Background Identification of specific genes and gene expression patterns important for bacterial survival, transmission and pathogenesis is critically needed to enable development of more effective pathogen control strategies. The stationary phase stress response transcriptome, including many σB-dependent genes, was defined for the human bacterial pathogen Listeria monocytogenes using RNA sequencing (RNA-Seq) with the Illumina Genome Analyzer. Specifically, bacterial transcriptomes were compared between stationary phase cells of L. monocytogenes 10403S and an otherwise isogenic ΔsigB mutant, which does not express the alternative σ factor σB, a major regulator of genes contributing to stress response, including stresses encountered upon entry into stationary phase. Results Overall, 83% of all L. monocytogenes genes were transcribed in stationary phase cells; 42% of currently annotated L. monocytogenes genes showed medium to high transcript levels under these conditions. A total of 96 genes had significantly higher transcript levels in 10403S than in ΔsigB, indicating σB-dependent transcription of these genes. RNA-Seq analyses indicate that a total of 67 noncoding RNA molecules (ncRNAs) are transcribed in stationary phase L. monocytogenes, including 7 previously unrecognized putative ncRNAs. Application of a dynamically trained Hidden Markov Model, in combination with RNA-Seq data, identified 65 putative σB promoters upstream of 82 of the 96 σB-dependent genes and upstream of the one σB-dependent ncRNA. The RNA-Seq data also enabled annotation of putative operons as well as visualization of 5'- and 3'-UTR regions. Conclusions The results from these studies provide powerful evidence that RNA-Seq data combined with appropriate bioinformatics tools allow quantitative characterization of prokaryotic transcriptomes, thus providing exciting new strategies for exploring transcriptional regulatory networks in bacteria. See minireivew http://jbiol.com/content/8/12/107.
Collapse
Affiliation(s)
- Haley F Oliver
- Department of Food Science, Cornell University, Ithaca, NY, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Some Listeria monocytogenes outbreak strains demonstrate significantly reduced invasion, inlA transcript levels, and swarming motility in vitro. Appl Environ Microbiol 2009; 75:5647-58. [PMID: 19581477 DOI: 10.1128/aem.00367-09] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Listeria monocytogenes can cause a severe invasive food-borne disease known as listeriosis, and large outbreaks of this disease occur occasionally. Based on molecular-subtype data, epidemic clone (EC) strains have been defined, including ECI and ECIa, which have caused listeriosis outbreaks on different continents. While a number of molecular-subtyping studies of outbreak strains have been reported, few comprehensive data sets of virulence-associated characteristics of these strains are available. We assembled a set of human clinical isolates from 15 outbreaks that occurred worldwide between 1975 and 2002. Initial characterization of these strains showed significant variation in the ability to invade human Caco-2 intestinal epithelial cells and HepG2 hepatic cells; four strains showed consistently reduced invasion in both cell lines. DNA sequencing of inlA, which encodes a protein required for efficient Caco-2 and HepG2 invasion, showed that none of the invasion-attenuated strains contained known virulence-attenuating mutations in inlA. Phylogenetic analyses of inlA sequences revealed a well-supported clade containing a fully invasive ECI strain and three invasion-attenuated ECI strains, along with a fully invasive ECIa strain and an invasion-attenuated ECIa strain. Of the four invasion-attenuated strains, one strain showed both reduced inlA transcript levels and impaired swarming, one strain showed reduced inlA transcript levels, and two strains showed reduced swarming. Overall, our data show that (i) L. monocytogenes strains from outbreaks vary significantly in invasion efficiency and (ii) different mechanisms may contribute to reduced invasion efficiency. Association between EC strains and listeriosis outbreaks may involve characteristics other than virulence phenotypes, including survival and growth in food-associated environments.
Collapse
|
45
|
Abstract
The bacterium Listeria monocytogenes is the causative agent of listeriosis, a highly fatal opportunistic foodborne infection. Listeria spp. are isolated from a diversity of environmental sources, including soil, water, effluents, a large variety of foods, and the feces of humans and animals. Recent outbreaks demonstrated that L. monocytogenes can cause gastroenteritis in otherwise healthy individuals and more severe invasive disease in immunocompromised patients. Common symptoms include fever, watery diarrhea, nausea, headache, and pains in joints and muscles. The intestinal tract is the major portal of entry for L. monocytogenes, whereby strains penetrate the mucosal tissue either directly, via invasion of enterocytes, or indirectly, via active penetration of the Peyer's patches. Studies have revealed the strategy taken by the bacteria to overcome changes in oxygen tension, osmolarity, acidity, and the sterilizing effects of bile or antimicrobial peptides to adapt to conditions in the gut. In addition, L. monocytogenes has evolved species-specific strategies for intestinal entry by exploiting the interaction between the internalin protein and its receptor E-cadherin, or inducing diarrhea and an inflammatory response via the activity of its hemolytic toxin, listeriolysin. The ability of these bacteria to survive in bile-rich environments, and to induce depletion of sentinel cells such as Paneth cells that monitor the luminal burden of commensal bacteria, suggest strategies that have evolved to promote intestinal survival. Preexisting gastrointestinal disease may be a risk factor for infection of the gastrointestinal tract with L. monocytogenes. Currently, there is enough evidence to warrant consideration of L. monocytogenes as a possible etiology in outbreaks of febrile gastroenteritis, and for further studies to examine the genetic structure of Listeria strains that have a propensity to cause gastrointestinal versus systemic infections.
