1
|
Yu J, Tang H, Chen Y, Wang Z, Huang W, Zhou T, Wen B, Wang C, Gu S, Ni J, Tao J, Wang D, Lu J, Xie Q, Yao YF. Salmonella utilizes L-arabinose to silence virulence gene expression for accelerated pathogen growth within the host. Gut Microbes 2025; 17:2467187. [PMID: 39954030 PMCID: PMC11834461 DOI: 10.1080/19490976.2025.2467187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/11/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
Carbon source is an important nutrient for bacteria to sustain growth and often acts as a signal that modulates virulence expression. L-arabinose is produced by plants and plays an important role in regulating the global gene expression of bacteria. Previously, we have shown that L-arabinose induces a more severe systemic infection in Salmonella-infected mice with normal microbiota, but does not affect the disease progression in mice with microbiota depleted by antibiotic treatment. The underlying mechanism remains elusive. In this study, we demonstrate that L-arabinose represses the expression of Salmonella type III secretion system 1 (T3SS-1) genes by negatively regulating the activity of the cyclic 3' 5'-AMP (cAMP)-cAMP receptor protein (CRP) complex. The cAMP-CRP complex can activate ribosome-associated inhibitor A, encoded by yfiA, to maintain the stability of HilD, a key transcriptional regulator of T3SS-1. L-arabinose supplementation promotes Salmonella initial bloom in the antibiotic-pretreated mouse gut and ultimately compensates for reduced virulence within the host. These results decipher the molecular mechanism by which cAMP-CRP directs regulatory changes of virulence in response to L-arabinose in Salmonella. It further implies that Salmonella exploits L-arabinose both as a nutrient and a regulatory signal to maintain a balance between growth and virulence within the host.
Collapse
Affiliation(s)
- Jingchen Yu
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huang Tang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yana Chen
- Department of Pediatrics, Anhui Provincial Hospital, The First Affiliated Hospital of USTC, Hefei, Anhui, China
| | - Zuoqiang Wang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wanqiu Huang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Zhou
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingjie Wen
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengyue Wang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuang Gu
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinjing Ni
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Tao
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Danni Wang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Lu
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Xie
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Feng Yao
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Diagnosis and Treatment of Respiratory Infectious Diseases (20dz2261100), Shanghai Key Laboratory of Emergency Prevention, Shanghai, China
| |
Collapse
|
2
|
Cooper KG, Kari L, Chong A, Tandon N, Doran K, Gomes Da Silva L, Cockrell DC, Baylink A, Steele-Mortimer O. HilD-regulated chemotaxis proteins contribute to Salmonella Typhimurium colonization in the gut. mBio 2025; 16:e0039025. [PMID: 39998229 PMCID: PMC11980550 DOI: 10.1128/mbio.00390-25] [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: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
In the enteric pathogen Salmonella Typhimurium, invasion and motility are coordinated by HilD, a master regulator that activates expression of genes encoding the type III secretion system 1 and some motility genes, including the chemotaxis gene mcpC. Previously, we have shown that McpC induces smooth swimming, which is important for type III secretion system 1-dependent invasion of epithelial cells. Here, we have studied another Salmonella-specific chemotaxis gene, mcpA, and demonstrate that it is also HilD regulated. Whereas HilD induction of mcpC occurs by direct derepression of H-NS, mcpA induction requires neither H-NS derepression nor the flagellar-specific sigma factor fliA; instead it occurs through a HilD-SprB regulatory cascade, providing experimental confirmation of previous transcriptional regulatory mapping. McpA and McpC contain methyl-accepting domains characteristic of bacterial chemoreceptors, and McpA also contains a chemoreceptor zinc-binding (CZB) protein domain found in a variety of bacterial proteins, many of which are involved in signaling or regulatory roles. Here, we show that, in a mouse model for acute Salmonella colitis, both mcpA and mcpC deletion mutants are outcompeted by wild-type Salmonella Typhimurium in the gut lumen. CZB domains bind Zn2+ through a conserved cysteine residue and are thought to perform redox-sensing through redox-initiated alterations in zinc homeostasis. We found that the conserved cysteine is required for McpA function in the mouse gut, thus demonstrating a virulence role for the CZB Zn2+-binding site during infection. IMPORTANCE The gut-adapted bacterium Salmonella Typhimurium causes inflammatory diarrhea via a process that involves active invasion of intestinal epithelial cells, secretion of inflammatory molecules, and recruitment of immune cells. Although bacterial motility and invasion of host cells are coordinated, how directed movement facilitates luminal survival and growth or invasion at the mucosal surface is not understood. Chemotaxis is the process by which bacteria control movement toward attractants and away from repellents. Previously, we identified a Salmonella-specific chemoreceptor, McpC, that is co-expressed with the invasion machinery and promotes smooth swimming for optimal host cell invasion. Here, we investigated another chemoreceptor, McpA, also regulated with invasion-associated genes and show it contributes to luminal expansion rather than invasion of epithelial cells. McpA activity requires a conserved Zn2+-binding domain, thought to be involved in sensing inflammation. This work demonstrates that coordination of invasion and chemotaxis plays a significant role in the gut.
Collapse
Affiliation(s)
- Kendal G. Cooper
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Laszlo Kari
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Audrey Chong
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Naman Tandon
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Kathleen Doran
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Lidiane Gomes Da Silva
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Diane C. Cockrell
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Arden Baylink
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Olivia Steele-Mortimer
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| |
Collapse
|
3
|
López-Pagán N, Rufián JS, Luneau J, Sánchez-Romero MA, Aussel L, van Vliet S, Ruiz-Albert J, Beuzón CR. Pseudomonas syringae subpopulations cooperate by coordinating flagellar and type III secretion spatiotemporal dynamics to facilitate plant infection. Nat Microbiol 2025; 10:958-972. [PMID: 40175722 PMCID: PMC11964935 DOI: 10.1038/s41564-025-01966-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 02/19/2025] [Indexed: 04/04/2025]
Abstract
Isogenic bacterial populations can display probabilistic cell-to-cell variation in response to challenges. This phenotypic heterogeneity can affect virulence in animals, but its impact on plant pathogens is unknown. Previously, we showed that expression of the type III secretion system (T3SS) of the plant pathogen Pseudomonas syringae displays phenotypic variation in planta. Here we use flow cytometry and microscopy to investigate single-cell flagellar expression in relation to T3SS expression, showing that both systems undergo phenotypic heterogeneity in vitro in apoplast-mimicking medium and within apoplastic microcolonies throughout colonization of Phaseolus vulgaris. Stochastic, spatial and time factors shape the dynamics of a phenotypically diverse pathogen population that displays division of labour during colonization: effectors produced by T3SS-expressing bacteria act as 'common goods' to suppress immunity, allowing motile flagella-expressing bacteria to increase and leave infected tissue before necrosis. These results showcase the mechanisms of bacterial specialization during plant colonization in an environmentally and agriculturally relevant system.
Collapse
Affiliation(s)
- Nieves López-Pagán
- Instituto de Hortofruticultura Subtropical y Mediterránea "La Mayora", Universidad de Málaga-Consejo Superior de Investigaciones Científicas (IHSM-UMA-CSIC), Málaga, Spain
- Department of Chemical Biology and Genetics, Centre of the Region Haná for Biotechnological and Agricultural Research, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - José S Rufián
- Instituto de Hortofruticultura Subtropical y Mediterránea "La Mayora", Universidad de Málaga-Consejo Superior de Investigaciones Científicas (IHSM-UMA-CSIC), Málaga, Spain
| | - Julien Luneau
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | | | - Laurent Aussel
- Aix Marseille Univ, CNRS, LCB UMR7283, IMM, Marseille, France
| | - Simon van Vliet
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Javier Ruiz-Albert
- Instituto de Hortofruticultura Subtropical y Mediterránea "La Mayora", Universidad de Málaga-Consejo Superior de Investigaciones Científicas (IHSM-UMA-CSIC), Málaga, Spain
| | - Carmen R Beuzón
- Instituto de Hortofruticultura Subtropical y Mediterránea "La Mayora", Universidad de Málaga-Consejo Superior de Investigaciones Científicas (IHSM-UMA-CSIC), Málaga, Spain.
| |
Collapse
|
4
|
Kroon S, Malcic D, Weidert L, Bircher L, Boldt L, Christen P, Kiefer P, Sintsova A, Nguyen BD, Barthel M, Steiger Y, Clerc M, Herzog MKM, Chen C, Gül E, Guery B, Slack E, Sunagawa S, Vorholt JA, Maier L, Lacroix C, Hausmann A, Hardt WD. Sublethal systemic LPS in mice enables gut-luminal pathogens to bloom through oxygen species-mediated microbiota inhibition. Nat Commun 2025; 16:2760. [PMID: 40113753 PMCID: PMC11926250 DOI: 10.1038/s41467-025-57979-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
Endotoxin-driven systemic immune activation is a common hallmark across various clinical conditions. During acute critical illness, elevated plasma lipopolysaccharide triggers non-specific systemic immune activation. In addition, a compositional shift in the gut microbiota, including an increase in gut-luminal opportunistic pathogens, is observed. Whether a causal link exists between acute endotoxemia and abundance of gut-luminal opportunistic pathogens is incompletely understood. Here, we model acute, pathophysiological lipopolysaccharide concentrations in mice and show that systemic exposure promotes a 100-10'000-fold expansion of Klebsiella pneumoniae, Escherichia coli, Enterococcus faecium and Salmonella Typhimurium in the gut within one day, without overt enteropathy. Mechanistically, this is driven by a Toll-like receptor 4-dependent increase in gut-luminal oxygen species levels, which transiently halts microbiota fermentation and fuels growth of gut-luminal facultative anaerobic pathogens through oxidative respiration. Thus, systemic immune activation transiently perturbs microbiota homeostasis and favours opportunistic pathogens, potentially increasing the risk of infection in critically ill patients.
Collapse
Affiliation(s)
- Sanne Kroon
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Dejan Malcic
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Lena Weidert
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Lea Bircher
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Leonardo Boldt
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany
- M3 Research Center for Malignome, Metabolome and Microbiome, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen, Tübingen, Germany
| | - Philipp Christen
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Patrick Kiefer
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Anna Sintsova
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Bidong D Nguyen
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Manja Barthel
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Yves Steiger
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Melanie Clerc
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Mathias K-M Herzog
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Carmen Chen
- Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ersin Gül
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Benoit Guery
- Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Emma Slack
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Shinichi Sunagawa
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Julia A Vorholt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Lisa Maier
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany
- M3 Research Center for Malignome, Metabolome and Microbiome, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen, Tübingen, Germany
| | - Christophe Lacroix
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Annika Hausmann
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
- reNEW - Novo Nordisk Foundation Center for Stem Cell Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
5
|
Hainzl T, Scortti M, Lindgren C, Grundström C, Krypotou E, Vázquez-Boland JA, Sauer-Eriksson AE. Structural basis of promiscuous inhibition of Listeria virulence activator PrfA by oligopeptides. Cell Rep 2025; 44:115290. [PMID: 39970044 DOI: 10.1016/j.celrep.2025.115290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/20/2024] [Accepted: 01/17/2025] [Indexed: 02/21/2025] Open
Abstract
The facultative pathogen Listeria monocytogenes uses a master regulator, PrfA, to tightly control the fitness-costly expression of its virulence factors. We found that PrfA activity is repressed via competitive occupancy of the binding site for the PrfA-activating cofactor, glutathione, by exogenous nutritional oligopeptides. The inhibitory peptides show different sequence and physicochemical properties, but how such a wide variety of oligopeptides can bind PrfA was unclear. Using crystal structure analysis of PrfA complexed with inhibitory tri- and tetrapeptides, we show here that the binding promiscuity is due to the ability of PrfA β5 in the glutathione-binding inter-domain tunnel to establish parallel or antiparallel β sheet-like interactions with the peptide backbone. Spacious tunnel pockets provide additional flexibility for unspecific peptide accommodation while providing selectivity for hydrophobic residues. Hydrophobic contributions from two adjacent peptide residues appear to be critical for efficient PrfA inhibitory binding. In contrast to glutathione, peptide binding prevents the conformational change required for the correct positioning of the DNA-binding helix-turn-helix motifs of PrfA, effectively inhibiting virulence gene expression.
Collapse
Affiliation(s)
- Tobias Hainzl
- Department of Chemistry and Umeå Centre for Microbial Research, Umeå University, 901 87 Umeå, Sweden
| | - Mariela Scortti
- Microbial Pathogenomics Group, Edinburgh Medical School (Biomedical Sciences), Edinburgh BioQuarter, IRR Bldg. South, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Cecilia Lindgren
- Department of Chemistry and Umeå Centre for Microbial Research, Umeå University, 901 87 Umeå, Sweden
| | - Christin Grundström
- Department of Chemistry and Umeå Centre for Microbial Research, Umeå University, 901 87 Umeå, Sweden
| | - Emilia Krypotou
- Microbial Pathogenomics Group, Edinburgh Medical School (Biomedical Sciences), Edinburgh BioQuarter, IRR Bldg. South, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - José A Vázquez-Boland
- Microbial Pathogenomics Group, Edinburgh Medical School (Biomedical Sciences), Edinburgh BioQuarter, IRR Bldg. South, University of Edinburgh, Edinburgh EH16 4UU, UK.
| | | |
Collapse
|
6
|
Santamaria de Souza N, Cherrak Y, Andersen TB, Vetsch M, Barthel M, Kroon S, Bakkeren E, Schubert C, Christen P, Kiefer P, Vorholt JA, Nguyen BD, Hardt WD. Context-dependent change in the fitness effect of (in)organic phosphate antiporter glpT during Salmonella Typhimurium infection. Nat Commun 2025; 16:1912. [PMID: 39994176 PMCID: PMC11850910 DOI: 10.1038/s41467-025-56851-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Salmonella enterica is a frequent cause of foodborne diseases, which is attributed to its adaptability. Even within a single host, expressing a gene can be beneficial in certain infection stages but neutral or even detrimental in others as previously shown for flagellins. Mutants deficient for the conserved glycerol-3-phosphate and phosphate antiporter glpT have been shown to be positively selected in nature, clinical, and laboratory settings. This suggests that different selective pressures select for the presence or absence of GlpT in a context dependent fashion, a phenomenon known as antagonistic pleiotropy. Using mutant libraries and reporters, we investigated the fitness of glpT-deficient mutants during murine orogastric infection. While glpT-deficient mutants thrive during initial growth in the gut lumen, where GlpT's capacity to import phosphate is disadvantageous, they are counter-selected by macrophages. The dichotomy showcases the need to study the spatial and temporal heterogeneity of enteric pathogens' fitness across distinct lifestyles and niches. Insights into the differential adaptation during infection may reveal opportunities for therapeutic interventions.
