1
|
Listeria exploits IFITM3 to suppress antibacterial activity in phagocytes. Nat Commun 2021; 12:4999. [PMID: 34404769 PMCID: PMC8371165 DOI: 10.1038/s41467-021-24982-0] [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: 04/22/2021] [Accepted: 07/18/2021] [Indexed: 12/20/2022] Open
Abstract
The type I interferon (IFN) signaling pathway has important functions in resistance to viral infection, with the downstream induction of interferon stimulated genes (ISG) protecting the host from virus entry, replication and spread. Listeria monocytogenes (Lm), a facultative intracellular foodborne pathogen, can exploit the type I IFN response as part of their pathogenic strategy, but the molecular mechanisms involved remain unclear. Here we show that type I IFN suppresses the antibacterial activity of phagocytes to promote systemic Lm infection. Mechanistically, type I IFN suppresses phagosome maturation and proteolysis of Lm virulence factors ActA and LLO, thereby promoting phagosome escape and cell-to-cell spread; the antiviral protein, IFN-induced transmembrane protein 3 (IFITM3), is required for this type I IFN-mediated alteration. Ifitm3-/- mice are resistant to systemic infection by Lm, displaying decreased bacterial spread in tissues, and increased immune cell recruitment and pro-inflammatory cytokine signaling. Together, our findings show how an antiviral mechanism in phagocytes can be exploited by bacterial pathogens, and implicate IFITM3 as a potential antimicrobial therapeutic target.
Collapse
|
2
|
Li M, Carpenter CE, Broadbent JR. Organic Acid Exposure Enhances Virulence in Some Listeria monocytogenes Strains Using the Galleria mellonella Infection Model. Front Microbiol 2021; 12:675241. [PMID: 34295317 PMCID: PMC8290484 DOI: 10.3389/fmicb.2021.675241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/14/2021] [Indexed: 01/01/2023] Open
Abstract
Prior research has suggested that the use of organic acids in the food industry may unintentionally enhance pathogenicity of Listeria monocytogenes strain N1-227 and R2-499. This study explored the connection between habituation to L-lactic acid or acetic acid and virulence in L. monocytogenes strains N1-227 and R2-499 using selected gene expression analysis and the in vivo Galleria mellonella wax worm model for infection. Expression of transcription factors (sigB and prfA) and genes related to acid resistance (gadD2, gadD3, and arcA) and bile resistance (bsh and bilE) or to virulence (inlA, inlB, hly, plcA, plcB, uhpT, and actA) was investigated by quantitative real-time PCR (qRT-PCR), while in vivo virulence was assessed by following the lethal time to 50% population mortality (LT50) of G. mellonella larvae after injection of untreated and habituated L. monocytogenes. Twenty minutes of habituation to the organic acids at pH 6.0 significantly increased expression of key acid and bile stress response genes in both strains, while expression of virulence genes was strain-dependent. The expression of transcription factor sigB was strain-dependent and there was no significant change in the expression of transcription factor prfA in both strains. Habituation to acid increased virulence of both strains as evidenced by decreased LT50 of G. mellonella larvae injected with Listeria habituated to either acid. In summary, habituation of both L. monocytogenes strains to organic acids up-regulated expression of several stress and virulence genes and concurrently increased virulence as measured using the G. mellonella model.
Collapse
Affiliation(s)
- Minghao Li
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT, United States
| | - Charles E Carpenter
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT, United States
| | - Jeff R Broadbent
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT, United States
| |
Collapse
|
3
|
Ma J, Ji Q, Wang S, Qiu J, Liu Q. Identification and evaluation of a panel of strong constitutive promoters in Listeria monocytogenes for improving the expression of foreign antigens. Appl Microbiol Biotechnol 2021; 105:5135-5145. [PMID: 34086117 PMCID: PMC8175932 DOI: 10.1007/s00253-021-11374-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/10/2021] [Accepted: 05/27/2021] [Indexed: 12/19/2022]
Abstract
Attenuated Listeria monocytogenes could be a potential vaccine vector for the immunotherapy of tumors or pathogens. However, the lack of reliable promoters has limited its ability to express foreign antigens. In the present study, 21 promoters were identified from Listeria monocytogenes through RNA-seq analysis under two pH conditions of pH 7.4 and pH 5.5. Based on the constructed fluorescence report system, 7 constitutive promoters exhibited higher strength than Phelp (1.8-fold to 5.4-fold), a previously reported strong promoter. Furthermore, the selected 5 constitutive promoters exhibited higher UreB production activity than Phelp (1.1-fold to 8.3-fold). Notably, a well-characterized constitutive promoter P18 was found with the highest activity of fluorescence intensity and UreB production. In summary, the study provides a panel of strong constitutive promoters for Listeria monocytogenes and offers a theoretical basis for mining constitutive promoters in other organisms. KEY POINTS: • Twenty-one promoters were identified from L. monocytogenes through RNA-seq. • Fluorescent tracer of L. monocytogenes (P18) was performed in vitro and in vivo. • A well-characterized constitutive promoter P18 could improve the expression level of a foreign antigen UreB in L. monocytogenes.
Collapse
Affiliation(s)
- Junfei Ma
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Qianyu Ji
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Shuying Wang
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jingxuan Qiu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Qing Liu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China. .,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China.
| |
Collapse
|
4
|
Characterization of the roles of activated charcoal and Chelex in the induction of PrfA regulon expression in complex medium. PLoS One 2021; 16:e0250989. [PMID: 33914817 PMCID: PMC8084165 DOI: 10.1371/journal.pone.0250989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 04/16/2021] [Indexed: 11/21/2022] Open
Abstract
The foodborne pathogen Listeria monocytogenes is able to survive across a wide range of intra- and extra-host environments by appropriately modulating gene expression patterns in response to different stimuli. Positive Regulatory Factor A (PrfA) is the major transcriptional regulator of virulence gene expression in L. monocytogenes. It has long been known that activated charcoal is required to induce the expression of PrfA-regulated genes in complex media, such as Brain Heart Infusion (BHI), but not in chemically defined media. In this study, we show that the expression of the PrfA-regulated hly, which encodes listeriolysin O, is induced 5- and 8-fold in L. monocytogenes cells grown in Chelex-treated BHI (Ch-BHI) and in the presence of activated charcoal (AC-BHI), respectively, relative to cells grown in BHI medium. Specifically, we show that metal ions present in BHI broth plays a role in the reduced expression of the PrfA regulon. In addition, we show that expression of hly is induced when the levels of bioavailable extra- or intercellular iron are reduced. L. monocytogenes cells grown Ch-BHI and AC-BHI media showed similar levels of resistance to the iron-activated antibiotic, streptonigrin, indicating that activated charcoal reduces the intracellular labile iron pool. Metal depletion and exogenously added glutathione contributed synergistically to PrfA-regulated gene expression since glutathione further increased hly expression in metal-depleted BHI but not in BHI medium. Analyses of transcriptional reporter fusion expression patterns revealed that genes in the PrfA regulon are differentially expressed in response to metal depletion, metal excess and exogenous glutathione. Our results suggest that metal ion abundance plays a role in modulating expression of PrfA-regulated virulence genes in L. monocytogenes.
Collapse
|
5
|
La Pietra L, Hudel M, Pillich H, Abu Mraheil M, Berisha B, Aden S, Hodnik V, Lochnit G, Rafiq A, Perniss A, Anderluh G, Chakraborty T. Phosphocholine Antagonizes Listeriolysin O-Induced Host Cell Responses of Listeria monocytogenes. J Infect Dis 2020; 222:1505-1516. [DOI: 10.1093/infdis/jiaa022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/20/2020] [Indexed: 12/20/2022] Open
Abstract
Abstract
Background
Bacterial toxins disrupt plasma membrane integrity with multitudinous effects on host cells. The secreted pore-forming toxin listeriolysin O (LLO) of the intracellular pathogen Listeria monocytogenes promotes egress of the bacteria from vacuolar compartments into the host cytosol often without overt destruction of the infected cell. Intracellular LLO activity is tightly controlled by host factors including compartmental pH, redox, proteolytic, and proteostatic factors, and inhibited by cholesterol.
Methods
Combining infection studies of L. monocytogenes wild type and isogenic mutants together with biochemical studies with purified phospholipases, we investigate the effect of their enzymatic activities on LLO.
Results
Here, we show that phosphocholine (ChoP), a reaction product of the phosphatidylcholine-specific phospholipase C (PC-PLC) of L. monocytogenes, is a potent inhibitor of intra- and extracellular LLO activities. Binding of ChoP to LLO is redox-independent and leads to the inhibition of LLO-dependent induction of calcium flux, mitochondrial damage, and apoptosis. ChoP also inhibits the hemolytic activities of the related cholesterol-dependent cytolysins (CDC), pneumolysin and streptolysin.