Collapse
|
46
|
Orsi RH, Sun Q, Wiedmann M. Genome-wide analyses reveal lineage specific contributions of positive selection and recombination to the evolution of Listeria monocytogenes. BMC Evol Biol 2008; 8:233. [PMID: 18700032 PMCID: PMC2532693 DOI: 10.1186/1471-2148-8-233] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2007] [Accepted: 08/12/2008] [Indexed: 12/30/2022] Open
Abstract
Background The genus Listeria includes two closely related pathogenic and non-pathogenic species, L. monocytogenes and L. innocua. L. monocytogenes is an opportunistic human foodborne and animal pathogen that includes two common lineages. While lineage I is more commonly found among human listeriosis cases, lineage II appears to be overrepresented among isolates from foods and environmental sources. This study used the genome sequences for one L. innocua strain and four L. monocytogenes strains representing lineages I and II, to characterize the contributions of positive selection and recombination to the evolution of the L. innocua/L. monocytogenes core genome. Results Among the 2267 genes in the L. monocytogenes/L. innocua core genome, 1097 genes showed evidence for recombination and 36 genes showed evidence for positive selection. Positive selection was strongly associated with recombination. Specifically, 29 of the 36 genes under positive selection also showed evidence for recombination. Recombination was more common among isolates in lineage II than lineage I; this trend was confirmed by sequencing five genes in a larger isolate set. Positive selection was more abundant in the ancestral branch of lineage II (20 genes) as compared to the ancestral branch of lineage I (9 genes). Additional genes under positive selection were identified in the branch separating the two species; for this branch, genes in the role category "Cell wall and membrane biogenesis" were significantly more likely to have evidence for positive selection. Positive selection of three genes was confirmed in a larger isolate set, which also revealed occurrence of multiple premature stop codons in one positively selected gene involved in flagellar motility (flaR). Conclusion While recombination and positive selection both contribute to evolution of L. monocytogenes, the relative contributions of these evolutionary forces seem to differ by L. monocytogenes lineages and appear to be more important in the evolution of lineage II, which seems to be found in a broader range of environments, as compared to the apparently more host adapted lineage I. Diversification of cell wall and membrane biogenesis and motility-related genes may play a particularly important role in the evolution of L. monocytogenes.
Collapse
Affiliation(s)
- Renato H Orsi
- Department of Food Science, Cornell University, Ithaca, NY, USA.