Collapse
Affiliation(s)
| | - Yassine Cherrak
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Thea Bill Andersen
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Michel Vetsch
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Manja Barthel
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Sanne Kroon
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Erik Bakkeren
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Christopher Schubert
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Philipp Christen
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Patrick Kiefer
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Julia A Vorholt
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Bidong D Nguyen
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Wolf-Dietrich Hardt
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
7
|
Walter S, Schatz V, Petzold J, Schmidt C, Hoffmann S, Jantsch J, Gerlach RG. O 2-dependent incapacitation of the Salmonella pathogenicity island 1 repressor HilE. Front Cell Infect Microbiol 2025; 15:1434254. [PMID: 40041146 PMCID: PMC11876186 DOI: 10.3389/fcimb.2025.1434254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
For successful colonization, pathogenic bacteria need to adapt their metabolism and virulence functions to challenging environments within their mammalian hosts that are frequently characterized by low oxygen (O2) tensions. Upon oral ingestion, the human pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium) is exposed to changing O2 and pH levels. Low concentrations of O2, which can enhance the virulence of enteroinvasive pathogens, facilitate the expression of the type three secretion system (T3SS-1) encoded by the Salmonella pathogenicity island 1 (SPI-1) that is critical for enteroinvasion and pathogenicity of S. Typhimurium. To study the impact of key environmental cues of the intestine when Salmonella encounter enterocytes, we established an in vitro growth model, which allows shifting the concentration of O2 from 0.5% to 11% and the pH from 5.9 to 7.4 in the presence of acetate and the alternative electron acceptor nitrate. Compared to normoxia, hypoxia elevated the expression of SPI-1 genes encoding T3SS-1 translocators and effectors, which resulted in higher invasion and effector translocation in epithelial cells. While hypoxia and pH shift only marginally altered the gene expression of SPI-1 regulators, including the SPI-1 repressor hilE, hypoxia and pH shift completely incapacitated HilE in a post-translational manner, ultimately promoting SPI-1 activity. From these findings, we conclude that O2-dependent HilE function allows for ultrasensitive adaptation of SPI-1 activity in environments with varying O2 availability such as the intestinal tract.
Collapse
Affiliation(s)
- Steffi Walter
- Project Group 5, Robert Koch Institute, Wernigerode, Germany
| | - Valentin Schatz
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, Regensburg, Germany
- Institute for Medical Microbiology, Immunology, and Hygiene, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Jana Petzold
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | | | | | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, Regensburg, Germany
- Institute for Medical Microbiology, Immunology, and Hygiene, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Roman G. Gerlach
- Project Group 5, Robert Koch Institute, Wernigerode, Germany
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
8
|
Bin Masroni MS, Koay ESC, Lee VKM, Ng SB, Tan SY, Tan KM, Archetti M, Leong SM. Sociobiology meets oncology: unraveling altruistic cooperation in cancer cells and its implications. Exp Mol Med 2025; 57:30-40. [PMID: 39774289 PMCID: PMC11799181 DOI: 10.1038/s12276-024-01387-9] [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: 08/08/2024] [Revised: 10/03/2024] [Accepted: 10/29/2024] [Indexed: 01/11/2025] Open
Abstract
Altruism, an act of benefiting others at a cost to the self, challenges our understanding of evolution. This Perspective delves into the importance of altruism in cancer cells and its implications for therapy. Against the backdrop of existing knowledge on various social organisms found in nature, we explore the mechanisms underlying the manifestation of altruism within breast tumors, revealing a complex interplay of seemingly counteracting cancer signaling pathways and processes that orchestrate the delicate balance between cost and benefit underlying altruistic cooperation. We also discuss how evolutionary game theory, coupled with contemporary molecular tools, may shed light on understudied mechanisms governing the dynamics of altruistic cooperation in cancer cells. Finally, we discuss how molecular insights gleaned from these mechanistic dissections may fuel advancements in our comprehension of altruism among cancer cells, with implications across multiple disciplines, offering innovative prospects for therapeutic strategies, molecular discoveries, and evolutionary investigations.
Collapse
Affiliation(s)
- Muhammad Sufyan Bin Masroni
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Evelyn Siew-Chuan Koay
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Victor Kwan Min Lee
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, Singapore
- Department of Pathology, National University Hospital, Singapore, Singapore
| | - Siok Bian Ng
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, Singapore
- Department of Pathology, National University Hospital, Singapore, Singapore
| | - Soo Yong Tan
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, Singapore
- Department of Pathology, National University Hospital, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Karen Meiling Tan
- Singapore Institute for Clinical Sciences, Brenner Centre for Molecular Medicine, Singapore, Singapore
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore
| | - Marco Archetti
- Department of Biology, Pennsylvania State University, University Park, PA, USA.
| | - Sai Mun Leong
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, Singapore.
| |
Collapse
|
9
|
Han R, Qian Y, Zheng C. A novel small RNA regulates Locus of Enterocyte Effacement and site-specific colonization of enterohemorrhagic Escherichia coli O157:H7 in gut. Front Cell Infect Microbiol 2025; 14:1517328. [PMID: 39882344 PMCID: PMC11774850 DOI: 10.3389/fcimb.2024.1517328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 12/11/2024] [Indexed: 01/31/2025] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) is a contagious foodborne pathogen that specifically colonizes the human large intestine, which is regulated by different environmental stimuli within the gut. Transcriptional regulation of EHEC virulence and infection has been extensively studied, while the posttranscriptional regulation of these processes by small RNAs (sRNAs) remains not fully understood. Here we present a virulence-regulating pathway in EHEC O157:H7, in which the sRNA EvrS binds to and destabilizes the mRNA of Z2269, a novel transcriptional regulator. In turn, Z2269 indirectly activates the expression of LEE (locus of enterocyte effacement) pathogenicity island through the master regulator Ler. Importantly, the expression of EvrS is modulated by environmental oxygen levels. EvrS also exhibits lower expression in the colon compared to the ileum, influencing the site-specific colonization of EHEC O157:H7 in mice. These results indicate that the oxygen status within the intestine may regulate the expression of EvrS, thereby modulating virulence factors of EHEC and contributing to successful infection in vivo. This study has broader implications for understanding sRNA functions in spatiotemporal virulence control of EHEC and may provide novel strategies to prevent EHEC infections.
Collapse
Affiliation(s)
- Runhua Han
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, China
- Department of Chemistry, University of Manitoba, Winnipeg, MB, Canada
| | - Ye Qian
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, China
| | - Chenguang Zheng
- College of Public Health, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
10
|
Schubert K, Braly M, Zhang J, Muscolo ME, Lam HN, Hug K, Moore H, McCausland JW, Terciano D, Lowe T, Lesser CF, Jacobs-Wagner C, Wang H, Auerbuch V. The polyadenylase PAPI is required for virulence plasmid maintenance in pathogenic bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617751. [PMID: 39416138 PMCID: PMC11482874 DOI: 10.1101/2024.10.11.617751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Many species of pathogenic bacteria harbor critical plasmid-encoded virulence factors, and yet the regulation of plasmid replication is often poorly understood despite playing a critical role in plasmid-encoded gene expression. Human pathogenic Yersinia, including the plague agent Y. pestis and its close relative Y. pseudotuberculosis, require the type III secretion system (T3SS) virulence factor to subvert host defense mechanisms and colonize host tissues. The Yersinia T3SS is encoded on the IncFII plasmid for Y ersinia virulence (pYV). Several layers of gene regulation enables a large increase in expression of Yersinia T3SS genes at mammalian body temperature. Surprisingly, T3SS expression is also controlled at the level of gene dosage. The number of pYV molecules relative to the number of chromosomes per cell, referred to as plasmid copy number, increases with temperature. The ability to increase and maintain elevated pYV plasmid copy number, and therefore T3SS gene dosage, at 37°C is important for Yersinia virulence. In addition, pYV is highly stable in Yersinia at all temperatures, despite being dispensable for growth outside the host. Yet how Yersinia reinforces elevated plasmid replication and plasmid stability remains unclear. In this study, we show that the chromosomal gene pcnB encoding the polyadenylase PAP I is required for regulation of pYV plasmid copy number (PCN), maintenance of pYV in the bacterial population outside the host, robust T3SS activity, and Yersinia virulence in a mouse infection model. Likewise, pcnB/PAP I is also required for robust expression of the Shigella flexneri virulence plasmid-encoded T3SS. Furthermore, Yersinia and Shigella pcnB/PAP I is required for maintaining normal PCN of model antimicrobial resistance (AMR) plasmids whose replication is regulated by sRNA, thereby increasing antibiotic resistance by ten-fold. These data suggest that pcnB/PAP I contributes to the spread and stabilization of virulence and AMR plasmids in bacterial pathogens, and is essential in maintaining the gene dosage required to mediate plasmid-encoded traits. Importantly pcnB/PAP I has been bioinformatically identified in many species of bacteria despite being studied in only a few species to date. Our work highlights the potential importance of pcnB/PAP I in antibiotic resistance, and shows for the first time that pcnB/PAP I reinforces PCN and virulence plasmid stability in natural pathogenic hosts with a direct impact on bacterial virulence.
Collapse
Affiliation(s)
- Katherine Schubert
- Department of Molecular, Cell, and Developmental Biology, UC Santa Cruz, Santa Cruz, CA 95064, United States
| | - Micah Braly
- Department of Microbiology and Environmental Toxicology, UC Santa Cruz, Santa Cruz, CA 95064, United States
| | - Jessica Zhang
- Department of Biology, Stanford University, Stanford, CA 94305, United States
| | - Michele E Muscolo
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, MA 02115, United States
| | - Hanh N Lam
- Department of Microbiology and Environmental Toxicology, UC Santa Cruz, Santa Cruz, CA 95064, United States
| | - Karen Hug
- Department of Microbiology and Environmental Toxicology, UC Santa Cruz, Santa Cruz, CA 95064, United States
| | - Henry Moore
- Department of Biomolecular Engineering, UC Santa Cruz, Santa Cruz, CA 95064, United States
| | - Joshua W McCausland
- Department of Biology, Stanford University, Stanford, CA 94305, United States
- Sarafan ChEM-H Institute, Stanford University, Stanford, CA 94305, United States
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Derfel Terciano
- Department of Microbiology and Environmental Toxicology, UC Santa Cruz, Santa Cruz, CA 95064, United States
| | - Todd Lowe
- Department of Biomolecular Engineering, UC Santa Cruz, Santa Cruz, CA 95064, United States
| | - Cammie F Lesser
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, MA 02115, United States
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, United States
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, United States
| | - Christine Jacobs-Wagner
- Department of Biology, Stanford University, Stanford, CA 94305, United States
- Sarafan ChEM-H Institute, Stanford University, Stanford, CA 94305, United States
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Helen Wang
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden
| | - Victoria Auerbuch
- Department of Microbiology and Environmental Toxicology, UC Santa Cruz, Santa Cruz, CA 95064, United States
| |
Collapse
|
11
|
Nguyen BD, Sintsova A, Schubert C, Sichert A, Scheidegger C, Näf J, Huttman J, Lentsch V, Keys T, Rutschmann C, Christen P, Kiefer P, Keller P, Barthel M, Cuenca M, Christen B, Sauer U, Slack E, Vorholt JA, Sunagawa S, Hardt WD. Salmonella Typhimurium screen identifies shifts in mixed-acid fermentation during gut colonization. Cell Host Microbe 2024; 32:1758-1773.e4. [PMID: 39293436 DOI: 10.1016/j.chom.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 07/10/2024] [Accepted: 08/21/2024] [Indexed: 09/20/2024]
Abstract
How enteric pathogens adapt their metabolism to a dynamic gut environment is not yet fully understood. To investigate how Salmonella enterica Typhimurium (S.Tm) colonizes the gut, we conducted an in vivo transposon mutagenesis screen in a gnotobiotic mouse model. Our data implicate mixed-acid fermentation in efficient gut-luminal growth and energy conservation throughout infection. During initial growth, the pathogen utilizes acetate fermentation and fumarate respiration. After the onset of gut inflammation, hexoses appear to become limiting, as indicated by carbohydrate analytics and the increased need for gluconeogenesis. In response, S.Tm adapts by ramping up ethanol fermentation for redox balancing and supplying the TCA cycle with α-ketoglutarate for additional energy. Our findings illustrate how S.Tm flexibly adapts mixed fermentation and its use of the TCA cycle to thrive in the changing gut environment. Similar metabolic wiring in other pathogenic Enterobacteriaceae may suggest a broadly conserved mechanism for gut colonization.