Conclusions
Our study uncovers a strategy used by L. monocytogenes to modulate cytotoxic LLO activity through the enzymatic activity of its PC-PLC. This mechanism appears to be widespread and also used by other CDC pore-forming toxin-producing bacteria.
Collapse
Affiliation(s)
- Luigi La Pietra
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Martina Hudel
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Helena Pillich
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Mobarak Abu Mraheil
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Besim Berisha
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| | - Saša Aden
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Vesna Hodnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Günter Lochnit
- Protein Analytics, Institute of Biochemistry, Justus-Liebig University Giessen, Giessen, Germany
| | - Amir Rafiq
- Institute of Anatomy and Cell Biology, Cardiopulmonary Institute, German Center for Lung Research, Justus-Liebig University Giessen, Giessen, Germany
| | - Alexander Perniss
- Institute of Anatomy and Cell Biology, Cardiopulmonary Institute, German Center for Lung Research, Justus-Liebig University Giessen, Giessen, Germany
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Trinad Chakraborty
- Institute of Medical Microbiology, German Center for Infection Research, Partner site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Giessen, Germany
| |
Collapse
|
6
|
Cross Talk between SigB and PrfA in Listeria monocytogenes Facilitates Transitions between Extra- and Intracellular Environments. Microbiol Mol Biol Rev 2019; 83:83/4/e00034-19. [PMID: 31484692 DOI: 10.1128/mmbr.00034-19] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The foodborne pathogen Listeria monocytogenes can modulate its transcriptome and proteome to ensure its survival during transmission through vastly differing environmental conditions. While L. monocytogenes utilizes a large array of regulators to achieve survival and growth in different intra- and extrahost environments, the alternative sigma factor σB and the transcriptional activator of virulence genes protein PrfA are two key transcriptional regulators essential for responding to environmental stress conditions and for host infection. Importantly, emerging evidence suggests that the shift from extrahost environments to the host gastrointestinal tract and, subsequently, to intracellular environments requires regulatory interplay between σB and PrfA at transcriptional, posttranscriptional, and protein activity levels. Here, we review the current evidence for cross talk and interplay between σB and PrfA and their respective regulons and highlight the plasticity of σB and PrfA cross talk and the role of this cross talk in facilitating successful transition of L. monocytogenes from diverse extrahost to diverse extra- and intracellular host environments.
Collapse
|
7
|
Stable Expression of Modified Green Fluorescent Protein in Group B Streptococci To Enable Visualization in Experimental Systems. Appl Environ Microbiol 2018; 84:AEM.01262-18. [PMID: 30006391 DOI: 10.1128/aem.01262-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 07/04/2018] [Indexed: 12/17/2022] Open
Abstract
Group B streptococcus (GBS) is a Gram-positive bacterium associated with various diseases in humans and animals. Many studies have examined GBS physiology, virulence, and microbe-host interactions using diverse imaging approaches, including fluorescence microscopy. Strategies to label and visualize GBS using fluorescence biomarkers have been limited to antibody-based methods or nonspecific stains that bind DNA or protein; an effective plasmid-based system to label GBS with a fluorescence biomarker would represent a useful visualization tool. In this study, we developed and validated a green fluorescent protein (GFP)-variant-expressing plasmid, pGU2664, which can be applied as a marker to visualize GBS in experimental studies. The synthetic constitutively active CP25 promoter drives strong and stable expression of the GFPmut3 biomarker in GBS strains carrying pGU2664. GBS maintains GFPmut3 activity at different phases of growth. The application of fluorescence polarization enables easy discrimination of GBS GFPmut3 activity from the autofluorescence of culture media commonly used to grow GBS. Differential interference contrast microscopy, in combination with epifluorescence microscopy to detect GFPmut3 in GBS, enabled visualization of bacterial attachment to live human epithelial cells in real time. Plasmid pGU2664 was also used to visualize phenotypic differences in the adherence of wild-type GBS and an isogenic gene-deficient mutant strain lacking CovR (the control of virulence regulator) in adhesion assays. The system for GFPmut3 expression in GBS described in this study provides a new tool for the visualization of this organism in diverse research applications. We discuss the advantages and consider the limitations of this fluorescent biomarker system developed for GBS.IMPORTANCE Group B streptococcus (GBS) is a bacterium associated with various diseases in humans and animals. This study describes the development of a strategy to label and visualize GBS using a fluorescence biomarker, termed GFPmut3. We show that this biomarker can be successfully applied to track the growth of bacteria in liquid medium, and it enables the detailed visualization of GBS in the context of live human cells in real-time microscopic analysis. The system for GFPmut3 expression in GBS described in this study provides a new tool for the visualization of this organism in diverse research applications.
Collapse
|
8
|
Secretion Chaperones PrsA2 and HtrA Are Required for Listeria monocytogenes Replication following Intracellular Induction of Virulence Factor Secretion. Infect Immun 2016; 84:3034-46. [PMID: 27481256 DOI: 10.1128/iai.00312-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 07/27/2016] [Indexed: 12/25/2022] Open
Abstract
The Gram-positive bacterium Listeria monocytogenes transitions from an environmental organism to an intracellular pathogen following its ingestion by susceptible mammalian hosts. Bacterial replication within the cytosol of infected cells requires activation of the central virulence regulator PrfA followed by a PrfA-dependent induction of secreted virulence factors. The PrfA-induced secreted chaperone PrsA2 and the chaperone/protease HtrA contribute to the folding and stability of select proteins translocated across the bacterial membrane. L. monocytogenes strains that lack both prsA2 and htrA exhibit near-normal patterns of growth in broth culture but are severely attenuated in vivo We hypothesized that, in the absence of PrsA2 and HtrA, the increase in PrfA-dependent protein secretion that occurs following bacterial entry into the cytosol results in misfolded proteins accumulating at the bacterial membrane with a subsequent reduction in intracellular bacterial viability. Consistent with this hypothesis, the introduction of a constitutively activated allele of prfA (prfA*) into ΔprsA2 ΔhtrA strains was found to essentially inhibit bacterial growth at 37°C in broth culture. ΔprsA2 ΔhtrA strains were additionally found to be defective for cell invasion and vacuole escape in selected cell types, steps that precede full PrfA activation. These data establish the essential requirement for PrsA2 and HtrA in maintaining bacterial growth under conditions of PrfA activation. In addition, chaperone function is required for efficient bacterial invasion and rapid vacuole lysis within select host cell types, indicating roles for PrsA2/HtrA prior to cytosolic PrfA activation and the subsequent induction of virulence factor secretion.
Collapse
|
9
|
Teh BS, Apel J, Shao Y, Boland W. Colonization of the Intestinal Tract of the Polyphagous Pest Spodoptera littoralis with the GFP-Tagged Indigenous Gut Bacterium Enterococcus mundtii. Front Microbiol 2016; 7:928. [PMID: 27379058 PMCID: PMC4906056 DOI: 10.3389/fmicb.2016.00928] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 05/31/2016] [Indexed: 11/16/2022] Open
Abstract
The alkaline gut of Lepidopterans plays a crucial role in shaping communities of bacteria. Enterococcus mundtii has emerged as one of the predominant gut microorganisms in the gastrointestinal tract of the major agricultural pest, Spodoptera littoralis. Therefore, it was selected as a model bacterium to study its adaptation to harsh alkaline gut conditions in its host insect throughout different stages of development (larvae, pupae, adults, and eggs). To date, the mechanism of bacterial survival in insects' intestinal tract has been unknown. Therefore, we have engineered a GFP-tagged species of bacteria, E. mundtii, to track how it colonizes the intestine of S. littoralis. Three promoters of different strengths were used to control the expression of GFP in E. mundtii. The promoter ermB was the most effective, exhibiting the highest GFP fluorescence intensity, and hence was chosen as our main construct. Our data show that the engineered fluorescent bacteria survived and proliferated in the intestinal tract of the insect at all life stages for up to the second generation following ingestion.