| | | | | |
Collapse
|
47
|
Gueriri I, Cyncynatus C, Dubrac S, Arana AT, Dussurget O, Msadek T. The DegU orphan response regulator of Listeria monocytogenes autorepresses its own synthesis and is required for bacterial motility, virulence and biofilm formation. Microbiology (Reading) 2008; 154:2251-2264. [DOI: 10.1099/mic.0.2008/017590-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Ibtissem Gueriri
- Unité de Biologie des Bactéries Pathogènes à Gram Positif, CNRS URA 2172, Institut Pasteur, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Camille Cyncynatus
- Unité de Biologie des Bactéries Pathogènes à Gram Positif, CNRS URA 2172, Institut Pasteur, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Sarah Dubrac
- Unité de Biologie des Bactéries Pathogènes à Gram Positif, CNRS URA 2172, Institut Pasteur, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Alejandro Toledo Arana
- Laboratory of Bacterial Biofilms, Instituto De Agrobiotecnologia Y Recursos Naturales, Universidad Pública de Navarra, 31006 Pamplona, Spain
| | - Olivier Dussurget
- Unité des Interactions Bactéries Cellules, INSERM U604, INRA USC2020, Institut Pasteur, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Tarek Msadek
- Unité de Biologie des Bactéries Pathogènes à Gram Positif, CNRS URA 2172, Institut Pasteur, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| |
Collapse
|
48
|
Bresolin G, Trček J, Scherer S, Fuchs TM. Presence of a functional flagellar cluster Flag-2 and low-temperature expression of flagellar genes in Yersinia enterocolitica W22703. MICROBIOLOGY-SGM 2008; 154:196-206. [PMID: 18174138 DOI: 10.1099/mic.0.2007/008458-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Twelve Yersinia enterocolitica mutants carrying luxCDABE-transposon insertions in motility and chemotaxis genes were isolated on the basis of strong low-temperature induction. Two transposons were located within an 11.2 kb enteric flagellar cluster 2 (Flag-2) of Y. enterocolitica biotype 2, serotype O : 9 strain W22703. The Flag-2 gene cluster is absent from the corresponding genomic location of the sequenced strain Y. enterocolitica biotype 1B, serotype O : 8 strain 8081. Evidence for the functionality of the O : 9 Flag-2 genes, probably located within the plasticity zone of the genome, is provided by swarming assays. PCR analysis of 49 strains revealed the presence of Flag-2 genes in biotypes 2-5, but not in biotypes 1A or 1B. Bioluminescence, measured between 6 and 37 degrees C, showed that the expression of all genes located in Flag-2 and in the known flagellar cluster, Flag-1, was highest at approximately 20 degrees C, and that expression of two Flag-2 genes is FlhC-dependent. In a motility assay, a non-motile and a hyper-motile phenotype resulted from knockout mutations of the Flag-1 genes fliS1 and fliT, respectively. Complemented strains validated these results, confirming the regulatory role of FliT.
Collapse
Affiliation(s)
- Geraldine Bresolin
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Abteilung Mikrobiologie, Technische Universität München, Weihenstephaner Berg 3, 85354 Freising, Germany
| | - Janja Trček
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Abteilung Mikrobiologie, Technische Universität München, Weihenstephaner Berg 3, 85354 Freising, Germany
| | - Siegfried Scherer
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Abteilung Mikrobiologie, Technische Universität München, Weihenstephaner Berg 3, 85354 Freising, Germany
| | - Thilo M Fuchs
- Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Abteilung Mikrobiologie, Technische Universität München, Weihenstephaner Berg 3, 85354 Freising, Germany
| |
Collapse
|
49
|
Dussurget O. Chapter 1 New Insights into Determinants of Listeria Monocytogenes Virulence. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2008; 270:1-38. [DOI: 10.1016/s1937-6448(08)01401-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
50
|
Severino P, Dussurget O, Vêncio RZN, Dumas E, Garrido P, Padilla G, Piveteau P, Lemaître JP, Kunst F, Glaser P, Buchrieser C. Comparative transcriptome analysis of Listeria monocytogenes strains of the two major lineages reveals differences in virulence, cell wall, and stress response. Appl Environ Microbiol 2007; 73:6078-88. [PMID: 17704270 PMCID: PMC2075013 DOI: 10.1128/aem.02730-06] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2006] [Accepted: 08/06/2007] [Indexed: 01/02/2023] Open
Abstract
Listeria monocytogenes is a food-borne, opportunistic, bacterial pathogen causing a wide spectrum of diseases, including meningitis, septicemia, abortion, and gastroenteritis, in humans and animals. Among the 13 L. monocytogenes serovars described, human listeriosis is mostly associated with strains of serovars 4b, 1/2b, and 1/2a. Within the species L. monocytogenes, three phylogenetic lineages are described. Serovar 1/2a belongs to phylogenetic lineage I, while serovars 4b and 1/2b group in phylogenetic lineage II. To explore the role of gene expression in the adaptation of L. monocytogenes strains of these two major lineages to different environments, as well as in virulence, we performed whole-genome expression profiling of six L. monocytogenes isolates of serovars 4b, 1/2b, and 1/2a of distinct origins, using a newly constructed Listeria multigenome DNA array. Comparison of the global gene expression profiles revealed differences among strains. The expression profiles of two strains having distinct 50% lethal doses, as assessed in the mouse model, were further analyzed. Gene ontology term enrichment analysis of the differentially expressed genes identified differences in protein-, nucleic acid-, carbon metabolism-, and virulence-related gene expression. Comparison of the expression profiles of the core genomes of all strains revealed differences between the two lineages with respect to cell wall synthesis, the stress-related sigma B regulon and virulence-related genes. These findings suggest different patterns of interaction with host cells and the environment, key factors for host colonization and survival in the environment.
Collapse
Affiliation(s)
- Patricia Severino
- Unité de Génomique des Microorganismes Pathogènes, Institut Pasteur, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|