Collapse
Affiliation(s)
- Bidong D Nguyen
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| | - Anna Sintsova
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Christopher Schubert
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Andreas Sichert
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Clio Scheidegger
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Jana Näf
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland; Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Julien Huttman
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Verena Lentsch
- Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Tim Keys
- Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | | | - Philipp Christen
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Patrick Kiefer
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Philipp Keller
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Manja Barthel
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Miguelangel Cuenca
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Beat Christen
- Institute of Microbiology, University of Stuttgart, Stuttgart, Germany
| | - Uwe Sauer
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Emma Slack
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland; Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Julia A Vorholt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Shinichi Sunagawa
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
12
|
Gory R, Personnic N, Blaha D. Unravelling the Roles of Bacterial Nanomachines Bistability in Pathogens' Life Cycle. Microorganisms 2024; 12:1930. [PMID: 39338604 PMCID: PMC11434070 DOI: 10.3390/microorganisms12091930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Bacterial nanomachines represent remarkable feats of evolutionary engineering, showcasing intricate molecular mechanisms that enable bacteria to perform a diverse array of functions essential to persist, thrive, and evolve within ecological and pathological niches. Injectosomes and bacterial flagella represent two categories of bacterial nanomachines that have been particularly well studied both at the molecular and functional levels. Among the diverse functionalities of these nanomachines, bistability emerges as a fascinating phenomenon, underscoring their dynamic and complex regulation as well as their contribution to shaping the bacterial community behavior during the infection process. In this review, we examine two closely related bacterial nanomachines, the type 3 secretion system, and the flagellum, to explore how the bistability of molecular-scale devices shapes the bacterial eco-pathological life cycle.
Collapse
Affiliation(s)
- Romain Gory
- Group Persistence and Single-Cell Dynamics of Respiratory Pathogens, CIRI-Centre International de Recherche en Infectiologie, CNRS, INSERM, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, 50 avenue Tony Garnier, 69007 Lyon, France
| | - Nicolas Personnic
- Group Persistence and Single-Cell Dynamics of Respiratory Pathogens, CIRI-Centre International de Recherche en Infectiologie, CNRS, INSERM, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, 50 avenue Tony Garnier, 69007 Lyon, France
| | - Didier Blaha
- Group Persistence and Single-Cell Dynamics of Respiratory Pathogens, CIRI-Centre International de Recherche en Infectiologie, CNRS, INSERM, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, 50 avenue Tony Garnier, 69007 Lyon, France
| |
Collapse
|
13
|
Greene J, Snyder RA, Cotten KL, Huiszoon RC, Chu S, Braza RED, Chapin AA, Stine JM, Bentley WE, Ghodssi R, Davis KM. Yersinia pseudotuberculosis growth arrest during type-III secretion system expression is associated with altered ribosomal protein expression and decreased gentamicin susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.02.610769. [PMID: 39282321 PMCID: PMC11398311 DOI: 10.1101/2024.09.02.610769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
It has been long appreciated that expression of the Yersinia type-III secretion system (T3SS) in culture is associated with growth arrest. Here we sought to understand whether this impacts expression of ribosomal protein genes, which were among the most highly abundant transcripts in exponential phase Yersinia pseudotuberculosis based on RNA-seq analysis. To visualize changes in ribosomal protein expression, we generated a fluorescent transcriptional reporter with the promoter upstream of rpsJ/S10 fused to a destabilized gfp variant. We confirmed reporter expression significantly increases in exponential phase and decreases as cells transition to stationary phase. We then utilized a mouse model of systemic Y. pseudotuberculosis infection to compare T3SS and S10 reporter expression during clustered bacterial growth in the spleen, and found that cells expressing high levels of the T3SS had decreased S10 levels, while cells with lower T3SS expression retained higher S10 expression. In bacteriological media, growth inhibition with T3SS induction and a reduction in S10 expression were observed in subsets of cells, while cells with high expression of both T3SS and S10 were also observed. Loss of T3SS genes resulted in rescued growth and heightened S10 expression. To understand if clustered growth impacted bacterial gene expression, we utilized droplet-based microfluidics to encapsulate bacteria in spherical agarose droplets, and also observed growth inhibition with high expression of T3SS and reduced S10 levels that better mirrored phenotypes observed in the mouse spleen. Finally, we show that T3SS expression is sufficient to promote tolerance to the ribosome-targeting antibiotic, gentamicin. Collectively, these data indicate that the growth arrest associated with T3SS induction leads to decreased expression of ribosomal protein genes, and this results in reduced antibiotic susceptibility.
Collapse
Affiliation(s)
- Justin Greene
- W. Harry Feinstone Department of Molecular Microbiology and Immunology Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Rhett A. Snyder
- W. Harry Feinstone Department of Molecular Microbiology and Immunology Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Katherine L. Cotten
- W. Harry Feinstone Department of Molecular Microbiology and Immunology Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ryan C. Huiszoon
- Institute for Systems Research, University of Maryland, College Park, MD, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
| | - Sangwook Chu
- Institute for Systems Research, University of Maryland, College Park, MD, USA
| | - Rezia Era D. Braza
- W. Harry Feinstone Department of Molecular Microbiology and Immunology Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ashley A. Chapin
- Institute for Systems Research, University of Maryland, College Park, MD, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
| | - Justin M. Stine
- Institute for Systems Research, University of Maryland, College Park, MD, USA
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
| | - William E. Bentley
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
| | - Reza Ghodssi
- Institute for Systems Research, University of Maryland, College Park, MD, USA
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
| | - Kimberly M. Davis
- W. Harry Feinstone Department of Molecular Microbiology and Immunology Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
14
|
Fieldman T. Evolutionary principles for modifying pathogen virulence. Crit Rev Microbiol 2024; 50:385-396. [PMID: 37146153 DOI: 10.1080/1040841x.2023.2203766] [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: 12/21/2022] [Revised: 03/31/2023] [Accepted: 04/10/2023] [Indexed: 05/07/2023]
Abstract
Current methods for combatting infectious diseases are largely limited to the prevention of infection, enhancing host immunity (via vaccination), and administration of small molecules to slow the growth of or kill pathogens (e.g. antimicrobials). Beyond efforts to deter the rise of antimicrobial resistance, little consideration is given to pathogen evolution. Natural selection will favor different levels of virulence under different circumstances. Experimental studies and a wealth of theoretical work have identified many likely evolutionary determinants of virulence. Some of these, such as transmission dynamics, are amenable to modification by clinicians and public health practitioners. In this article, we provide a conceptual overview of virulence, followed by an analysis of modifiable evolutionary determinants of virulence including vaccinations, antibiotics, and transmission dynamics. Finally, we discuss both the importance and limitations of taking an evolutionary approach to reducing pathogen virulence.
Collapse
Affiliation(s)
- Tom Fieldman
- Clinical Microbiology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
15
|
Rojas VK, Winter MG, Jimenez AG, Tanner NW, Crockett SL, Spiga L, Hendrixson DR, Winter SE. Infection-associated gene regulation of L-tartrate metabolism in Salmonella enterica serovar Typhimurium. mBio 2024; 15:e0035024. [PMID: 38682906 PMCID: PMC11237755 DOI: 10.1128/mbio.00350-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 03/28/2024] [Indexed: 05/01/2024] Open
Abstract
Enteric pathogens such as Salmonella enterica serovar Typhimurium experience spatial and temporal changes to the metabolic landscape throughout infection. Host reactive oxygen and nitrogen species non-enzymatically convert monosaccharides to alpha hydroxy acids, including L-tartrate. Salmonella utilizes L-tartrate early during infection to support fumarate respiration, while L-tartrate utilization ceases at later time points due to the increased availability of exogenous electron acceptors such as tetrathionate, nitrate, and oxygen. It remains unknown how Salmonella regulates its gene expression to metabolically adapt to changing nutritional environments. Here, we investigated how the transcriptional regulation for L-tartrate metabolism in Salmonella is influenced by infection-relevant cues. L-tartrate induces the transcription of ttdBAU, genes involved in L-tartrate utilization. L-tartrate metabolism is negatively regulated by two previously uncharacterized transcriptional regulators TtdV (STM3357) and TtdW (STM3358), and both TtdV and TtdW are required for the sensing of L-tartrate. The electron acceptors nitrate, tetrathionate, and oxygen repress ttdBAU transcription via the two-component system ArcAB. Furthermore, the regulation of L-tartrate metabolism is required for optimal fitness in a mouse model of Salmonella-induced colitis. TtdV, TtdW, and ArcAB allow for the integration of two cues, i.e., substrate availability and availability of exogenous electron acceptors, to control L-tartrate metabolism. Our findings provide novel insights into how Salmonella prioritizes the utilization of different electron acceptors for respiration as it experiences transitional nutrient availability throughout infection. IMPORTANCE Bacterial pathogens must adapt their gene expression profiles to cope with diverse environments encountered during infection. This coordinated process is carried out by the integration of cues that the pathogen senses to fine-tune gene expression in a spatiotemporal manner. Many studies have elucidated the regulatory mechanisms of how Salmonella sense metabolites in the gut to activate or repress its virulence program; however, our understanding of how Salmonella coordinates its gene expression to maximize the utilization of carbon and energy sources found in transitional nutrient niches is not well understood. In this study, we discovered how Salmonella integrates two infection-relevant cues, substrate availability and exogenous electron acceptors, to control L-tartrate metabolism. From our experiments, we propose a model for how L-tartrate metabolism is regulated in response to different metabolic cues in addition to characterizing two previously unknown transcriptional regulators. This study expands our understanding of how microbes combine metabolic cues to enhance fitness during infection.
Collapse
Affiliation(s)
- Vivian K. Rojas
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, California, USA
| | - Maria G. Winter
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, California, USA
| | - Angel G. Jimenez
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Natasha W. Tanner
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, California, USA
| | - Stacey L. Crockett
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Luisella Spiga
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - David R. Hendrixson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sebastian E. Winter
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, California, USA
- Department of Medical Microbiology and Immunology, UC Davis School of Medicine, Davis, California, USA
| |
Collapse
|
16
|
Sutar AA, Dashpute RS, Shinde YD, Mukherjee S, Chowdhury C. A Systemic Review on Fitness and Survival of Salmonella in Dynamic Environment and Conceivable Ways of Its Mitigation. Indian J Microbiol 2024; 64:267-286. [PMID: 39011015 PMCID: PMC11246371 DOI: 10.1007/s12088-023-01176-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 12/05/2023] [Indexed: 07/17/2024] Open
Abstract
Gastroenteritis caused by non-typhoidal Salmonella still prevails resulting in several recent outbreaks affecting many people worldwide. The presence of invasive non-typhoidal Salmonella is exemplified by several characteristic symptoms and their severity relies on prominent risk factors. The persistence of this pathogen can be attributed to its broad host range, complex pathogenicity and virulence and adeptness in survival under challenging conditions inside the host. Moreover, a peculiar aid of the ever-changing climatic conditions grants this organism with remarkable potential to survive within the environment. Abusive use of antibiotics for the treatment of gastroenteritis has led to the emergence of multiple drug resistance, making the infections difficult to treat. This review emphasizes the importance of early detection of Salmonella, along with strategies for accomplishing it, as well as exploring alternative treatment approaches. The exceptional characteristics exhibited by Salmonella, like strategies of infection, persistence, and survival parallelly with multiple drug resistance, make this pathogen a prominent concern to human health.
Collapse
Affiliation(s)
- Ajit A Sutar
- Biochemical Sciences Division, CSIR- National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, MH 411008 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| | - Rohit S Dashpute
- Biochemical Sciences Division, CSIR- National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, MH 411008 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| | - Yashodhara D Shinde
- Biochemical Sciences Division, CSIR- National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, MH 411008 India
| | - Srestha Mukherjee
- Biochemical Sciences Division, CSIR- National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, MH 411008 India
| | - Chiranjit Chowdhury
- Biochemical Sciences Division, CSIR- National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, MH 411008 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| |
Collapse
|
17
|
Ernst C, Andreassen PR, Giger GH, Nguyen BD, Gäbelein CG, Guillaume-Gentil O, Fattinger SA, Sellin ME, Hardt WD, Vorholt JA. Direct Salmonella injection into enteroid cells allows the study of host-pathogen interactions in the cytosol with high spatiotemporal resolution. PLoS Biol 2024; 22:e3002597. [PMID: 38684033 PMCID: PMC11057982 DOI: 10.1371/journal.pbio.3002597] [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: 09/28/2023] [Accepted: 03/21/2024] [Indexed: 05/02/2024] Open
Abstract
Intestinal epithelial cells (IECs) play pivotal roles in nutrient uptake and in the protection against gut microorganisms. However, certain enteric pathogens, such as Salmonella enterica serovar Typhimurium (S. Tm), can invade IECs by employing flagella and type III secretion systems (T3SSs) with cognate effector proteins and exploit IECs as a replicative niche. Detection of flagella or T3SS proteins by IECs results in rapid host cell responses, i.e., the activation of inflammasomes. Here, we introduce a single-cell manipulation technology based on fluidic force microscopy (FluidFM) that enables direct bacteria delivery into the cytosol of single IECs within a murine enteroid monolayer. This approach allows to specifically study pathogen-host cell interactions in the cytosol uncoupled from preceding events such as docking, initiation of uptake, or vacuole escape. Consistent with current understanding, we show using a live-cell inflammasome reporter that exposure of the IEC cytosol to S. Tm induces NAIP/NLRC4 inflammasomes via its known ligands flagellin and T3SS rod and needle. Injected S. Tm mutants devoid of these invasion-relevant ligands were able to grow in the cytosol of IECs despite the absence of T3SS functions, suggesting that, in the absence of NAIP/NLRC4 inflammasome activation and the ensuing cell death, no effector-mediated host cell manipulation is required to render the epithelial cytosol growth-permissive for S. Tm. Overall, the experimental system to introduce S. Tm into single enteroid cells enables investigations into the molecular basis governing host-pathogen interactions in the cytosol with high spatiotemporal resolution.