Collapse
Affiliation(s)
- Beng-Soon Teh
- Department of Bioorganic Chemistry, Max Planck Institute for Chemical Ecology Jena, Germany
| | - Johanna Apel
- Clinic for Internal Medicine II, Department of Haematology and Medical Oncology University Hospital Jena, Germany
| | - Yongqi Shao
- Laboratory of Invertebrate Pathology, College of Animal Sciences, Zhejiang University Hangzhou, China
| | - Wilhelm Boland
- Department of Bioorganic Chemistry, Max Planck Institute for Chemical Ecology Jena, Germany
| |
Collapse
|
10
|
Perforin-2 Protects Host Cells and Mice by Restricting the Vacuole to Cytosol Transitioning of a Bacterial Pathogen. Infect Immun 2016; 84:1083-1091. [PMID: 26831467 DOI: 10.1128/iai.01434-15] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 01/23/2016] [Indexed: 12/20/2022] Open
Abstract
The host-encoded Perforin-2 (encoded by the macrophage-expressed gene 1, Mpeg1), which possesses a pore-forming MACPF domain, reduces the viability of bacterial pathogens that reside within membrane-bound compartments. Here, it is shown that Perforin-2 also restricts the proliferation of the intracytosolic pathogen Listeria monocytogenes Within a few hours of systemic infection, the massive proliferation of L. monocytogenes in Perforin-2(-/-)mice leads to a rapid appearance of acute disease symptoms. We go on to show in cultured Perforin-2(-/-)cells that the vacuole-to-cytosol transitioning of L. monocytogenesis greatly accelerated. Unexpectedly, we found that in Perforin-2(-/-)macrophages,Listeria-containing vacuoles quickly (≤ 15 min) acidify, and that this was coincident with greater virulence gene expression, likely accounting for the more rapid translocation of L. monocytogenes to its replicative niche in the cytosol. This hypothesis was supported by our finding that aL. monocytogenes strain expressing virulence factors at a constitutively high level replicated equally well in Perforin-2(+/+)and Perforin-2(-/-)macrophages. Our findings suggest that the protective role of Perforin-2 against listeriosis is based on it limiting the intracellular replication of the pathogen. This cellular activity of Perforin-2 may derive from it regulating the acidification of Listeria-containing vacuoles, thereby depriving the pathogen of favorable intracellular conditions that promote its virulence gene activity.
Collapse
|
11
|
Spears PA, Havell EA, Hamrick TS, Goforth JB, Levine AL, Abraham ST, Heiss C, Azadi P, Orndorff PE. Listeria monocytogenes wall teichoic acid decoration in virulence and cell-to-cell spread. Mol Microbiol 2016; 101:714-30. [PMID: 26871418 DOI: 10.1111/mmi.13353] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2016] [Indexed: 12/11/2022]
Abstract
Wall teichoic acid (WTA) comprises a class of glycopolymers covalently attached to the peptidoglycan of gram positive bacteria. In Listeria monocytogenes, mutations that prevent addition of certain WTA decorating sugars are attenuating. However, the steps required for decoration and the pathogenic process interrupted are not well described. We systematically examined the requirement for WTA galactosylation in a mouse oral-virulent strain by first creating mutations in four genes whose products conferred resistance to a WTA-binding bacteriophage. WTA biochemical and structural studies indicated that galactosylated WTA was directly required for bacteriophage adsorption and that mutant WTA lacked appreciable galactose in all except one mutant - which retained a level ca. 7% of the parent. All mutants were profoundly attenuated in orally infected mice and were impaired in cell-to-cell spread in vitro. Confocal microscopy of cytosolic mutants revealed that all expressed ActA on their cell surface and formed actin tails with a frequency similar to the parent. However, the mutant tails were significantly shorter - suggesting a defect in actin based motility. Roles for the gene products in WTA galactosylation are proposed. Identification and interruption of WTA decoration pathways may provide a general strategy to discover non-antibiotic therapeutics for gram positive infections. © 2016 John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- Patricia A Spears
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27606, USA
| | - Edward A Havell
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27606, USA
| | - Terri S Hamrick
- Department of Microbiology and Immunology, School of Osteopathic Medicine, Campbell University, Buies Creek, NC, 27506, USA
| | - John B Goforth
- Department of Microbiology and Immunology, School of Osteopathic Medicine, Campbell University, Buies Creek, NC, 27506, USA
| | - Alexandra L Levine
- Department of Microbiology and Immunology, School of Osteopathic Medicine, Campbell University, Buies Creek, NC, 27506, USA
| | - S Thomas Abraham
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Campbell University, Buies Creek, NC, 27506, USA
| | - Christian Heiss
- Complex Carbohydrate Research Center, The University of Georgia, Athens, GA, 30602, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, The University of Georgia, Athens, GA, 30602, USA
| | - Paul E Orndorff
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27606, USA
| |
Collapse
|
12
|
Cemma M, Lam GY, Stöckli M, Higgins DE, Brumell JH. Strain-Specific Interactions of Listeria monocytogenes with the Autophagy System in Host Cells. PLoS One 2015; 10:e0125856. [PMID: 25970638 PMCID: PMC4430529 DOI: 10.1371/journal.pone.0125856] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 03/26/2015] [Indexed: 12/22/2022] Open
Abstract
Listeria monocytogenes is an intracellular bacterial pathogen that can replicate in the cytosol of host cells. These bacteria undergo actin-based motility in the cytosol via expression of ActA, which recruits host actin-regulatory proteins to the bacterial surface. L. monocytogenes is thought to evade killing by autophagy using ActA-dependent mechanisms. ActA-independent mechanisms of autophagy evasion have also been proposed, but remain poorly understood. Here we examined autophagy of non-motile (ΔactA) mutants of L. monocytogenes strains 10403S and EGD-e, two commonly studied strains of this pathogen. The ΔactA mutants displayed accumulation of ubiquitinated proteins and p62/SQSTM1 on their surface. However, only strain EGD-e ΔactA displayed colocalization with the autophagy marker LC3 at 8 hours post infection. A bacteriostatic agent (chloramphenicol) was required for LC3 recruitment to 10403S ΔactA, suggesting that these bacteria produce a factor for autophagy evasion. Internalin K was proposed to block autophagy of L. monocytogenes in the cytosol of host cells. However, deletion of inlK in either the wild-type or ΔactA background of strain 10403S had no impact on autophagy evasion by bacteria, indicating it does not play an essential role in evading autophagy. Replication of ΔactA mutants of strain EGD-e and 10403S was comparable to their parent wild-type strain in macrophages. Thus, ΔactA mutants of L. monocytogenes can block killing by autophagy at a step downstream of protein ubiquitination and, in the case of strain EGD-e, downstream of LC3 recruitment to bacteria. Our findings highlight the strain-specific differences in the mechanisms that L. monocytogenes uses to evade killing by autophagy in host cells.
Collapse
Affiliation(s)
- Marija Cemma
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Grace Y. Lam
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Martina Stöckli
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02115, United States of America
| | - Darren E. Higgins
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02115, United States of America
| | - John H. Brumell
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
- * E-mail:
| |
Collapse
|
13
|
|
14
|
Carvalho F, Sousa S, Cabanes D. How Listeria monocytogenes organizes its surface for virulence. Front Cell Infect Microbiol 2014; 4:48. [PMID: 24809022 PMCID: PMC4010754 DOI: 10.3389/fcimb.2014.00048] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/02/2014] [Indexed: 02/04/2023] Open
Abstract
Listeria monocytogenes is a Gram-positive pathogen responsible for the manifestation of human listeriosis, an opportunistic foodborne disease with an associated high mortality rate. The key to the pathogenesis of listeriosis is the capacity of this bacterium to trigger its internalization by non-phagocytic cells and to survive and even replicate within phagocytes. The arsenal of virulence proteins deployed by L. monocytogenes to successfully promote the invasion and infection of host cells has been progressively unveiled over the past decades. A large majority of them is located at the cell envelope, which provides an interface for the establishment of close interactions between these bacterial factors and their host targets. Along the multistep pathways carrying these virulence proteins from the inner side of the cytoplasmic membrane to their cell envelope destination, a multiplicity of auxiliary proteins must act on the immature polypeptides to ensure that they not only maturate into fully functional effectors but also are placed or guided to their correct position in the bacterial surface. As the major scaffold for surface proteins, the cell wall and its metabolism are critical elements in listerial virulence. Conversely, the crucial physical support and protection provided by this structure make it an ideal target for the host immune system. Therefore, mechanisms involving fine modifications of cell envelope components are activated by L. monocytogenes to render it less recognizable by the innate immunity sensors or more resistant to the activity of antimicrobial effectors. This review provides a state-of-the-art compilation of the mechanisms used by L. monocytogenes to organize its surface for virulence, with special focus on those proteins that work “behind the frontline”, either supporting virulence effectors or ensuring the survival of the bacterium within its host.