Collapse
Affiliation(s)
- Chantal Ernst
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | | | - Gabriel H. Giger
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Bidong D. Nguyen
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | | | | | - Stefan A. Fattinger
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Mikael E. Sellin
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Julia A. Vorholt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
Figueroa-Bossi N, Fernández-Fernández R, Kerboriou P, Bouloc P, Casadesús J, Sánchez-Romero MA, Bossi L. Transcription-driven DNA supercoiling counteracts H-NS-mediated gene silencing in bacterial chromatin. Nat Commun 2024; 15:2787. [PMID: 38555352 PMCID: PMC10981669 DOI: 10.1038/s41467-024-47114-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 03/19/2024] [Indexed: 04/02/2024] Open
Abstract
In all living cells, genomic DNA is compacted through interactions with dedicated proteins and/or the formation of plectonemic coils. In bacteria, DNA compaction is achieved dynamically, coordinated with dense and constantly changing transcriptional activity. H-NS, a major bacterial nucleoid structuring protein, is of special interest due to its interplay with RNA polymerase. H-NS:DNA nucleoprotein filaments inhibit transcription initiation by RNA polymerase. However, the discovery that genes silenced by H-NS can be activated by transcription originating from neighboring regions has suggested that elongating RNA polymerases can disassemble H-NS:DNA filaments. In this study, we present evidence that transcription-induced counter-silencing does not require transcription to reach the silenced gene; rather, it exerts its effect at a distance. Counter-silencing is suppressed by introducing a DNA gyrase binding site within the intervening segment, suggesting that the long-range effect results from transcription-driven positive DNA supercoils diffusing toward the silenced gene. We propose a model wherein H-NS:DNA complexes form in vivo on negatively supercoiled DNA, with H-NS bridging the two arms of the plectoneme. Rotational diffusion of positive supercoils generated by neighboring transcription will cause the H-NS-bound negatively-supercoiled plectoneme to "unroll" disrupting the H-NS bridges and releasing H-NS.
Collapse
Affiliation(s)
- Nara Figueroa-Bossi
- Université Paris-Saclay, CEA, CNRS, Institut de Biologie Intégrative de la Cellule (I2BC), Gif-sur-Yvette, France
| | - Rocío Fernández-Fernández
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Patricia Kerboriou
- Université Paris-Saclay, CEA, CNRS, Institut de Biologie Intégrative de la Cellule (I2BC), Gif-sur-Yvette, France
| | - Philippe Bouloc
- Université Paris-Saclay, CEA, CNRS, Institut de Biologie Intégrative de la Cellule (I2BC), Gif-sur-Yvette, France
| | - Josep Casadesús
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | | | - Lionello Bossi
- Université Paris-Saclay, CEA, CNRS, Institut de Biologie Intégrative de la Cellule (I2BC), Gif-sur-Yvette, France.
| |
Collapse
|
19
|
Lories B, Belpaire TER, Smeets B, Steenackers HP. Competition quenching strategies reduce antibiotic tolerance in polymicrobial biofilms. NPJ Biofilms Microbiomes 2024; 10:23. [PMID: 38503782 PMCID: PMC10951329 DOI: 10.1038/s41522-024-00489-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/20/2024] [Indexed: 03/21/2024] Open
Abstract
Bacteria typically live in dense communities where they are surrounded by other species and compete for a limited amount of resources. These competitive interactions can induce defensive responses that also protect against antimicrobials, potentially complicating the antimicrobial treatment of pathogens residing in polymicrobial consortia. Therefore, we evaluate the potential of alternative antivirulence strategies that quench this response to competition. We test three competition quenching approaches: (i) interference with the attack mechanism of surrounding competitors, (ii) inhibition of the stress response systems that detect competition, and (iii) reduction of the overall level of competition in the community by lowering the population density. We show that either strategy can prevent the induction of antimicrobial tolerance of Salmonella Typhimurium in response to competitors. Competition quenching strategies can thus reduce tolerance of pathogens residing in polymicrobial communities and could contribute to the improved eradication of these pathogens via traditional methods.
Collapse
Affiliation(s)
- Bram Lories
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium
| | - Tom E R Belpaire
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium
- Division of Mechatronics, Biostatistics, and Sensors (MeBioS), Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Bart Smeets
- Division of Mechatronics, Biostatistics, and Sensors (MeBioS), Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Hans P Steenackers
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium.
| |
Collapse
|
20
|
Zhu H, Xiong Y, Jiang Z, Liu Q, Wang J. Quantifying Dynamic Phenotypic Heterogeneity in Resistant Escherichia coli under Translation-Inhibiting Antibiotics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304548. [PMID: 38193201 PMCID: PMC10953537 DOI: 10.1002/advs.202304548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/20/2023] [Indexed: 01/10/2024]
Abstract
Understanding the phenotypic heterogeneity of antibiotic-resistant bacteria following treatment and the transitions between different phenotypes is crucial for developing effective infection control strategies. The study expands upon previous work by explicating chloramphenicol-induced phenotypic heterogeneities in growth rate, gene expression, and morphology of resistant Escherichia coli using time-lapse microscopy. Correlating the bacterial growth rate and cspC expression, four interchangeable phenotypic subpopulations across varying antibiotic concentrations are identified, surpassing the previously described growth rate bistability. Notably, bacterial cells exhibiting either fast or slow growth rates can concurrently harbor subpopulations characterized by high and low gene expression levels, respectively. To elucidate the mechanisms behind this enhanced heterogeneity, a concise gene expression network model is proposed and the biological significance of the four phenotypes is further explored. Additionally, by employing Hidden Markov Model fitting and integrating the non-equilibrium landscape and flux theory, the real-time data encompassing diverse bacterial traits are analyzed. This approach reveals dynamic changes and switching kinetics in different cell fates, facilitating the quantification of observable behaviors and the non-equilibrium dynamics and thermodynamics at play. The results highlight the multi-dimensional heterogeneous behaviors of antibiotic-resistant bacteria under antibiotic stress, providing new insights into the compromised antibiotic efficacy, microbial response, and associated evolution processes.
Collapse
Affiliation(s)
- Haishuang Zhu
- State Key Laboratory of Electroanalytical ChemistryChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
- School of Applied Chemistry and EngineeringUniversity of Science and Technology of ChinaHefeiAnhui230026China
| | - Yixiao Xiong
- State Key Laboratory of Electroanalytical ChemistryChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
- School of Applied Chemistry and EngineeringUniversity of Science and Technology of ChinaHefeiAnhui230026China
| | - Zhenlong Jiang
- State Key Laboratory of Electroanalytical ChemistryChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
| | - Qiong Liu
- State Key Laboratory of Electroanalytical ChemistryChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
| | - Jin Wang
- Department of ChemistryPhysics and Applied MathematicsState University of New York at Stony Brook.Stony BrookNew York11794‐3400USA
| |
Collapse
|
21
|
Zhang C, Datta S, Ratcliff WC, Hammer BK. Constitutive expression of the Type VI Secretion System carries no measurable fitness cost in Vibrio cholerae. Ecol Evol 2024; 14:e11081. [PMID: 38435022 PMCID: PMC10905242 DOI: 10.1002/ece3.11081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 02/03/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024] Open
Abstract
The Type VI Secretion System (T6SS) is a widespread and highly effective mechanism of microbial warfare; it confers the ability to efficiently kill susceptible cells within close proximity. Due to its large physical size, complexity, and ballistic basis for intoxication, it has widely been assumed to incur significant growth costs in the absence of improved competitive outcomes. In this study, we precisely examine the fitness costs of constitutive T6SS firing in the bacterium Vibrio cholerae. We find that, contrary to expectations, constitutive expression of the T6SS has a negligible impact on growth, reducing growth fitness by 0.025 ± 0.5% (95% CI) relative to a T6SS- control. Mathematical modeling of microbial populations demonstrates that, due to clonal interference, constitutive expression of the T6SS will often be neutral, with little impact on evolutionary outcomes. Our findings underscore the importance of precisely measuring the fitness costs of microbial social behaviors and help explain the prevalence of the T6SS across Gram-negative bacteria.
Collapse
Affiliation(s)
- Christopher Zhang
- School of Biological SciencesGeorgia Institute of TechnologyAtlantaGeorgiaUSA
- Interdisciplinary Graduate Program in Quantitative BiosciencesGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Sayantan Datta
- School of Biological SciencesGeorgia Institute of TechnologyAtlantaGeorgiaUSA
- Interdisciplinary Graduate Program in Quantitative BiosciencesGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - William C. Ratcliff
- School of Biological SciencesGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Brian K. Hammer
- School of Biological SciencesGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| |
Collapse
|
22
|
Rojas VK, Winter MG, Jimenez AG, Tanner NW, Crockett SL, Spiga L, Hendrixson DR, Winter SE. Gene regulation of infection-associated L-tartrate metabolism in Salmonella enterica serovar Typhimurium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.578992. [PMID: 38370731 PMCID: PMC10871181 DOI: 10.1101/2024.02.05.578992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Enteric pathogens such as Salmonella enterica serovar Typhimurium experience spatial and temporal changes to the metabolic landscape throughout infection. Host reactive oxygen and nitrogen species non-enzymatically convert monosaccharides to alpha hydroxy acids, including L-tartrate. Salmonella utilizes L-tartrate early during infection to support fumarate respiration, while L-tartrate utilization ceases at later time points due to the increased availability of exogenous electron acceptors such as tetrathionate, nitrate, and oxygen. It remains unknown how Salmonella regulates its gene expression to metabolically adapt to changing nutritional environments. Here, we investigated how the transcriptional regulation for L-tartrate metabolism in Salmonella is influenced by infection-relevant cues. L-tartrate induces the transcription of ttdBAU, genes involved in L-tartrate utilization. L-tartrate metabolism is negatively regulated by two previously uncharacterized transcriptional regulators TtdV (STM3357) and TtdW (STM3358), and both TtdV and TtdW are required for sensing of L-tartrate. The electron acceptors nitrate, tetrathionate, and oxygen repress ttdBAU transcription via the two-component system ArcAB. Furthermore, regulation of L-tartrate metabolism is required for optimal fitness in a mouse model of Salmonella-induced colitis. TtdV, TtdW, and ArcAB allow for the integration of two cues, substrate availability and availability of exogenous electron acceptors, to control L-tartrate metabolism. Our findings provide novel insights into how Salmonella prioritizes utilization of different electron acceptors for respiration as it experiences transitional nutrient availability throughout infection.
Collapse
Affiliation(s)
- Vivian K. Rojas
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, CA, USA
| | - Maria G. Winter
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, CA, USA
| | - Angel G. Jimenez
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Current address: Infectious Diseases, Genentech, South San Francisco, California, USA
| | - Natasha W. Tanner
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, CA, USA
| | - Stacey L. Crockett
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Luisella Spiga
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Current address: Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David R. Hendrixson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sebastian E. Winter
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis School of Medicine, Davis, CA, USA
- Department of Medical Microbiology and Immunology, UC Davis School of Medicine, Davis, CA, USA
| |
Collapse
|
23
|
Wimmi S, Fleck M, Helbig C, Brianceau C, Langenfeld K, Szymanski WG, Angelidou G, Glatter T, Diepold A. Pilotins are mobile T3SS components involved in assembly and substrate specificity of the bacterial type III secretion system. Mol Microbiol 2024; 121:304-323. [PMID: 38178634 DOI: 10.1111/mmi.15223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 12/17/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024]
Abstract
In animal pathogens, assembly of the type III secretion system injectisome requires the presence of so-called pilotins, small lipoproteins that assist the formation of the secretin ring in the outer membrane. Using a combination of functional assays, interaction studies, proteomics, and live-cell microscopy, we determined the contribution of the pilotin to the assembly, function, and substrate selectivity of the T3SS and identified potential new downstream roles of pilotin proteins. In absence of its pilotin SctG, Yersinia enterocolitica forms few, largely polar injectisome sorting platforms and needles. Accordingly, most export apparatus subcomplexes are mobile in these strains, suggesting the absence of fully assembled injectisomes. Remarkably, while absence of the pilotin all but prevents export of early T3SS substrates, such as the needle subunits, it has little effect on secretion of late T3SS substrates, including the virulence effectors. We found that although pilotins interact with other injectisome components such as the secretin in the outer membrane, they mostly localize in transient mobile clusters in the bacterial membrane. Together, these findings provide a new view on the role of pilotins in the assembly and function of type III secretion injectisomes.
Collapse
Affiliation(s)
- Stephan Wimmi
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Moritz Fleck
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Carlos Helbig
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Corentin Brianceau
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Katja Langenfeld
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Witold G Szymanski
- Mass Spectrometry and Proteomics Facility, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Georgia Angelidou
- Mass Spectrometry and Proteomics Facility, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Timo Glatter
- Mass Spectrometry and Proteomics Facility, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Andreas Diepold
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| |
Collapse
|
24
|
Taillefer B, Giraud JF, Cascales E. No fitness cost entailed by type VI secretion system synthesis, assembly, contraction, or disassembly in enteroaggregative Escherichia coli. J Bacteriol 2023; 205:e0035723. [PMID: 37971272 PMCID: PMC10729742 DOI: 10.1128/jb.00357-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023] Open
Abstract
IMPORTANCE Bacteria use weapons to deliver effectors into target cells. One of these weapons, the type VI secretion system (T6SS), assembles a contractile tail acting as a spring to propel a toxin-loaded needle. Due to its size and mechanism of action, the T6SS was intuitively thought to be energetically costly. Here, using a combination of mutants and growth measurements in liquid medium, on plates, and in competition experiments, we show that the T6SS does not entail a growth cost to enteroaggregative Escherichia coli.