Collapse
Affiliation(s)
- Filipe Carvalho
- Group of Molecular Microbiology, Unit of Infection and Immunity, Instituto de Biologia Molecular e Celular, University of Porto Porto, Portugal
| | - Sandra Sousa
- Group of Molecular Microbiology, Unit of Infection and Immunity, Instituto de Biologia Molecular e Celular, University of Porto Porto, Portugal
| | - Didier Cabanes
- Group of Molecular Microbiology, Unit of Infection and Immunity, Instituto de Biologia Molecular e Celular, University of Porto Porto, Portugal
| |
Collapse
|
15
|
Deshayes C, Bielecka MK, Cain RJ, Scortti M, de las Heras A, Pietras Z, Luisi BF, Núñez Miguel R, Vázquez-Boland JA. Allosteric mutants show that PrfA activation is dispensable for vacuole escape but required for efficient spread and Listeria survival in vivo. Mol Microbiol 2012; 85:461-77. [PMID: 22646689 PMCID: PMC3443378 DOI: 10.1111/j.1365-2958.2012.08121.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The transcriptional regulator PrfA controls key virulence determinants of the facultative intracellular pathogen Listeria monocytogenes. PrfA-dependent gene expression is strongly induced within host cells. While the basis of this activation is unknown, the structural homology of PrfA with the cAMP receptor protein (Crp) and the finding of constitutively activated PrfA* mutants suggests it may involve ligand-induced allostery. Here, we report the identification of a solvent-accessible cavity within the PrfA N-terminal domain that may accommodate an activating ligand. The pocket occupies a similar position to the cAMP binding site in Crp but lacks the cyclic nucleotide-anchoring motif and has its entrance on the opposite side of the β-barrel. Site-directed mutations in this pocket impaired intracellular PrfA-dependent gene activation without causing extensive structural/functional alterations to PrfA. Two substitutions, L48F and Y63W, almost completely abolished intracellular virulence gene induction and thus displayed the expected phenotype for allosteric activation-deficient PrfA mutations. Neither PrfA(allo) substitution affected vacuole escape and initial intracellular growth of L. monocytogenes in epithelial cells and macrophages but caused defective cell-to-cell spread and strong attenuation in mice. Our data support the hypothesis that PrfA is allosterically activated during intracellular infection and identify the probable binding site for the effector ligand. They also indicate that PrfA allosteric activation is not required for early intracellular survival but is essential for full Listeria virulence and colonization of host tissues.
Collapse
Affiliation(s)
- Caroline Deshayes
- Centres for Infectious Diseases and Immunity, Infection & Evolution, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Lam GY, Czuczman MA, Higgins DE, Brumell JH. Interactions of Listeria monocytogenes with the Autophagy System of Host Cells. Adv Immunol 2012; 113:7-18. [DOI: 10.1016/b978-0-12-394590-7.00008-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
Proteomic expression profiles of virulent and avirulent strains of Listeria monocytogenes isolated from macrophages. J Proteomics 2011; 74:1906-17. [DOI: 10.1016/j.jprot.2011.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 04/29/2011] [Accepted: 05/02/2011] [Indexed: 11/20/2022]
|
18
|
Invasive extravillous trophoblasts restrict intracellular growth and spread of Listeria monocytogenes. PLoS Pathog 2011; 7:e1002005. [PMID: 21408203 PMCID: PMC3048367 DOI: 10.1371/journal.ppat.1002005] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 12/29/2010] [Indexed: 11/19/2022] Open
Abstract
Listeria monocytogenes is a facultative intracellular bacterial pathogen that can infect the placenta, a chimeric organ made of maternal and fetal cells. Extravillous trophoblasts (EVT) are specialized fetal cells that invade the uterine implantation site, where they come into direct contact with maternal cells. We have shown previously that EVT are the preferred site of initial placental infection. In this report, we infected primary human EVT with L. monocytogenes. EVT eliminated ∼80% of intracellular bacteria over 24-hours. Bacteria were unable to escape into the cytoplasm and remained confined to vacuolar compartments that became acidified and co-localized with LAMP1, consistent with bacterial degradation in lysosomes. In human placental organ cultures bacterial vacuolar escape rates differed between specific trophoblast subpopulations. The most invasive EVT—those that would be in direct contact with maternal cells in vivo—had lower escape rates than trophoblasts that were surrounded by fetal cells and tissues. Our results suggest that EVT present a bottleneck in the spread of L. monocytogenes from mother to fetus by inhibiting vacuolar escape, and thus intracellular bacterial growth. However, if L. monocytogenes is able to spread beyond EVT it can find a more hospitable environment. Our results elucidate a novel aspect of the maternal-fetal barrier. Infection of the placenta and fetus is an important cause of pregnancy complications and fetal and neonatal morbidity and mortality. Listeria monocytogenes is an intracellular bacterial pathogen that causes pregnancy-related infections in humans. The pathogenesis of listeriosis during pregnancy is poorly understood. We have previously shown that transmission of L. monocytogenes from maternal cells and tissues to fetal cells occurs in the uterine implantation site, and that a small subpopulation of specialized fetal cells called extravillous trophoblasts are the preferred initial site of infection. Here we use primary human placental organ and cell culture systems to characterize the intracellular fate of L. monocytogenes in extravillous trophoblasts. We found that these cells entrap bacteria in vacuolar compartments where they are degraded and therefore reduce bacterial dissemination into deeper structures of the placenta. Our study provides new insights into the nature of the maternal-fetal barrier. Extravillous trophoblasts that are accessible to infection with intracellular pathogens from infected maternal cells have host defense mechanisms that constitute a bottleneck in maternal-fetal transmission.
Collapse
|
19
|
Cortes-Bratti X, Bassères E, Herrera-Rodriguez F, Botero-Kleiven S, Coppotelli G, Andersen JB, Masucci MG, Holmgren A, Chaves-Olarte E, Frisan T, Avila-Cariño J. Thioredoxin 80-activated-monocytes (TAMs) inhibit the replication of intracellular pathogens. PLoS One 2011; 6:e16960. [PMID: 21365006 PMCID: PMC3041819 DOI: 10.1371/journal.pone.0016960] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 01/09/2011] [Indexed: 12/18/2022] Open
Abstract
Background Thioredoxin 80 (Trx80) is an 80 amino acid natural cleavage product of Trx, produced primarily by monocytes. Trx80 induces differentiation of human monocytes into a novel cell type, named Trx80-activated-monocytes (TAMs). Principal Findings In this investigation we present evidence for a role of TAMs in the control of intracellular bacterial infections. As model pathogens we have chosen Listeria monocytogenes and Brucella abortus which replicate in the cytosol and the endoplasmic reticulum respectively. Our data indicate that TAMs efficiently inhibit intracellular growth of both L. monocytogenes and B. abortus. Further analysis shows that Trx80 activation prevents the escape of GFP-tagged L. monocytogenes into the cytosol, and induces accumulation of the bacteria within the lysosomes. Inhibition of the lysosomal activity by chloroquine treatment resulted in higher replication of bacteria in TAMs compared to that observed in control cells 24 h post-infection, indicating that TAMs kill bacteria by preventing their escape from the endosomal compartments, which progress into a highly degradative phagolysosome. Significance Our results show that Trx80 potentiates the bactericidal activities of professional phagocytes, and contributes to the first line of defense against intracellular bacteria.
Collapse
Affiliation(s)
- Ximena Cortes-Bratti
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Eugénie Bassères
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Fabiola Herrera-Rodriguez
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología Universidad de Costa Rica, San José, Costa Rica
| | | | - Giuseppe Coppotelli
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jens B. Andersen
- Department of Microbiology and Risk Assessment, National Food Institute, Soeborg, Denmark
| | - Maria G. Masucci
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Arne Holmgren
- Department of Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Esteban Chaves-Olarte
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología Universidad de Costa Rica, San José, Costa Rica
| | - Teresa Frisan
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (TF); (JA)
| | - Javier Avila-Cariño
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (TF); (JA)
| |
Collapse
|
20
|
Williams D, Dunn S, Richardson A, Frank JF, Smith MA. Time course of fetal tissue invasion by Listeria monocytogenes following an oral inoculation in pregnant guinea pigs. J Food Prot 2011; 74:248-53. [PMID: 21333144 DOI: 10.4315/0362-028x.jfp-10-163] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Pregnant women are 20 times more likely to develop listeriosis than are members of the general population, and infection can result in abortion, stillbirth, or neonatal illness. The objective of this study was to orally challenge pregnant guinea pigs with Listeria monocytogenes to assess maternal and fetal tissue invasion at postinoculation days 2, 6, 9, and 21. The time course of invasion was followed by fluorescence microscopy and a traditional culture method. Guinea pigs were treated on gestation day 35 with L. monocytogenes doses ranging from 10(4) to 10(8) CFU. L. monocytogenes was isolated and viewed in maternal and fetal tissues as early as 2 days postinoculation. L. monocytogenes was isolated from placentas, fetal livers and brains, and maternal spleens at similar rates, suggesting that invasion of the spleen could be indicative of fetal invasion. When comparing fecal shedding, all animals treated with 10(4) CFU were shedding L. monocytogenes by postinoculation day 7, and all animals treated with the higher doses (10(6) or 10(8) CFU) were shedding L. monocytogenes by postinoculation day 5. These data suggest that L. monocytogenes crosses the fetoplacental barrier and invades the fetus by day 2 after maternal ingestion. When comparing the sensitivities of microscopy and culture, neither method consistently detected L. monocytogenes at a higher rate. However, detection in individual tissues differed. Microscopy was significantly more sensitive with fetal liver (P<0.001) and brain (P<0.001) at the highest dose of 10(8) CFU, but at the lowest dose of 10(4) CFU culture was significantly more sensitive with maternal spleen (P=0.04).