Collapse
Affiliation(s)
- Boris Taillefer
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM, UMR 7255), Institut de Microbiologie de la Méditerranée (IMM), Aix Marseille Univ, CNRS, Marseille, France
| | - Julien F. Giraud
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM, UMR 7255), Institut de Microbiologie de la Méditerranée (IMM), Aix Marseille Univ, CNRS, Marseille, France
| | - Eric Cascales
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM, UMR 7255), Institut de Microbiologie de la Méditerranée (IMM), Aix Marseille Univ, CNRS, Marseille, France
| |
Collapse
|
25
|
Ciolli Mattioli C, Eisner K, Rosenbaum A, Wang M, Rivalta A, Amir A, Golding I, Avraham R. Physiological stress drives the emergence of a Salmonella subpopulation through ribosomal RNA regulation. Curr Biol 2023; 33:4880-4892.e14. [PMID: 37879333 PMCID: PMC10843543 DOI: 10.1016/j.cub.2023.09.064] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/24/2023] [Accepted: 09/26/2023] [Indexed: 10/27/2023]
Abstract
Bacteria undergo cycles of growth and starvation to which they must adapt swiftly. One important strategy for adjusting growth rates relies on ribosomal levels. Although high ribosomal levels are required for fast growth, their dynamics during starvation remain unclear. Here, we analyzed ribosomal RNA (rRNA) content of individual Salmonella cells by using fluorescence in situ hybridization (rRNA-FISH) and measured a dramatic decrease in rRNA numbers only in a subpopulation during nutrient limitation, resulting in a bimodal distribution of cells with high and low rRNA content. During nutritional upshifts, the two subpopulations were associated with distinct phenotypes. Using a transposon screen coupled with rRNA-FISH, we identified two mutants, DksA and RNase I, acting on rRNA transcription shutdown and degradation, which abolished the formation of the subpopulation with low rRNA content. Our work identifies a bacterial mechanism for regulation of ribosomal bimodality that may be beneficial for population survival during starvation.
Collapse
Affiliation(s)
- Camilla Ciolli Mattioli
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Kfir Eisner
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Aviel Rosenbaum
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Mengyu Wang
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Andre' Rivalta
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ariel Amir
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ido Golding
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Roi Avraham
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
26
|
Davis KM. mSphere of Influence: If virulence is energetically costly, how can it be maintained? mSphere 2023; 8:e0031223. [PMID: 37676232 PMCID: PMC10597342 DOI: 10.1128/msphere.00312-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023] Open
Abstract
Kimberly Davis works in the field of bacterial pathogenesis and studies heterogeneity in bacterial populations within host tissues. In this mSphere of Influence article, she reflects on how the paper "Stabilization of cooperative virulence by the expression of an avirulent phenotype" by M. Diard et al. (M. Diard et al., Nature 494:353-6, 2013, DOI: 10.1038/nature11913) impacted the way she thinks about bacterial population dynamics and the costs and benefits of producing virulence factors during infection.
Collapse
Affiliation(s)
- Kimberly Michele Davis
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
27
|
Pokorzynski ND, Groisman EA. How Bacterial Pathogens Coordinate Appetite with Virulence. Microbiol Mol Biol Rev 2023; 87:e0019822. [PMID: 37358444 PMCID: PMC10521370 DOI: 10.1128/mmbr.00198-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023] Open
Abstract
Cells adjust growth and metabolism to nutrient availability. Having access to a variety of carbon sources during infection of their animal hosts, facultative intracellular pathogens must efficiently prioritize carbon utilization. Here, we discuss how carbon source controls bacterial virulence, with an emphasis on Salmonella enterica serovar Typhimurium, which causes gastroenteritis in immunocompetent humans and a typhoid-like disease in mice, and propose that virulence factors can regulate carbon source prioritization by modifying cellular physiology. On the one hand, bacterial regulators of carbon metabolism control virulence programs, indicating that pathogenic traits appear in response to carbon source availability. On the other hand, signals controlling virulence regulators may impact carbon source utilization, suggesting that stimuli that bacterial pathogens experience within the host can directly impinge on carbon source prioritization. In addition, pathogen-triggered intestinal inflammation can disrupt the gut microbiota and thus the availability of carbon sources. By coordinating virulence factors with carbon utilization determinants, pathogens adopt metabolic pathways that may not be the most energy efficient because such pathways promote resistance to antimicrobial agents and also because host-imposed deprivation of specific nutrients may hinder the operation of certain pathways. We propose that metabolic prioritization by bacteria underlies the pathogenic outcome of an infection.
Collapse
Affiliation(s)
- Nick D. Pokorzynski
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
| | - Eduardo A. Groisman
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
- Yale Microbial Sciences Institute, West Haven, Connecticut, USA
| |
Collapse
|
28
|
Anderson JC. Ill Communication: Host Metabolites as Virulence-Regulating Signals for Plant-Pathogenic Bacteria. ANNUAL REVIEW OF PHYTOPATHOLOGY 2023; 61:49-71. [PMID: 37253693 DOI: 10.1146/annurev-phyto-021621-114026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Plant bacterial pathogens rely on host-derived signals to coordinate the deployment of virulence factors required for infection. In this review, I describe how diverse plant-pathogenic bacteria detect and respond to plant-derived metabolic signals for the purpose of virulence gene regulation. I highlight examples of how pathogens perceive host metabolites through membrane-localized receptors as well as intracellular response mechanisms. Furthermore, I describe how individual strains may coordinate their virulence using multiple distinct host metabolic signals, and how plant signals may positively or negatively regulate virulence responses. I also describe how plant defenses may interfere with the perception of host metabolites as a means to dampen pathogen virulence. The emerging picture is that recognition of host metabolic signals for the purpose of virulence gene regulation represents an important primary layer of interaction between pathogenic bacteria and host plants that shapes infection outcomes.
Collapse
Affiliation(s)
- Jeffrey C Anderson
- Department of Botany and Plant Pathology, Oregon State University, Corvallis, Oregon, USA;
| |
Collapse
|
29
|
Nava-Galeana J, Núñez C, Bustamante VH. Proteomic analysis reveals the global effect of the BarA/SirA-Csr regulatory cascade in Salmonella Typhimurium grown in conditions that favor the expression of invasion genes. J Proteomics 2023; 286:104960. [PMID: 37451358 DOI: 10.1016/j.jprot.2023.104960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023]
Abstract
In many bacteria, the BarA/SirA and Csr regulatory systems control expression of genes encoding a wide variety of cellular functions. The BarA/SirA two-component system induces the expression of CsrB and CsrC, two small non-coding RNAs that sequester CsrA, a protein that binds to target mRNAs and thus negatively or positively regulates their expression. BarA/SirA and CsrB/C induce expression of the Salmonella Pathogenicity Island 1 (SPI-1) genes required for Salmonella invasion of host cells. To further investigate the regulatory role of the BarA/SirA and Csr systems in Salmonella, we performed LC-MS/MS proteomic analysis using the WT S. Typhimurium strain and its derived ΔsirA and ΔcsrB ΔcsrC mutants grown in SPI-1-inducing conditions. The expression of 164 proteins with a wide diversity, or unknown, functions was significantly affected positively or negatively by the absence of SirA and/or CsrB/C. Interestingly, 19 proteins were identified as new targets for SirA-CsrB/C. Our results support that SirA and CsrB/C act in a cascade fashion to regulate gene expression in S. Typhimurium in the conditions tested. Notably, our results show that SirA-CsrB/C-CsrA controls expression of proteins required for the replication of Salmonella in the intestinal lumen, in an opposite way to its control exerted on the SPI-1 proteins. SIGNIFICANCE: The BarA/SirA and Csr global regulatory systems control a wide range of cellular processes, including the expression of virulence genes. For instance, in Salmonella, BarA/SirA and CsrB/C positively regulate expression of the SPI-1 genes, which are required for Salmonella invasion to host cells. In this study, by performing a proteomic analysis, we identified 164 proteins whose expression was positively or negatively controlled by SirA and CsrB/C in SPI-1-inducing conditions, including 19 new possible targets of these systems. Our results support the action of SirA and CsrB/C in a cascade fashion to control different cellular processes in Salmonella. Interestingly, our data indicate that SirA-CsrB/C-CsrA controls inversely the expression of proteins required for invasion of the intestinal epithelium and for replication in the intestinal lumen, which suggests a role for this regulatory cascade as a molecular switch for Salmonella virulence. Thus, our study further expands the insight into the regulatory mechanisms governing the virulence and physiology of an important pathogen.
Collapse
Affiliation(s)
- Jessica Nava-Galeana
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| | - Cinthia Núñez
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| | - Víctor H Bustamante
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico.
| |
Collapse
|
30
|
Nava-Galeana J, Yakhnin H, Babitzke P, Bustamante VH. CsrA Positively and Directly Regulates the Expression of the pdu, pocR, and eut Genes Required for the Luminal Replication of Salmonella Typhimurium. Microbiol Spectr 2023; 11:e0151623. [PMID: 37358421 PMCID: PMC10433801 DOI: 10.1128/spectrum.01516-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/26/2023] [Indexed: 06/27/2023] Open
Abstract
Enteric pathogens, such as Salmonella, have evolved to thrive in the inflamed gut. Genes located within the Salmonella pathogenicity island 1 (SPI-1) mediate the invasion of cells from the intestinal epithelium and the induction of an intestinal inflammatory response. Alternative electron acceptors become available in the inflamed gut and are utilized by Salmonella for luminal replication through the metabolism of propanediol and ethanolamine, using the enzymes encoded by the pdu and eut genes. The RNA-binding protein CsrA inhibits the expression of HilD, which is the central transcriptional regulator of the SPI-1 genes. Previous studies suggest that CsrA also regulates the expression of the pdu and eut genes, but the mechanism for this regulation is unknown. In this work, we show that CsrA positively regulates the pdu genes by binding to the pocR and pduA transcripts as well as the eut genes by binding to the eutS transcript. Furthermore, our results show that the SirA-CsrB/CsrC-CsrA regulatory cascade controls the expression of the pdu and eut genes mediated by PocR or EutR, which are the positive AraC-like transcriptional regulators for the pdu and eut genes, respectively. By oppositely regulating the expression of genes for invasion and for luminal replication, the SirA-CsrB/CsrC-CsrA regulatory cascade could be involved in the generation of two Salmonella populations that cooperate for intestinal colonization and transmission. Our study provides new insight into the regulatory mechanisms that govern Salmonella virulence. IMPORTANCE The regulatory mechanisms that control the expression of virulence genes are essential for bacteria to infect hosts. Salmonella has developed diverse regulatory mechanisms to colonize the host gut. For instance, the SirA-CsrB/CsrC-CsrA regulatory cascade controls the expression of the SPI-1 genes, which are required for this bacterium to invade intestinal epithelium cells and for the induction of an intestinal inflammatory response. In this study, we determine the mechanisms by which the SirA-CsrB/CsrC-CsrA regulatory cascade controls the expression of the pdu and eut genes, which are necessary for the replication of Salmonella in the intestinal lumen. Thus, our data, together with the results of previous reports, indicate that the SirA-CsrB/CsrC-CsrA regulatory cascade has an important role in the intestinal colonization by Salmonella.
Collapse
Affiliation(s)
- Jessica Nava-Galeana
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Helen Yakhnin
- Department of Biochemistry and Molecular Biology, Center for RNA Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Paul Babitzke
- Department of Biochemistry and Molecular Biology, Center for RNA Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Víctor H. Bustamante
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| |
Collapse
|
31
|
Gül E, Bakkeren E, Salazar G, Steiger Y, Abi Younes A, Clerc M, Christen P, Fattinger SA, Nguyen BD, Kiefer P, Slack E, Ackermann M, Vorholt JA, Sunagawa S, Diard M, Hardt WD. The microbiota conditions a gut milieu that selects for wild-type Salmonella Typhimurium virulence. PLoS Biol 2023; 21:e3002253. [PMID: 37651408 PMCID: PMC10499267 DOI: 10.1371/journal.pbio.3002253] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/13/2023] [Accepted: 07/13/2023] [Indexed: 09/02/2023] Open
Abstract
Salmonella Typhimurium elicits gut inflammation by the costly expression of HilD-controlled virulence factors. This inflammation alleviates colonization resistance (CR) mediated by the microbiota and thereby promotes pathogen blooms. However, the inflamed gut-milieu can also select for hilD mutants, which cannot elicit or maintain inflammation, therefore causing a loss of the pathogen's virulence. This raises the question of which conditions support the maintenance of virulence in S. Typhimurium. Indeed, it remains unclear why the wild-type hilD allele is dominant among natural isolates. Here, we show that microbiota transfer from uninfected or recovered hosts leads to rapid clearance of hilD mutants that feature attenuated virulence, and thereby contributes to the preservation of the virulent S. Typhimurium genotype. Using mouse models featuring a range of microbiota compositions and antibiotic- or inflammation-inflicted microbiota disruptions, we found that irreversible disruption of the microbiota leads to the accumulation of hilD mutants. In contrast, in models with a transient microbiota disruption, selection for hilD mutants was prevented by the regrowing microbiota community dominated by Lachnospirales and Oscillospirales. Strikingly, even after an irreversible microbiota disruption, microbiota transfer from uninfected donors prevented the rise of hilD mutants. Our results establish that robust S. Typhimurium gut colonization hinges on optimizing its manipulation of the host: A transient and tempered microbiota perturbation is favorable for the pathogen to both flourish in the inflamed gut and also minimize loss of virulence. Moreover, besides conferring CR, the microbiota may have the additional consequence of maintaining costly enteropathogen virulence mechanisms.
Collapse
Affiliation(s)
- Ersin Gül
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Erik Bakkeren
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Department of Biology, University of Oxford, Oxford, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Guillem Salazar
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Institute of Microbiology and Swiss Institute of Bioinformatics, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Yves Steiger
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Andrew Abi Younes
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Melanie Clerc
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Philipp Christen
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Stefan A. Fattinger
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Bidong D. Nguyen
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Patrick Kiefer
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Emma Slack
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Institute for Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Martin Ackermann
- Institute of Biogeochemistry and Pollutant Dynamics, Department of Environmental Systems Science, ETH Zurich, Zurich, Switzerland
- Department of Environmental Microbiology, Eawag, Duebendorf, Switzerland
| | - Julia A. Vorholt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Shinichi Sunagawa
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Institute of Microbiology and Swiss Institute of Bioinformatics, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Médéric Diard
- Biozentrum, University of Basel, Basel, Switzerland
- Botnar Research Centre for Child Health, Basel, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Heyman O, Yehezkel D, Ciolli Mattioli C, Blumberger N, Rosenberg G, Solomon A, Hoffman D, Bossel Ben-Moshe N, Avraham R. Paired single-cell host profiling with multiplex-tagged bacterial mutants reveals intracellular virulence-immune networks. Proc Natl Acad Sci U S A 2023; 120:e2218812120. [PMID: 37399397 PMCID: PMC10334762 DOI: 10.1073/pnas.2218812120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 05/24/2023] [Indexed: 07/05/2023] Open
Abstract
Encounters between host cells and intracellular bacterial pathogens lead to complex phenotypes that determine the outcome of infection. Single-cell RNA sequencing (scRNA-seq) is increasingly used to study the host factors underlying diverse cellular phenotypes but has limited capacity to analyze the role of bacterial factors. Here, we developed scPAIR-seq, a single-cell approach to analyze infection with a pooled library of multiplex-tagged, barcoded bacterial mutants. Infected host cells and barcodes of intracellular bacterial mutants are both captured by scRNA-seq to functionally analyze mutant-dependent changes in host transcriptomes. We applied scPAIR-seq to macrophages infected with a library of Salmonella Typhimurium secretion system effector mutants. We analyzed redundancy between effectors and mutant-specific unique fingerprints and mapped the global virulence network of each individual effector by its impact on host immune pathways. ScPAIR-seq is a powerful tool to untangle bacterial virulence strategies and their complex interplay with host defense strategies that drive infection outcome.