Collapse
Affiliation(s)
- Denita Williams
- Department of Environmental Health Science, University of Georgia, Athens, Georgia 30602, USA
| | | | | | | | | |
Collapse
|
21
|
Bruno JC, Freitag NE. Constitutive activation of PrfA tilts the balance of Listeria monocytogenes fitness towards life within the host versus environmental survival. PLoS One 2010; 5:e15138. [PMID: 21151923 PMCID: PMC2998416 DOI: 10.1371/journal.pone.0015138] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2010] [Accepted: 10/25/2010] [Indexed: 01/21/2023] Open
Abstract
PrfA is a key regulator of Listeria monocytogenes pathogenesis and induces the expression of multiple virulence factors within the infected host. PrfA is post-translationally regulated such that the protein becomes activated upon bacterial entry into the cell cytosol. The signal that triggers PrfA activation remains unknown, however mutations have been identified (prfA* mutations) that lock the protein into a high activity state. In this report we examine the consequences of constitutive PrfA activation on L. monocytogenes fitness both in vitro and in vivo. Whereas prfA* mutants were hyper-virulent during animal infection, the mutants were compromised for fitness in broth culture and under conditions of stress. Broth culture prfA*-associated fitness defects were alleviated when glycerol was provided as the principal carbon source; under these conditions prfA* mutants exhibited a competitive advantage over wild type strains. Glycerol and other three carbon sugars have been reported to serve as primary carbon sources for L. monocytogenes during cytosolic growth, thus prfA* mutants are metabolically-primed for replication within eukaryotic cells. These results indicate the critical need for environment-appropriate regulation of PrfA activity to enable L. monocytogenes to optimize bacterial fitness inside and outside of host cells.
Collapse
Affiliation(s)
- Joseph C. Bruno
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Nancy E. Freitag
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
22
|
van der Veen S, Abee T. Mixed species biofilms of Listeria monocytogenes and Lactobacillus plantarum show enhanced resistance to benzalkonium chloride and peracetic acid. Int J Food Microbiol 2010; 144:421-31. [PMID: 21084128 DOI: 10.1016/j.ijfoodmicro.2010.10.029] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 09/29/2010] [Accepted: 10/25/2010] [Indexed: 11/17/2022]
Abstract
We investigated the formation of single and mixed species biofilms of Listeria monocytogenes strains EGD-e and LR-991, with Lactobacillus plantarum WCFS1 as secondary species, and their resistance to the disinfectants benzalkonium chloride and peracetic acid. Modulation of growth, biofilm formation, and biofilm composition was achieved by addition of manganese sulfate and/or glucose to the BHI medium. Composition analyses of the mixed species biofilms using plate counts and fluorescence microscopy with dual fluorophores showed that mixed species biofilms were formed in BHI (total count, 8-9 log₁₀ cfu/well) and that they contained 1-2 log₁₀ cfu/well more L. monocytogenes than L. plantarum cells. Addition of manganese sulfate resulted in equal numbers of both species (total count, 8 log₁₀ cfu/well) in the mixed species biofilm, while manganese sulfate in combination with glucose, resulted in 1-2 log₁₀ more L. plantarum than L. monocytogenes cells (total count, 9 log₁₀ cfu/well). Corresponding single species biofilms of L. monocytogenes and L. plantarum contained up to 9 log₁₀ cfu/well. Subsequent disinfection treatments showed mixed species biofilms to be more resistant to treatments with the selected disinfectants. In BHI with additional manganese sulfate, both L. monocytogenes strains and L. plantarum grown in the mixed species biofilm showed less than 2 log₁₀ cfu/well inactivation after exposure for 15 min to 100 μg/ml benzalkonium chloride, while single species biofilms of both L. monocytogenes strains showed 4.5 log₁₀ cfu/well inactivation and single species biofilms of L. plantarum showed 3.3 log₁₀ cfu/well inactivation. Our results indicate that L. monocytogenes and L. plantarum mixed species biofilms can be more resistant to disinfection treatments than single species biofilms.
Collapse
Affiliation(s)
- Stijn van der Veen
- Top Institute Food and Nutrition (TIFN), Nieuwe Kanaal 9A, 6709 PA Wageningen, The Netherlands.
| | | |
Collapse
|
23
|
Douce G, Ross K, Cowan G, Ma J, Mitchell TJ. Novel mucosal vaccines generated by genetic conjugation of heterologous proteins to pneumolysin (PLY) from Streptococcus pneumoniae. Vaccine 2010; 28:3231-7. [PMID: 20188176 DOI: 10.1016/j.vaccine.2010.02.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Revised: 02/05/2010] [Accepted: 02/10/2010] [Indexed: 10/19/2022]
Abstract
Induction of immunity at mucosal surfaces is thought to be an essential feature in the protection of the host against the many pathogens that gain access through these surfaces. Here we describe how strong local and systemic immune responses can be generated when proteins are genetically conjugated to pneumolysin (PLY) from Streptococcus pneumoniae. Using green fluorescent protein (eGFP) and PsaA from S. pneumoniae, we have shown that genetic fusion (eGFPPLY and PsaAPLY) is essential to ensure high levels of antigen specific IgG and IgA in the serum and at mucosal surfaces. This form of vaccination is highly effective with antigen specific antibodies detected after a single dose of nanogram quantities of the conjugated proteins. In addition, generation of a non-toxic variant (eGFPDelta6PLY) indicated that while the toxic activity of PLY was not essential for adjuvanticity, it contributed to the magnitude of the response generated. Whilst vaccination with the PsaAPLY fusion proteins did not protect the animals from challenge, these studies confirm the utility of pneumolysin to act as a novel mucosal adjuvant to substantially increase the local and systemic humoral response to genetically fused protein antigens.
Collapse
Affiliation(s)
- Gill Douce
- Division of Infection and Immunity, Faculty of Biomedical and Life Sciences, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow G12 8TA, UK
| | | | | | | | | |
Collapse
|
24
|
Abstract
Listeria monocytogenes infection in mice is a highly prolific model of bacterial infection. Several in vivo imaging approaches have been used to study host cell dynamics in response to infection, including bioluminescence imaging, confocal microscopy and two-photon microscopy, The application of in vivo imaging to study transgenic mouse models is providing unprecedented opportunities to test specific molecular mechanistic theories about how the host immune response unfolds. In complementary studies, in vivo imaging can be performed using genetically engineered bacterial mutants to assess the impact of specific virulence factors in host cell invasion and pathogenesis. The purpose of this chapter is to provide a general rationale for why in vivo imaging is important, provide an overview of various techniques highlighting the strengths and weaknesses of each, and provide examples of how various imaging techniques have been used to study Listeria infection. Lastly, our goal is to make the reader aware of the tremendous potential these approaches hold for studying host-pathogen interactions.
Collapse
|
25
|
Miner MD, Port GC, Freitag NE. Functional impact of mutational activation on the Listeria monocytogenes central virulence regulator PrfA. MICROBIOLOGY-SGM 2008; 154:3579-3589. [PMID: 18957610 DOI: 10.1099/mic.0.2008/021063-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The transcriptional activator PrfA is required for the expression of virulence factors necessary for Listeria monocytogenes pathogenesis. PrfA is believed to become activated following L. monocytogenes entry into the cytosol of infected host cells, resulting in the induction of target genes whose products are required for bacterial intracellular growth and cell-to-cell spread. Several mutations have been identified that appear to lock PrfA into its highly activated cytosolic form (known as prfA* mutations). In this study PrfA and five PrfA* mutant proteins exhibiting differing degrees of activity were purified and analysed to define the influences of the mutations on distinct aspects of PrfA activity. Based on limited proteolytic digestion, conformational changes were detected for the PrfA* mutant proteins in comparison to wild-type PrfA. For all but one mutant (PrfA Y63C), the DNA binding affinity as measured by electophoretic mobility shift assay appeared to directly correlate with levels of PrfA mutational activation, such that the high-activity mutants exhibited the largest increases in DNA binding affinity and moderately activated mutants exhibited more moderate increases. Surprisingly, the ability of PrfA and PrfA* mutants to form dimers in solution appeared to inversely correlate with levels of PrfA-dependent gene expression. Based on comparisons of protein activity and structural similarities with PrfA family members Crp and CooA, the prfA* mutations modify distinct aspects of PrfA activity that include DNA binding and protein-protein interactions.
Collapse
Affiliation(s)
- Maurine D Miner
- Seattle Biomedical Research Institute, Seattle, WA, USA.,Program in Pathobiology, University of Washington, Seattle, WA, USA
| | - Gary C Port
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA.,Seattle Biomedical Research Institute, Seattle, WA, USA
| | - Nancy E Freitag
- Seattle Biomedical Research Institute, Seattle, WA, USA.,Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA.,Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA.,Program in Pathobiology, University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
Miner MD, Port GC, Bouwer HGA, Chang JC, Freitag NE. A novel prfA mutation that promotes Listeria monocytogenes cytosol entry but reduces bacterial spread and cytotoxicity. Microb Pathog 2008; 45:273-81. [PMID: 18675335 DOI: 10.1016/j.micpath.2008.06.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2008] [Revised: 06/17/2008] [Accepted: 06/26/2008] [Indexed: 10/21/2022]
Abstract
Listeria monocytogenes is an environmental bacterium that becomes a pathogen following ingestion by a mammalian host. The transition from environmental organism to pathogen requires significant changes in gene expression, including the increased expression of gene products that contribute to bacterial growth within host cells. PrfA is an L. monocytogenes transcriptional regulator that becomes activated upon bacterial entry into mammalian cells and induces the expression of gene products required for virulence. How PrfA activation occurs is not known, however several mutations have been identified that increase PrfA activity in strains grown in vitro (prfA mutations). Here we describe a novel prfA mutation that enhances extracellular PrfA-dependent gene expression but in contrast to prfA mutants inhibits the cytosol-mediated induction of virulence genes. prfA Y154C strains entered cells and escaped from phagosomes with an efficiency similar to wild type bacteria, however the mutation prevented efficient L. monocytogenes actin polymerization and reduced spread of bacteria to adjacent cells. The prfA Y154C mutation severely attenuated bacterial virulence in mice but the mutant strains did generate target antigen specific CD8(+) effector cells. Interestingly, the prfA Y154C mutant was significantly less cytotoxic for host cells than wild type L. monocytogenes. The prfA Y154C mutant strain may therefore represent a novel attenuated strain of L. monocytogenes for antigen delivery with reduced host cell toxicity.
Collapse
Affiliation(s)
- Maurine D Miner
- Department of Pathobiology, University of Washington, Seattle, WA, USA
| | | | | | | | | |
Collapse
|
27
|
Drevets DA, Bronze MS. Listeria monocytogenes: epidemiology, human disease, and mechanisms of brain invasion. ACTA ACUST UNITED AC 2008; 53:151-65. [PMID: 18462388 DOI: 10.1111/j.1574-695x.2008.00404.x] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Listeria monocytogenes is a facultative intracellular bacterium that has predilection for causing central nervous systemic infections in humans and domesticated animals. This pathogen can be found worldwide in the food supply and most L. monocytogenes infections are acquired through ingestion of contaminated food. The main clinical syndromes caused by L. monocytogenes include febrile gastroenteritis, perinatal infection, and systemic infections marked by central nervous system infections with or without bacteremia. Experimental infection of mice has been used for over 50 years as a model system to study the pathogenesis of this organism including the mechanisms by which it invades the brain. Data from this model indicate that a specific subset of monocytes, distinguished in part by high expression of the Ly-6C antigen, become parasitized in the bone marrow and have a key role in transporting intracellular bacteria across the blood-brain barriers and into the central nervous system. This Minireview will summarize recent epidemiologic and clinical information regarding L. monocytogenes as a human pathogen and will discuss current in vitro and in vivo data relevant to the role of parasitized monocytes and the pathogenetic mechanisms that underlie its formidable ability to invade the central nervous system.
Collapse
Affiliation(s)
- Douglas A Drevets
- Department of Medicine, Oklahoma University Health Sciences Center and the Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| | | |
Collapse
|
28
|
The Listeria monocytogenes virulence factor InlJ is specifically expressed in vivo and behaves as an adhesin. Infect Immun 2008; 76:1368-78. [PMID: 18227172 DOI: 10.1128/iai.01519-07] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The food-borne pathogen Listeria monocytogenes is adapted to a diversity of environments, such as soil, food, body fluids, and the cytosol of eukaryotic cells. The transition between saprophytic and pathogenic life is mediated through complex regulatory pathways that modulate the expression of virulence factors. Here we examined the expression of inlJ, a recently identified gene encoding a protein of the LPXTG-internalin family and involved in pathogenesis. We show that inlJ expression is controlled neither by the major listerial regulator of virulence genes, PrfA, nor by AxyR, a putative AraC regulator encoded by a gene adjacent to inlJ and divergently transcribed. The InlJ protein is not produced by bacteria grown in vitro in brain heart infusion medium or replicating in the cytosol of tissue-cultured cells. In contrast, it is efficiently produced and localized at the surface of bacteria present in the liver and blood of infected animals. Strikingly, the expression of inlJ by a heterologous promoter in L. monocytogenes or L. innocua promotes bacterial adherence to human cells in vitro. Taken together, these results strongly suggest that InlJ acts as a novel L. monocytogenes sortase-anchored adhesin specifically expressed during infection in vivo.
Collapse
|
29
|
Hara H, Kawamura I, Nomura T, Tominaga T, Tsuchiya K, Mitsuyama M. Cytolysin-dependent escape of the bacterium from the phagosome is required but not sufficient for induction of the Th1 immune response against Listeria monocytogenes infection: distinct role of Listeriolysin O determined by cytolysin gene replacement. Infect Immun 2007; 75:3791-801. [PMID: 17517863 PMCID: PMC1951982 DOI: 10.1128/iai.01779-06] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Listeria monocytogenes evades the antimicrobial mechanisms of macrophages by escaping from the phagosome into the cytosolic space via a unique cytolysin that targets the phagosomal membrane, listeriolysin O (LLO), encoded by hly. Gamma interferon (IFN-gamma), which is known to play a pivotal role in the induction of Th1-dependent protective immunity in mice, appears to be produced, depending on the bacterial virulence factor. To determine whether the LLO molecule (the major virulence factor of L. monocytogenes) is indispensable or the escape of bacteria from the phagosome is sufficient to induce IFN-gamma production, we first constructed an hly-deleted mutant of L. monocytogenes and then established isogenic L. monocytogenes mutants expressing LLO or ivanolysin O (ILO), encoded by ilo from Listeria ivanovii. LLO-expressing L. monocytogenes was highly capable of inducing IFN-gamma production and Listeria-specific protective immunity, while the hly-deleted mutant was not. In contrast, the level of IFN-gamma induced by ILO-expressing L. monocytogenes was significantly lower both in vitro and in vivo, despite the ability of this strain to escape the phagosome and the intracellular multiplication at a level equivalent to that of LLO-expressing L. monocytogenes. Only a negligible level of protective immunity was induced in mice against challenge with LLO- and ILO-expressing L. monocytogenes. These results clearly show that escape of the bacterium from the phagosome is a prerequisite but is not sufficient for the IFN-gamma-dependent Th1 response against L. monocytogenes, and some distinct molecular nature of LLO is indispensable for the final induction of IFN-gamma that is essentially required to generate a Th1-dependent immune response.