Collapse
Affiliation(s)
- Ori Heyman
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Dror Yehezkel
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Camilla Ciolli Mattioli
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Neta Blumberger
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Gili Rosenberg
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Aryeh Solomon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Dotan Hoffman
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Noa Bossel Ben-Moshe
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Roi Avraham
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| |
Collapse
|
33
|
Saleh DO, Horstmann JA, Giralt-Zúñiga M, Weber W, Kaganovitch E, Durairaj AC, Klotzsch E, Strowig T, Erhardt M. SPI-1 virulence gene expression modulates motility of Salmonella Typhimurium in a proton motive force- and adhesins-dependent manner. PLoS Pathog 2023; 19:e1011451. [PMID: 37315106 DOI: 10.1371/journal.ppat.1011451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/01/2023] [Indexed: 06/16/2023] Open
Abstract
Both the bacterial flagellum and the evolutionary related injectisome encoded on the Salmonella pathogenicity island 1 (SPI-1) play crucial roles during the infection cycle of Salmonella species. The interplay of both is highlighted by the complex cross-regulation that includes transcriptional control of the flagellar master regulatory operon flhDC by HilD, the master regulator of SPI-1 gene expression. Contrary to the HilD-dependent activation of flagellar gene expression, we report here that activation of HilD resulted in a dramatic loss of motility, which was dependent on the presence of SPI-1. Single cell analyses revealed that HilD-activation triggers a SPI-1-dependent induction of the stringent response and a substantial decrease in proton motive force (PMF), while flagellation remains unaffected. We further found that HilD activation enhances the adhesion of Salmonella to epithelial cells. A transcriptome analysis revealed a simultaneous upregulation of several adhesin systems, which, when overproduced, phenocopied the HilD-induced motility defect. We propose a model where the SPI-1-dependent depletion of the PMF and the upregulation of adhesins upon HilD-activation enable flagellated Salmonella to rapidly modulate their motility during infection, thereby enabling efficient adhesion to host cells and delivery of effector proteins.
Collapse
Affiliation(s)
- Doaa Osama Saleh
- Institute for Biology/Molecular Microbiology, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Julia A Horstmann
- Junior Research Group Infection Biology of Salmonella, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - María Giralt-Zúñiga
- Institute for Biology/Molecular Microbiology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Willi Weber
- Institute for Biology, Experimental Biophysics/Mechanobiology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Eugen Kaganovitch
- Max Planck Institute for Terrestrial Microbiology and Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
| | - Abilash Chakravarthy Durairaj
- Junior Research Group Infection Biology of Salmonella, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Enrico Klotzsch
- Institute for Biology, Experimental Biophysics/Mechanobiology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Marc Erhardt
- Institute for Biology/Molecular Microbiology, Humboldt-Universität zu Berlin, Berlin, Germany
- Max Planck Unit for the Science of Pathogens, Berlin, Germany
| |
Collapse
|
34
|
Lindsay RJ, Holder PJ, Talbot NJ, Gudelj I. Metabolic efficiency reshapes the seminal relationship between pathogen growth rate and virulence. Ecol Lett 2023; 26:896-907. [PMID: 37056166 PMCID: PMC10947253 DOI: 10.1111/ele.14218] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 04/15/2023]
Abstract
A cornerstone of classical virulence evolution theories is the assumption that pathogen growth rate is positively correlated with virulence, the amount of damage pathogens inflict on their hosts. Such theories are key for incorporating evolutionary principles into sustainable disease management strategies. Yet, empirical evidence raises doubts over this central assumption underpinning classical theories, thus undermining their generality and predictive power. In this paper, we identify a key component missing from current theories which redefines the growth-virulence relationship in a way that is consistent with data. By modifying the activity of a single metabolic gene, we engineered strains of Magnaporthe oryzae with different nutrient acquisition and growth rates. We conducted in planta infection studies and uncovered an unexpected non-monotonic relationship between growth rate and virulence that is jointly shaped by how growth rate and metabolic efficiency interact. This novel mechanistic framework paves the way for a much-needed new suite of virulence evolution theories.
Collapse
Affiliation(s)
| | | | - Nicholas J. Talbot
- The Sainsbury LaboratoryUniversity of East Anglia, Norwich Research ParkNorwichUK
| | - Ivana Gudelj
- Biosciences and Living Systems InstituteUniversity of ExeterExeterUK
| |
Collapse
|
35
|
Schott S, Scheuer R, Ermoli F, Glatter T, Evguenieva-Hackenberg E, Diepold A. A ParDE toxin-antitoxin system is responsible for the maintenance of the Yersinia virulence plasmid but not for type III secretion-associated growth inhibition. Front Cell Infect Microbiol 2023; 13:1166077. [PMID: 37228670 PMCID: PMC10203498 DOI: 10.3389/fcimb.2023.1166077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
Many Gram-negative pathogens utilize the type III secretion system (T3SS) to translocate virulence-promoting effector proteins into eukaryotic host cells. The activity of this system results in a severe reduction of bacterial growth and division, summarized as secretion-associated growth inhibition (SAGI). In Yersinia enterocolitica, the T3SS and related proteins are encoded on a virulence plasmid. We identified a ParDE-like toxin-antitoxin system on this virulence plasmid in genetic proximity to yopE, encoding a T3SS effector. Effectors are strongly upregulated upon activation of the T3SS, indicating a potential role of the ParDE system in the SAGI or maintenance of the virulence plasmid. Expression of the toxin ParE in trans resulted in reduced growth and elongated bacteria, highly reminiscent of the SAGI. Nevertheless, the activity of ParDE is not causal for the SAGI. T3SS activation did not influence ParDE activity; conversely, ParDE had no impact on T3SS assembly or activity itself. However, we found that ParDE ensures the presence of the T3SS across bacterial populations by reducing the loss of the virulence plasmid, especially under conditions relevant to infection. Despite this effect, a subset of bacteria lost the virulence plasmid and regained the ability to divide under secreting conditions, facilitating the possible emergence of T3SS-negative bacteria in late acute and persistent infections.
Collapse
Affiliation(s)
- Saskia Schott
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Robina Scheuer
- Department of Microbiology and Molecular Biology, Justus Liebig University Gießen, Gießen, Germany
| | - Francesca Ermoli
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Timo Glatter
- Core Facility for Mass spectrometry & Proteomics, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | | | - Andreas Diepold
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| |
Collapse
|
36
|
Hernández-Ledesma A, Cabrera-Díaz E, Arvizu-Medrano SM, Gómez-Baltazar A, Hernández-Iturriaga M, Godínez-Oviedo A. Virulence and antimicrobial resistance profiles of Salmonella enterica isolated from foods, humans, and the environment in Mexico. Int J Food Microbiol 2023; 391-393:110135. [PMID: 36827747 DOI: 10.1016/j.ijfoodmicro.2023.110135] [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: 11/03/2022] [Revised: 01/11/2023] [Accepted: 02/09/2023] [Indexed: 02/21/2023]
Abstract
Salmonella enterica genotypic and phenotypic characteristics play an important role in its pathogenesis, which could be influenced by its origin. This study evaluated the association among the antimicrobial resistance, virulence, and origin of circulating S. enterica strains in Mexico, isolated from foods, humans, and the environment. The antimicrobial susceptibility to fourteen antibiotics by the Kirby-Bauer method (n = 117), and the presence of thirteen virulence genes by multiplex PCR (n = 153) and by sequence alignments (n = 2963) were evaluated. In addition, a set of S. enterica isolates from Mexico (n = 344) previously characterized according to their genotypic and phenotypic print was included to increase the coverage of the association analysis. Strains with the presence of sopE and strains with the absence of sspH1 were significantly associated with multidrug-resistant (MDR) phenotypes (p < 0.05). The origin of the strains had significant associations with the antimicrobial profiles and some virulence genes (hilA, orgA, sifA, ssaQ, sseL, sspH1, pefA, and spvC) (p < 0.05). Animal-origin food isolates showed the highest frequency of MDR (57.2 %), followed by human isolates (30.0 %). Also, sspH1, pefA, and spvC were found in major frequency in human (32.4 %, 31.0 %, 31.7 %) and animal-origin foods (41.6 %, 10.6 %, 10.6 %) isolates. The findings highlighted that antimicrobial profiles and specific virulence genes of S. enterica strains are related to their origin. Similar genotypic and phenotypic characteristics between human and animal-origin foods isolates were found, suggesting that animal-origin foods isolates are the most responsible for human cases. The revealed associations can be used to improve risk estimation assessments in national food safety surveillance programs.
Collapse
Affiliation(s)
- A Hernández-Ledesma
- Departamento de Investigación y Posgrado de Alimentos, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de las Campanas s/n, Col. Las Campanas, 76010 Querétaro, Qro., Mexico
| | - E Cabrera-Díaz
- Departamento de Salud Pública, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Camino Ramón Padilla Sánchez 2100, Zapopan, Jalisco 45200, Mexico
| | - S M Arvizu-Medrano
- Departamento de Investigación y Posgrado de Alimentos, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de las Campanas s/n, Col. Las Campanas, 76010 Querétaro, Qro., Mexico
| | - A Gómez-Baltazar
- Departamento de Investigación y Posgrado de Alimentos, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de las Campanas s/n, Col. Las Campanas, 76010 Querétaro, Qro., Mexico
| | - M Hernández-Iturriaga
- Departamento de Investigación y Posgrado de Alimentos, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de las Campanas s/n, Col. Las Campanas, 76010 Querétaro, Qro., Mexico
| | - A Godínez-Oviedo
- Departamento de Investigación y Posgrado de Alimentos, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de las Campanas s/n, Col. Las Campanas, 76010 Querétaro, Qro., Mexico.
| |
Collapse
|
37
|
Fernández-Fernández R, López-Igual R, Casadesús J, Sánchez-Romero MA. Analysis of Salmonella lineage-specific traits upon cell sorting. Front Cell Infect Microbiol 2023; 13:1146070. [PMID: 37065195 PMCID: PMC10090396 DOI: 10.3389/fcimb.2023.1146070] [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: 01/16/2023] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
Microbial cell individuality is receiving increasing interest in the scientific community. Individual cells within clonal populations exhibit noticeable phenotypic heterogeneity. The advent of fluorescent protein technology and advances in single-cell analysis has revealed phenotypic cell variant in bacterial populations. This heterogeneity is evident in a wide range of phenotypes, for example, individual cells display variable degrees of gene expression and survival under selective conditions and stresses, and can exhibit differing propensities to host interactions. Last few years, numerous cell sorting approaches have been employed for resolving the properties of bacterial subpopulations. This review provides an overview of applications of cell sorting to analyze Salmonella lineage-specific traits, including bacterial evolution studies, gene expression analysis, response to diverse cellular stresses and characterization of diverse bacterial phenotypic variants.
Collapse
Affiliation(s)
- Rocío Fernández-Fernández
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Seville, Spain
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| | - Rocío López-Igual
- Instituto de Bioquímica Vegetal y Fotosíntesis, Universidad de Sevilla and C.S.I.C., Seville, Spain
| | - Josep Casadesús
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| | - María Antonia Sánchez-Romero
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
- *Correspondence: María Antonia Sánchez-Romero,
| |
Collapse
|
38
|
Vincent MS, Vergnes A, Ezraty B. Chlorate Contamination in Commercial Growth Media as a Source of Phenotypic Heterogeneity within Bacterial Populations. Microbiol Spectr 2023; 11:e0499122. [PMID: 36752622 PMCID: PMC10100951 DOI: 10.1128/spectrum.04991-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/18/2023] [Indexed: 02/09/2023] Open
Abstract
Under anaerobic conditions, chlorate is reduced to chlorite, a cytotoxic compound that triggers oxidative stress within bacterial cultures. We previously found that BD Bacto Casamino Acids were contaminated with chlorate. In this study, we investigated whether chlorate contamination is detectable in other commercial culture media. We provide evidence that in addition to different batches of BD Bacto Casamino Acids, several commercial agar powders are contaminated with chlorate. A direct consequence of this contamination is that, during anaerobic growth, Escherichia coli cells activate the expression of msrP, a gene encoding periplasmic methionine sulfoxide reductase, which repairs oxidized protein-bound methionine. We further demonstrate that during aerobic growth, progressive oxygen depletion triggers msrP expression in a subpopulation of cells due to the presence of chlorate. Hence, we propose that chlorate contamination in commercial growth media is a source of phenotypic heterogeneity within bacterial populations. IMPORTANCE Agar is arguably the most utilized solidifying agent for microbiological media. In this study, we show that agar powders from different suppliers, as well as certain batches of BD Bacto Casamino Acids, contain significant levels of chlorate. We demonstrate that this contamination induces the expression of a methionine sulfoxide reductase, suggesting the presence of intracellular oxidative damage. Our results should alert the microbiology community to a pitfall in the cultivation of microorganisms under laboratory conditions.
Collapse
Affiliation(s)
- Maxence S. Vincent
- Aix-Marseille Université, CNRS, Laboratoire de Chimie Bactérienne, Institut de Microbiologie de la Méditerranée, Marseille, France
| | - Alexandra Vergnes
- Aix-Marseille Université, CNRS, Laboratoire de Chimie Bactérienne, Institut de Microbiologie de la Méditerranée, Marseille, France
| | - Benjamin Ezraty
- Aix-Marseille Université, CNRS, Laboratoire de Chimie Bactérienne, Institut de Microbiologie de la Méditerranée, Marseille, France
| |
Collapse
|
39
|
Small RNAs Activate Salmonella Pathogenicity Island 1 by Modulating mRNA Stability through the hilD mRNA 3' Untranslated Region. J Bacteriol 2023; 205:e0033322. [PMID: 36472436 PMCID: PMC9879128 DOI: 10.1128/jb.00333-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Salmonella enterica serovar Typhimurium is an enteric pathogen associated with foodborne disease. Salmonella invades the intestinal epithelium using a type three secretion system encoded on Salmonella pathogenicity island 1 (SPI-1). SPI-1 genes are tightly regulated by a complex feed-forward loop to ensure proper spatial and temporal expression. Most regulatory input is integrated at HilD, through control of hilD mRNA translation or HilD protein activity. The hilD mRNA possesses a 310-nucleotide 3' untranslated region (UTR) that influences HilD and SPI-1 expression, and this regulation is dependent on Hfq and RNase E, cofactors known to mediate small RNA (sRNA) activities. Thus, we hypothesized that the hilD mRNA 3' UTR is a target for sRNAs. Here, we show that two sRNAs, SdsR and Spot 42, regulate SPI-1 by targeting different regions of the hilD mRNA 3' UTR. Regulatory activities of these sRNAs depended on Hfq and RNase E, in agreement with previous roles found for both at the hilD 3' UTR. Salmonella mutants lacking SdsR and Spot 42 had decreased virulence in a mouse model of infection. Collectively, this work suggests that these sRNAs targeting the hilD mRNA 3' UTR increase hilD mRNA levels by interfering with RNase E-dependent mRNA degradation and that this regulatory effect is required for Salmonella invasiveness. Our work provides novel insights into mechanisms of sRNA regulation at bacterial mRNA 3' UTRs and adds to our knowledge of post-transcriptional regulation of the SPI-1 complex feed-forward loop. IMPORTANCE Salmonella enterica serovar Typhimurium is a prominent foodborne pathogen, infecting millions of people a year. To express virulence genes at the correct time and place in the host, Salmonella uses a complex regulatory network that senses environmental conditions. Known for their role in allowing quick responses to stress and virulence conditions, we investigated the role of small RNAs in facilitating precise expression of virulence genes. We found that the 3' untranslated region of the hilD mRNA, encoding a key virulence regulator, is a target for small RNAs and RNase E. The small RNAs stabilize hilD mRNA to allow proper expression of Salmonella virulence genes in the host.
Collapse
|
40
|
The Xenogeneic Silencer Histone-Like Nucleoid-Structuring Protein Mediates the Temperature and Salinity-Dependent Regulation of the Type III Secretion System 2 in Vibrio parahaemolyticus. J Bacteriol 2023; 205:e0026622. [PMID: 36468869 PMCID: PMC9879105 DOI: 10.1128/jb.00266-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The marine bacterium Vibrio parahaemolyticus is a major seafood-borne pathogen that causes acute diarrhea in humans. A crucial virulence determinant of V. parahaemolyticus is the type III secretion system 2 (T3SS2), which is encoded on the Vibrio parahaemolyticus pathogenicity island (Vp-PAI), in which gene expression is dependent on environmental cues, such as temperature and salinity. This characteristic may implicate the adaptation of V. parahaemolyticus from its natural habitat to the human body environment during infection; however, the underlying mechanism remains unknown. Here, we describe the regulatory role of the histone-like nucleoid-structuring protein (H-NS), which is a xenogeneic silencing protein, in T3SS2 gene expression through the conditional silencing of the gene encoding a master regulator of Vp-PAI, VtrB. The hns deletion canceled the temperature- and salinity-dependent differential T3SS2 gene expression. H-NS bound to the vtrB promoter containing AT-rich sequences, and the binding sites partially overlapped the binding sites of two positive regulators of vtrB (i.e., VtrA and ToxR), which may block the transcriptional activation of vtrB. H-NS-family proteins multimerize along the DNA strand, forming stiffened filament and/or bridging DNA duplexes for its target silencing. In V. parahaemolyticus, mutations at conserved residues that are required for the multimerization of H-NS abolished the repressive activity on VtrB expression, supporting the contention that H-NS multimerization is also critical for vtrB silencing in V. parahaemolyticus. Taken together, these findings demonstrate the principal role of H-NS as a thermal and salt switch with sensory and regulatory properties for ensuring T3SS2 gene regulation in V. parahaemolyticus. IMPORTANCE In the major seafood-borne pathogen Vibrio parahaemolyticus, the type III secretion system 2 (T3SS2) is a major virulence factor that is responsible for the enterotoxicity of this bacterium. The expression of T3SS2 varies according to changes in temperature and salinity, but the mechanism via which T3SS2 expression is regulated in response to such physical cues remains unknown. Here, we report that H-NS, a xenogeneic silencer that is widespread in Gram-negative bacteria, modulates the entirety of T3SS2 gene expression through the transcriptional silencing of the gene encoding the T3SS2 master regulator VtrB in a temperature- and salinity-dependent manner. Thus, our findings provide insights into how this pathogen achieves the appropriate control of the expression of virulence genes in the transition between aquatic and human environments.
Collapse
|
41
|
Abstract
The ability of bacteria to respond to changes in their environment is critical to their survival, allowing them to withstand stress, form complex communities, and induce virulence responses during host infection. A remarkable feature of many of these bacterial responses is that they are often variable across individual cells, despite occurring in an isogenic population exposed to a homogeneous environmental change, a phenomenon known as phenotypic heterogeneity. Phenotypic heterogeneity can enable bet-hedging or division of labor strategies that allow bacteria to survive fluctuating conditions. Investigating the significance of phenotypic heterogeneity in environmental transitions requires dynamic, single-cell data. Technical advances in quantitative single-cell measurements, imaging, and microfluidics have led to a surge of publications on this topic. Here, we review recent discoveries on single-cell bacterial responses to environmental transitions of various origins and complexities, from simple diauxic shifts to community behaviors in biofilm formation to virulence regulation during infection. We describe how these studies firmly establish that this form of heterogeneity is prevalent and a conserved mechanism by which bacteria cope with fluctuating conditions. We end with an outline of current challenges and future directions for the field. While it remains challenging to predict how an individual bacterium will respond to a given environmental input, we anticipate that capturing the dynamics of the process will begin to resolve this and facilitate rational perturbation of environmental responses for therapeutic and bioengineering purposes.
Collapse
|
42
|
Abstract
Bacterial populations can survive exposure to antibiotics through transient phenotypic and gene expression changes. These changes can be attributed to a small subpopulation of bacteria, giving rise to antibiotic persistence. Although this phenomenon has been known for decades, much remains to be learned about the mechanisms that drive persister formation. The RNA-binding protein ProQ has recently emerged as a global regulator of gene expression. Here, we show that ProQ impacts persister formation in Salmonella. In vitro, ProQ contributes to growth arrest in a subset of cells that are able to survive treatment at high concentrations of different antibiotics. The underlying mechanism for ProQ-dependent persister formation involves the activation of metabolically costly processes, including the flagellar pathway and the type III protein secretion system encoded on Salmonella pathogenicity island 2. Importantly, we show that the ProQ-dependent phenotype is relevant during macrophage infection and allows Salmonella to survive the combined action of host immune defenses and antibiotics. Together, our data highlight the importance of ProQ in Salmonella persistence and pathogenesis. IMPORTANCE Bacteria can avoid eradication by antibiotics through a phenomenon known as persistence. Persister cells arise through phenotypic heterogeneity and constitute a small fraction of dormant cells within a population of actively growing bacteria, which is susceptible to antibiotic killing. In this study, we show that ProQ, an RNA-binding protein and global regulator of gene expression, promotes persisters in the human pathogen Salmonella enterica serovar Typhimurium. Bacteria lacking the proQ gene outcompete wild-type bacteria under laboratory conditions, are less prone to enter growth dormancy, and form fewer persister cells. The basis for these phenotypes lies in ProQ's ability to activate energy-consuming cellular processes, including flagellar motility and protein secretion. Importantly, we show that ProQ contributes to the persister phenotype during Salmonella infection of macrophages, indicating an important role of this global regulator in Salmonella pathogenesis.
Collapse
|
43
|
Fernández-Fernández R, Olivenza DR, Sánchez-Romero MA. Identifying Bacterial Lineages in Salmonella by Flow Cytometry. EcoSal Plus 2022; 10:eESP00182021. [PMID: 35148202 PMCID: PMC10729938 DOI: 10.1128/ecosalplus.esp-0018-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 12/21/2021] [Indexed: 12/16/2022]
Abstract
Advances in technologies that permit high-resolution analysis of events in single cells have revealed that phenotypic heterogeneity is a widespread phenomenon in bacteria. Flow cytometry has the potential to describe the distribution of cellular properties within a population of bacterial cells and has yielded invaluable information about the ability of isogenic cells to diversify into phenotypic subpopulations. This review will discuss several single-cell approaches that have recently been applied to define phenotypic heterogeneity in populations of Salmonella enterica.
Collapse
Affiliation(s)
| | - David R. Olivenza
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| | | |
Collapse
|
44
|
Raad N, Tandon D, Hapfelmeier S, Polacek N. The stationary phase-specific sRNA FimR2 is a multifunctional regulator of bacterial motility, biofilm formation and virulence. Nucleic Acids Res 2022; 50:11858-11875. [PMID: 36354005 PMCID: PMC9723502 DOI: 10.1093/nar/gkac1025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 10/06/2022] [Accepted: 10/20/2022] [Indexed: 11/11/2022] Open
Abstract
Bacterial pathogens employ a plethora of virulence factors for host invasion, and their use is tightly regulated to maximize infection efficiency and manage resources in a nutrient-limited environment. Here we show that during Escherichia coli stationary phase the 3' UTR-derived small non-coding RNA FimR2 regulates fimbrial and flagellar biosynthesis at the post-transcriptional level, leading to biofilm formation as the dominant mode of survival under conditions of nutrient depletion. FimR2 interacts with the translational regulator CsrA, antagonizing its functions and firmly tightening control over motility and biofilm formation. Generated through RNase E cleavage, FimR2 regulates stationary phase biology by fine-tuning target mRNA levels independently of the chaperones Hfq and ProQ. The Salmonella enterica orthologue of FimR2 induces effector protein secretion by the type III secretion system and stimulates infection, thus linking the sRNA to virulence. This work reveals the importance of bacterial sRNAs in modulating various aspects of bacterial physiology including stationary phase and virulence.
Collapse
Affiliation(s)
- Nicole Raad
- Department of Chemistry, Biochemistry, and Pharmaceutical Sciences, University of Bern, Bern, Switzerland,Graduate School for Cellular and Biomedical Sciences, Bern, Switzerland
| | - Disha Tandon
- Graduate School for Cellular and Biomedical Sciences, Bern, Switzerland,Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | | | - Norbert Polacek
- To whom correspondence should be addressed. Tel: +41 31 684 43 20;
| |
Collapse
|
45
|
Wiradiputra MRD, Khuntayaporn P, Thirapanmethee K, Chomnawang MT. Toxin-Antitoxin Systems: A Key Role on Persister Formation in Salmonella enterica Serovar Typhimurium. Infect Drug Resist 2022; 15:5813-5829. [PMID: 36213766 PMCID: PMC9541301 DOI: 10.2147/idr.s378157] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 09/16/2022] [Indexed: 11/05/2022] Open
Abstract
The toxin and antitoxin modules in bacteria consist of a toxin molecule that has activity to inhibit various cellular processes and its cognate antitoxin that neutralizes the toxin. This system is considered taking part in the formation of persister cells, which are a subpopulation of recalcitrant cells able to survive antimicrobial treatment without any resistance mechanisms. Importantly, persisters have been associated with long-term infections and treatment failures in healthcare settings. It is a public health concern since persisters can be involved in the evolution and dissemination of antimicrobial resistance amidst the aggravating spread of multidrug-resistant bacteria and insufficient novel antimicrobial therapy to tackle this issue. Salmonella enterica serovar Typhimurium is one of the most prevalent Salmonella serotypes in the world and is a leading cause of food-borne salmonellosis. S. Typhimurium has been known to cause persistent infection and a wealth of investigations on Salmonella persisters indicates that toxin and antitoxin modules play a role in mediating the phenotypic switch of persisters, rendering its survival ability in the presence of antimicrobial agents. In this review, we discuss findings regarding mechanisms that underly persistence in S. Typhimurium, especially the involvement of toxin and antitoxin modules.