Collapse
Affiliation(s)
- Hideki Hara
- Department of Microbiology, Kyoto University Graduate School of Medicine, Yoshida-konoecho, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
The PrfA protein, a member of the Crp/Cap-Fnr family of bacterial transcription factors, controls the expression of key virulence determinants of the facultative intracellular pathogen Listeria monocytogenes. Each of the steps of the listerial intracellular infection cycle-host cell invasion, phagosomal escape, cytosolic replication, and direct cell-to-cell spread-is mediated by products of the PrfA regulon. Only 10 of the 2853 genes of the L. monocytogenes EGDe genome have been confirmed as bona fide (directly regulated) members of this regulon, a number surprisingly small given the apparent complexity of listerial intracellular parasitism. PrfA activates transcription by binding as a dimer to a palindromic promoter element of canonical sequence tTAACanntGTtAa, with seven invariant nucleotides (in capitals) and a two-mismatch tolerance. PrfA integrates a number of environmental and bacteria-derived signals to ensure the correct spatio-temporal and niche-adapted expression of the regulon, with maximum induction in the host cell cytosol and repression in the environmental habitat. Regulation operates through changes in PrfA activity-presumably by cofactor-mediated allosteric shift-and concentration, and involves transcriptional, translational and post-translational control mechanisms. There is evidence that PrfA exerts a more global influence on L. monocytogenes physiology via indirect mechanisms.
Collapse
Affiliation(s)
- Mariela Scortti
- Bacterial Molecular Pathogenesis Group, Veterinary Molecular Microbiology Section, Faculty of Medical and Veterinary Sciences, University of Bristol, Langford, UK
| | | | | | | | | |
Collapse
|
31
|
Ebersbach G, Jacobs-Wagner C. Exploration into the spatial and temporal mechanisms of bacterial polarity. Trends Microbiol 2007; 15:101-8. [PMID: 17275310 DOI: 10.1016/j.tim.2007.01.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Revised: 01/04/2007] [Accepted: 01/23/2007] [Indexed: 10/23/2022]
Abstract
The recognition of bacterial asymmetry is not new: the first high-resolution microscopy studies revealed that bacteria come in a multitude of shapes and sometimes carry asymmetrically localized external structures such as flagella on the cell surface. Even so, the idea that bacteria could have an inherent overall polarity, which affects not only their outer appearance but also many of their vital processes, has only recently been appreciated. In this review, we focus on recent advances in our understanding of the molecular mechanisms underlying the establishment of polarized functions and cell polarity in bacteria.
Collapse
Affiliation(s)
- Gitte Ebersbach
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | | |
Collapse
|
32
|
Westcott MM, Henry CJ, Cook AS, Grant KW, Hiltbold EM. Differential susceptibility of bone marrow-derived dendritic cells and macrophages to productive infection with Listeria monocytogenes. Cell Microbiol 2007; 9:1397-411. [PMID: 17250592 DOI: 10.1111/j.1462-5822.2006.00880.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Dendritic cells (DC) are required for the immune response against Listeria monocytogenes and are permissive for infection in vivo and in vitro. However, it is unclear if DC provide a desirable intracellular niche for bacterial growth. To address this issue, we have compared the behaviour of L. monocytogenes in murine bone marrow-derived DC and macrophages (BMM). Similar to BMM, bacteria escaped to the cytosol in DC, replicated, and spread to adjacent cells. However, DC infection was less robust in terms of intracellular doubling time and total increase in bacterial numbers. Immunofluorescence analysis using a strain of L. monocytogenes that expresses green fluorescent protein upon bacterial entry into the cytosol suggested that a subpopulation of DC restricted bacteria to vacuoles, a finding that was confirmed by electron microscopy. In unstimulated DC cultures, L. monocytogenes replicated preferentially in phenotypically immature cells. Furthermore, DC that were induced to mature prior to infection were poor hosts for bacterial growth. We conclude that DC provide a suboptimal niche for L. monocytogenes growth, and this is at least in part a function of the DC maturation state. Therefore, the generation of an effective T cell response may be a net effect of both productive and non-productive infection of DC.
Collapse
Affiliation(s)
- Marlena M Westcott
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | | | | | | | | |
Collapse
|
33
|
Dubail I, Bigot A, Lazarevic V, Soldo B, Euphrasie D, Dupuis M, Charbit A. Identification of an essential gene of Listeria monocytogenes involved in teichoic acid biogenesis. J Bacteriol 2006; 188:6580-91. [PMID: 16952950 PMCID: PMC1595501 DOI: 10.1128/jb.00771-06] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Listeria monocytogenes is a facultative intracellular gram-positive bacterium responsible for severe opportunistic infections in humans and animals. We had previously identified a gene encoding a putative UDP-N-acetylglucosamine 2-epimerase, a precursor of the teichoic acid linkage unit, in the genome of L monocytogenes strain EGD-e. This gene, now designated lmo2537, encodes a protein that shares 62% identity with the cognate epimerase MnaA of Bacillus subtilis and 55% identity with Cap5P of Staphylococcus aureus. Here, we addressed the role of lmo2537 in L. monocytogenes pathogenesis by constructing a conditional knockout mutant. The data presented here demonstrate that lmo2537 is an essential gene of L. monocytogenes that is involved in teichoic acid biogenesis. In vivo, the conditional mutant is very rapidly eliminated from the target organs of infected mice and thus is totally avirulent.
Collapse
Affiliation(s)
- Iharilalao Dubail
- Faculté de Médecine Necker, 156, Rue de Vaugirard, 75730 Paris Cedex 15, France
| | | | | | | | | | | | | |
Collapse
|
34
|
Gray MJ, Freitag NE, Boor KJ. How the bacterial pathogen Listeria monocytogenes mediates the switch from environmental Dr. Jekyll to pathogenic Mr. Hyde. Infect Immun 2006; 74:2505-12. [PMID: 16622185 PMCID: PMC1459693 DOI: 10.1128/iai.74.5.2505-2512.2006] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Michael J Gray
- Department of Food Science, Cornell University, Ithaca, NY 14853, USA
| | | | | |
Collapse
|
35
|
Scortti M, Lacharme-Lora L, Wagner M, Chico-Calero I, Losito P, Vázquez-Boland JA. Coexpression of virulence and fosfomycin susceptibility in Listeria: molecular basis of an antimicrobial in vitro-in vivo paradox. Nat Med 2006; 12:515-7. [PMID: 16633349 DOI: 10.1038/nm1396] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2005] [Accepted: 03/23/2006] [Indexed: 11/08/2022]
Abstract
Discrepancies between resistance in vitro and therapeutic efficacy in vivo are generally attributed to failure of laboratory susceptibility tests to reflect an antibiotic's pharmacokinetic or pharmacodynamic properties. We show here that this phenomenon can result from differential in vitro-in vivo expression of bacterial determinants of antibiotic susceptibility. We found that an in vivo-induced virulence factor, Hpt, also mediates uptake of fosfomycin in Listeria monocytogenes. These bacteria therefore seem resistant to fosfomycin in vitro, although they are in fact susceptible to the antibiotic during infection.
Collapse
Affiliation(s)
- Mariela Scortti
- Bacterial Molecular Pathogenesis Group, Faculty of Medical and Veterinary Sciences, University of Bristol, Langford BS40 5DU, UK
| | | | | | | | | | | |
Collapse
|
36
|
Joseph B, Przybilla K, Stühler C, Schauer K, Slaghuis J, Fuchs TM, Goebel W. Identification of Listeria monocytogenes genes contributing to intracellular replication by expression profiling and mutant screening. J Bacteriol 2006; 188:556-68. [PMID: 16385046 PMCID: PMC1347271 DOI: 10.1128/jb.188.2.556-568.2006] [Citation(s) in RCA: 226] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A successful transition of Listeria monocytogenes from the extracellular to the intracellular environment requires a precise adaptation response to conditions encountered in the host milieu. Although many key steps in the intracellular lifestyle of this gram-positive pathogen are well characterized, our knowledge about the factors required for cytosolic proliferation is still rather limited. We used DNA microarray and real-time reverse transcriptase PCR analyses to investigate the transcriptional profile of intracellular L. monocytogenes following epithelial cell infection. Approximately 19% of the genes were differentially expressed by at least 1.6-fold relative to their level of transcription when grown in brain heart infusion medium, including genes encoding transporter proteins essential for the uptake of carbon and nitrogen sources, factors involved in anabolic pathways, stress proteins, transcriptional regulators, and proteins of unknown function. To validate the biological relevance of the intracellular gene expression profile, a random mutant library of L. monocytogenes was constructed by insertion-duplication mutagenesis and screened for intracellular-growth-deficient strains. By interfacing the results of both approaches, we provide evidence that L. monocytogenes can use alternative carbon sources like phosphorylated glucose and glycerol and nitrogen sources like ethanolamine during replication in epithelial cells and that the pentose phosphate cycle, but not glycolysis, is the predominant pathway of sugar metabolism in the host environment. Additionally, we show that the synthesis of arginine, isoleucine, leucine, and valine, as well as a species-specific phosphoenolpyruvate-dependent phosphotransferase system, play a major role in the intracellular growth of L. monocytogenes.