Collapse
Affiliation(s)
- Made Rai Dwitya Wiradiputra
- Antimicrobial Resistance Interdisciplinary Group (AmRIG), Faculty of Pharmacy, Mahidol University, Bangkok, Thailand,Biopharmaceutical Sciences Program, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Piyatip Khuntayaporn
- Antimicrobial Resistance Interdisciplinary Group (AmRIG), Faculty of Pharmacy, Mahidol University, Bangkok, Thailand,Department of Microbiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Krit Thirapanmethee
- Antimicrobial Resistance Interdisciplinary Group (AmRIG), Faculty of Pharmacy, Mahidol University, Bangkok, Thailand,Department of Microbiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Mullika Traidej Chomnawang
- Antimicrobial Resistance Interdisciplinary Group (AmRIG), Faculty of Pharmacy, Mahidol University, Bangkok, Thailand,Department of Microbiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand,Correspondence: Mullika Traidej Chomnawang, Department of Microbiology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand, Tel +66 2 644 8692, Email
| |
Collapse
|
46
|
Luneau JS, Baudin M, Quiroz Monnens T, Carrère S, Bouchez O, Jardinaud M, Gris C, François J, Ray J, Torralba B, Arlat M, Lewis JD, Lauber E, Deutschbauer AM, Noël LD, Boulanger A. Genome-wide identification of fitness determinants in the Xanthomonas campestris bacterial pathogen during early stages of plant infection. THE NEW PHYTOLOGIST 2022; 236:235-248. [PMID: 35706385 PMCID: PMC9543026 DOI: 10.1111/nph.18313] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/07/2022] [Indexed: 05/31/2023]
Abstract
Plant diseases are an important threat to food production. While major pathogenicity determinants required for disease have been extensively studied, less is known on how pathogens thrive during host colonization, especially at early infection stages. Here, we used randomly barcoded-transposon insertion site sequencing (RB-TnSeq) to perform a genome-wide screen and identify key bacterial fitness determinants of the vascular pathogen Xanthomonas campestris pv campestris (Xcc) during infection of the cauliflower host plant (Brassica oleracea). This high-throughput analysis was conducted in hydathodes, the natural entry site of Xcc, in xylem sap and in synthetic media. Xcc did not face a strong bottleneck during hydathode infection. In total, 181 genes important for fitness were identified in plant-associated environments with functional enrichment in genes involved in metabolism but only few genes previously known to be involved in virulence. The biological relevance of 12 genes was independently confirmed by phenotyping single mutants. Notably, we show that XC_3388, a protein with no known function (DUF1631), plays a key role in the adaptation and virulence of Xcc possibly through c-di-GMP-mediated regulation. This study revealed yet unsuspected social behaviors adopted by Xcc individuals when confined inside hydathodes at early infection stages.
Collapse
Affiliation(s)
- Julien S. Luneau
- LIPME, Université de Toulouse, INRAE, CNRSUniversité Paul Sabatier31320Castanet‐TolosanFrance
| | - Maël Baudin
- Plant Gene Expression Center, USDAAlbanyCA94710USA
- Department of Plant and Microbial BiologyUniversity of CaliforniaBerkeleyCA94720USA
| | - Thomas Quiroz Monnens
- LIPME, Université de Toulouse, INRAE, CNRSUniversité Paul Sabatier31320Castanet‐TolosanFrance
| | - Sébastien Carrère
- LIPME, Université de Toulouse, INRAE, CNRSUniversité Paul Sabatier31320Castanet‐TolosanFrance
| | - Olivier Bouchez
- Genotoul Genome & Transcriptome (GeT‐PlaGe), INRAE31320Castanet‐TolosanFrance
| | | | - Carine Gris
- LIPME, Université de Toulouse, INRAE, CNRSUniversité Paul Sabatier31320Castanet‐TolosanFrance
| | - Jonas François
- LIPME, Université de Toulouse, INRAE, CNRSUniversité Paul Sabatier31320Castanet‐TolosanFrance
| | - Jayashree Ray
- Environmental Genomics and Systems Biology DivisionLawrence Berkeley National LaboratoryBerkeleyCA94720USA
| | - Babil Torralba
- LIPME, Université de Toulouse, INRAE, CNRSUniversité Paul Sabatier31320Castanet‐TolosanFrance
| | - Matthieu Arlat
- LIPME, Université de Toulouse, INRAE, CNRSUniversité Paul Sabatier31320Castanet‐TolosanFrance
| | - Jennifer D. Lewis
- Plant Gene Expression Center, USDAAlbanyCA94710USA
- Department of Plant and Microbial BiologyUniversity of CaliforniaBerkeleyCA94720USA
| | - Emmanuelle Lauber
- LIPME, Université de Toulouse, INRAE, CNRSUniversité Paul Sabatier31320Castanet‐TolosanFrance
| | - Adam M. Deutschbauer
- Department of Plant and Microbial BiologyUniversity of CaliforniaBerkeleyCA94720USA
- Environmental Genomics and Systems Biology DivisionLawrence Berkeley National LaboratoryBerkeleyCA94720USA
| | - Laurent D. Noël
- LIPME, Université de Toulouse, INRAE, CNRSUniversité Paul Sabatier31320Castanet‐TolosanFrance
| | - Alice Boulanger
- LIPME, Université de Toulouse, INRAE, CNRSUniversité Paul Sabatier31320Castanet‐TolosanFrance
| |
Collapse
|
47
|
Elpers L, Deiwick J, Hensel M. Effect of Environmental Temperatures on Proteome Composition of Salmonella enterica Serovar Typhimurium. Mol Cell Proteomics 2022; 21:100265. [PMID: 35788066 PMCID: PMC9396072 DOI: 10.1016/j.mcpro.2022.100265] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 06/17/2022] [Accepted: 06/30/2022] [Indexed: 12/29/2022] Open
Abstract
Salmonella enterica serovar Typhimurium (STM) is a major cause of gastroenteritis and transmitted by consumption of contaminated food. STM is associated to food originating from animals (pork, chicken, eggs) or plants (vegetables, fruits, nuts, and herbs). Infection of warm-blooded mammalian hosts by STM and the underlying complex regulatory network of virulence gene expression depend on various environmental conditions encountered in hosts. However, less is known about the proteome and possible regulatory networks for gene expression of STM outside the preferred host. Nutritional limitations and changes in temperature are the most obvious stresses outside the native host. Thus, we analyzed the proteome profile of STM grown in rich medium (LB medium) or minimal medium (PCN medium) at temperatures ranging from 8 °C to 37 °C. LB medium mimics the nutritional rich environment inside the host, whereas minimal PCN medium represents nutritional limitations outside the host, found during growth of fresh produce (field conditions). Further, the range of temperatures analyzed reflects conditions within natural hosts (37 °C), room temperature (20 °C), during growth under agricultural conditions (16 °C and 12 °C), and during food storage (8 °C). Implications of altered nutrient availability and growth temperature on STM proteomes were analyzed by HPLC/MS-MS and label-free quantification. Our study provides first insights into the complex adaptation of STM to various environmental temperatures, which allows STM not only to infect mammalian hosts but also to enter new infection routes that have been poorly studied so far. With the present dataset, global virulence factors, their impact on infection routes, and potential anti-infective strategies can now be investigated in detail. Especially, we were able to demonstrate functional flagella at 12 °C growth temperature for STM with an altered motility behavior.
Collapse
Affiliation(s)
- Laura Elpers
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany
| | - Jörg Deiwick
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany
| | - Michael Hensel
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany; CellNanOs - Center of Cellular Nanoanalytics Osnabrück, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany.
| |
Collapse
|
48
|
Figueroa-Bossi N, Sánchez-Romero MA, Kerboriou P, Naquin D, Mendes C, Bouloc P, Casadesús J, Bossi L. Pervasive transcription enhances the accessibility of H-NS-silenced promoters and generates bistability in Salmonella virulence gene expression. Proc Natl Acad Sci U S A 2022; 119:e2203011119. [PMID: 35858437 PMCID: PMC9335307 DOI: 10.1073/pnas.2203011119] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/01/2022] [Indexed: 01/21/2023] Open
Abstract
In Escherichia coli and Salmonella, many genes silenced by the nucleoid structuring protein H-NS are activated upon inhibiting Rho-dependent transcription termination. This response is poorly understood and difficult to reconcile with the view that H-NS acts mainly by blocking transcription initiation. Here we have analyzed the basis for the up-regulation of H-NS-silenced Salmonella pathogenicity island 1 (SPI-1) in cells depleted of Rho-cofactor NusG. Evidence from genetic experiments, semiquantitative 5' rapid amplification of complementary DNA ends sequencing (5' RACE-Seq), and chromatin immunoprecipitation sequencing (ChIP-Seq) shows that transcription originating from spurious antisense promoters, when not stopped by Rho, elongates into a H-NS-bound regulatory region of SPI-1, displacing H-NS and rendering the DNA accessible to the master regulator HilD. In turn, HilD's ability to activate its own transcription triggers a positive feedback loop that results in transcriptional activation of the entire SPI-1. Significantly, single-cell analyses revealed that this mechanism is largely responsible for the coexistence of two subpopulations of cells that either express or do not express SPI-1 genes. We propose that cell-to-cell differences produced by stochastic spurious transcription, combined with feedback loops that perpetuate the activated state, can generate bimodal gene expression patterns in bacterial populations.
Collapse
Affiliation(s)
- Nara Figueroa-Bossi
- Université Paris-Saclay, CEA, CNRS, Institut de Biologie Intégrative de la Cellule (I2BC), 91190 Gif-sur-Yvette, France
| | - María Antonia Sánchez-Romero
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Patricia Kerboriou
- Université Paris-Saclay, CEA, CNRS, Institut de Biologie Intégrative de la Cellule (I2BC), 91190 Gif-sur-Yvette, France
| | - Delphine Naquin
- Université Paris-Saclay, CEA, CNRS, Institut de Biologie Intégrative de la Cellule (I2BC), 91190 Gif-sur-Yvette, France
| | - Clara Mendes
- Université Paris-Saclay, CEA, CNRS, Institut de Biologie Intégrative de la Cellule (I2BC), 91190 Gif-sur-Yvette, France
| | - Philippe Bouloc
- Université Paris-Saclay, CEA, CNRS, Institut de Biologie Intégrative de la Cellule (I2BC), 91190 Gif-sur-Yvette, France
| | - Josep Casadesús
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Lionello Bossi
- Université Paris-Saclay, CEA, CNRS, Institut de Biologie Intégrative de la Cellule (I2BC), 91190 Gif-sur-Yvette, France
| |
Collapse
|
49
|
Marro FC, Laurent F, Josse J, Blocker AJ. Methods to monitor bacterial growth and replicative rates at the single-cell level. FEMS Microbiol Rev 2022; 46:6623663. [PMID: 35772001 PMCID: PMC9629498 DOI: 10.1093/femsre/fuac030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 06/01/2022] [Accepted: 06/28/2022] [Indexed: 01/09/2023] Open
Abstract
The heterogeneity of bacterial growth and replicative rates within a population was proposed a century ago notably to explain the presence of bacterial persisters. The term "growth rate" at the single-cell level corresponds to the increase in size or mass of an individual bacterium while the "replicative rate" refers to its division capacity within a defined temporality. After a decades long hiatus, recent technical innovative approaches allow population growth and replicative rates heterogeneity monitoring at the single-cell level resuming in earnest. Among these techniques, the oldest and widely used is time-lapse microscopy, most recently combined with microfluidics. We also discuss recent fluorescence dilution methods informing only on replicative rates and best suited. Some new elegant single cell methods so far only sporadically used such as buoyant mass measurement and stable isotope probing have emerged. Overall, such tools are widely used to investigate and compare the growth and replicative rates of bacteria displaying drug-persistent behaviors to that of bacteria growing in specific ecological niches or collected from patients. In this review, we describe the current methods available, discussing both the type of queries these have been used to answer and the specific strengths and limitations of each method.
Collapse
Affiliation(s)
- Florian C Marro
- Evotec ID Lyon, In Vitro Biology, Infectious Diseases and Antibacterials Unit, Gerland, 69007 Lyon, France,CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
| | - Frédéric Laurent
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France,Institut des Sciences Pharmaceutiques et Biologiques (ISPB), Université Claude Bernard Lyon 1, Lyon, France,Centre de Référence pour la prise en charge des Infections ostéo-articulaires complexes (CRIOAc Lyon; www.crioac-lyon.fr), Hospices Civils de Lyon, Lyon, France,Laboratoire de bactériologie, Institut des Agents Infectieux, French National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France
| | - Jérôme Josse
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France,Institut des Sciences Pharmaceutiques et Biologiques (ISPB), Université Claude Bernard Lyon 1, Lyon, France,Centre de Référence pour la prise en charge des Infections ostéo-articulaires complexes (CRIOAc Lyon; www.crioac-lyon.fr), Hospices Civils de Lyon, Lyon, France
| | - Ariel J Blocker
- Corresponding author. Evotec ID Lyon, In Vitro Biology, Infectious Diseases and Antibacterials Unit, France. E-mail:
| |
Collapse
|
50
|
Mitchell MK, Ellermann M. Long Chain Fatty Acids and Virulence Repression in Intestinal Bacterial Pathogens. Front Cell Infect Microbiol 2022; 12:928503. [PMID: 35782143 PMCID: PMC9247172 DOI: 10.3389/fcimb.2022.928503] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
When bacterial pathogens enter the gut, they encounter a complex milieu of signaling molecules and metabolites produced by host and microbial cells or derived from external sources such as the diet. This metabolomic landscape varies throughout the gut, thus establishing a biogeographical gradient of signals that may be sensed by pathogens and resident bacteria alike. Enteric bacterial pathogens have evolved elaborate mechanisms to appropriately regulate their virulence programs, which involves sensing and responding to many of these gut metabolites to facilitate successful gut colonization. Long chain fatty acids (LCFAs) represent major constituents of the gut metabolome that can impact bacterial functions. LCFAs serve as important nutrient sources for all cellular organisms and can function as signaling molecules that regulate bacterial metabolism, physiology, and behaviors. Moreover, in several enteric pathogens, including Salmonella enterica, Listeria monocytogenes, Vibrio cholerae, and enterohemorrhagic Escherichia coli, LCFA sensing results in the transcriptional repression of virulence through two general mechanisms. First, some LCFAs function as allosteric inhibitors that decrease the DNA binding affinities of transcriptional activators of virulence genes. Second, some LCFAs also modulate the activation of histidine kinase receptors, which alters downstream intracellular signaling networks to repress virulence. This mini-review will summarize recent studies that have investigated the molecular mechanisms by which different LCFA derivatives modulate the virulence of enteric pathogens, while also highlighting important gaps in the field regarding the roles of LCFAs as determinants of infection and disease.
Collapse
|