Collapse
Affiliation(s)
- Biju Joseph
- Theodor-Boveri-Institut (Biozentrum), Lehrstuhl für Mikrobiologie, Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
37
|
Rafelski SM, Theriot JA. Mechanism of polarization of Listeria monocytogenes surface protein ActA. Mol Microbiol 2006; 59:1262-79. [PMID: 16430699 PMCID: PMC1413586 DOI: 10.1111/j.1365-2958.2006.05025.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2005] [Indexed: 11/26/2022]
Abstract
The polar distribution of the ActA protein on the surface of the Gram-positive intracellular bacterial pathogen, Listeria monocytogenes, is required for bacterial actin-based motility and successful infection. ActA spans both the bacterial membrane and the peptidoglycan cell wall. We have directly examined the de novo ActA polarization process in vitro by using an ActA-RFP (red fluorescent protein) fusion. After induction of expression, ActA initially appeared at distinct sites along the sides of bacteria and was then redistributed over the entire cylindrical cell body through helical cell wall growth. The accumulation of ActA at the bacterial poles displayed slower kinetics, occurring over several bacterial generations. ActA accumulated more efficiently at younger, less inert poles, and proper polarization required an optimal balance between protein secretion and bacterial growth rates. Within infected host cells, younger generations of L. monocytogenes initiated motility more quickly than older ones, consistent with our in vitro observations of de novo ActA polarization. We propose a model in which the polarization of ActA, and possibly other Gram-positive cell wall-associated proteins, may be a direct consequence of the differential cell wall growth rates along the bacterium and dependent on the relative rates of protein secretion, protein degradation and bacterial growth.
Collapse
Affiliation(s)
- Susanne M Rafelski
- Departments of Biochemistry and Stanford University Medical Center279 W. Campus Dr, Stanford, CA 94305-5307, USA
| | - Julie A Theriot
- Microbiology and Immunology, Stanford University Medical Center279 W. Campus Dr, Stanford, CA 94305-5307, USA.
| |
Collapse
|
38
|
Pentecost M, Otto G, Theriot JA, Amieva MR. Listeria monocytogenes invades the epithelial junctions at sites of cell extrusion. PLoS Pathog 2006; 2:e3. [PMID: 16446782 PMCID: PMC1354196 DOI: 10.1371/journal.ppat.0020003] [Citation(s) in RCA: 155] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Accepted: 12/19/2005] [Indexed: 12/19/2022] Open
Abstract
Listeria monocytogenes causes invasive disease by crossing the intestinal epithelial barrier. This process depends on the interaction between the bacterial surface protein Internalin A and the host protein E-cadherin, located below the epithelial tight junctions at the lateral cell-to-cell contacts. We used polarized MDCK cells as a model epithelium to determine how L. monocytogenes breaches the tight junctions to gain access to this basolateral receptor protein. We determined that L. monocytogenes does not actively disrupt the tight junctions, but finds E-cadherin at a morphologically distinct subset of intercellular junctions. We identified these sites as naturally occurring regions where single senescent cells are expelled and detached from the epithelium by extrusion. The surrounding cells reorganize to form a multicellular junction that maintains epithelial continuity. We found that E-cadherin is transiently exposed to the lumenal surface at multicellular junctions during and after cell extrusion, and that L. monocytogenes takes advantage of junctional remodeling to adhere to and subsequently invade the epithelium. In intact epithelial monolayers, an anti-E-cadherin antibody specifically decorates multicellular junctions and blocks L. monocytogenes adhesion. Furthermore, an L. monocytogenes mutant in the Internalin A gene is completely deficient in attachment to the epithelial apical surface and is unable to invade. We hypothesized that L. monocytogenes utilizes analogous extrusion sites for epithelial invasion in vivo. By infecting rabbit ileal loops, we found that the junctions at the cell extrusion zone of villus tips are the specific target for L. monocytogenes adhesion and invasion. Thus, L. monocytogenes exploits the dynamic nature of epithelial renewal and junctional remodeling to breach the intestinal barrier.
Collapse
Affiliation(s)
- Mickey Pentecost
- Department of Microbiology and Immunology, Stanford University, Stanford, California, United States of America
| | - Glen Otto
- Department of Comparative Medicine, Stanford University, Stanford, California, United States of America
| | - Julie A Theriot
- Department of Microbiology and Immunology, Stanford University, Stanford, California, United States of America
- Department of Biochemistry, Stanford University, Stanford, California, United States of America
| | - Manuel R Amieva
- Department of Microbiology and Immunology, Stanford University, Stanford, California, United States of America
- Department of Pediatrics, Stanford University, Stanford, California, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
39
|
Shen A, Higgins DE. The 5' untranslated region-mediated enhancement of intracellular listeriolysin O production is required for Listeria monocytogenes pathogenicity. Mol Microbiol 2005; 57:1460-73. [PMID: 16102013 DOI: 10.1111/j.1365-2958.2005.04780.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Listeriolysin O (LLO) and ActA are essential virulence determinants for Listeria monocytogenes pathogenesis. Transcription of actA and hly, encoding LLO, is regulated by PrfA and increases dramatically during intracellular infection. The 5' untranslated regions (5' UTRs) of actA and prfA have been shown to upregulate expression of their respective gene products. Here, we demonstrate that the hly 5' UTR plays a critical role in regulating expression of LLO during intracellular infection. Deletion of the hly 5' UTR, while retaining the hly ribosome binding site, had a moderate effect on LLO production during growth in broth culture, yet resulted in a marked decrease in LLO levels during intracellular infection. The diminished level of LLO resulted in a significant defect in bacterial cell-to-cell spread during intracellular infection and a 10-fold reduction in virulence during in vivo infection of mice. Insertion of the hly 5' UTR sequence between a heterologous promoter and reporter gene sequences indicated that the hly 5' UTR functions independent of PrfA-mediated transcription and can enhance expression of cis-associated genes through a mechanism that appears to act at both a post-transcriptional and translational level. The ability of the hly 5' UTR to increase gene expression can be exploited to achieve PrfA-independent complementation of virulence genes and high-level expression of single copy heterologous genes in L. monocytogenes.
Collapse
Affiliation(s)
- Aimee Shen
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, MA 02115-6092, USA
| | | |
Collapse
|
40
|
Rafelski SM, Theriot JA. Bacterial shape and ActA distribution affect initiation of Listeria monocytogenes actin-based motility. Biophys J 2005; 89:2146-58. [PMID: 15980176 PMCID: PMC1366716 DOI: 10.1529/biophysj.105.061168] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Accepted: 06/16/2005] [Indexed: 11/18/2022] Open
Abstract
We have examined the process by which the intracellular bacterial pathogen Listeria monocytogenes initiates actin-based motility and determined the contribution of the variable surface distribution of the ActA protein to initiation and steady-state movement. To directly correlate ActA distributions to actin dynamics and motility of live bacteria, ActA was fused to a monomeric red fluorescent protein (mRFP1). Actin comet tail formation and steady-state bacterial movement rates both depended on ActA distribution, which in turn was tightly coupled to the bacterial cell cycle. Motility initiation was found to be a highly complex, multistep process for bacteria, in contrast to the simple symmetry breaking previously observed for ActA-coated spherical beads. F-actin initially accumulated along the sides of the bacterium and then slowly migrated to the bacterial pole expressing the highest density of ActA as a tail formed. Early movement was highly unstable with extreme changes in speed and frequent stops. Over time, saltatory motility and sensitivity to the immediate environment decreased as bacterial movement became robust at a constant steady-state speed.
Collapse
Affiliation(s)
- Susanne M Rafelski
- Department of Biochemistry, Stanford University Medical Center, Stanford, California 94305-5307, USA
| | | |
Collapse
|
41
|
Beurg M, Hafidi A, Skinner L, Cowan G, Hondarrague Y, Mitchell TJ, Dulon D. The mechanism of pneumolysin-induced cochlear hair cell death in the rat. J Physiol 2005; 568:211-27. [PMID: 16051626 PMCID: PMC1474774 DOI: 10.1113/jphysiol.2005.092478] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Streptoccocus pneumoniae infection can result in local and systemic diseases such as otitis media, pneumonia and meningitis. Sensorineural hearing loss associated with this infection is mediated by the release of an exotoxin, pneumolysin. The goal of the present study was to characterize the mechanisms of pneumolysin toxicity in cochlear hair cells in vitro. Pneumolysin induced severe damage in cochlear hair cells, ranging from stereocilia disorganization to total cell loss. Surprisingly, pneumolysin-induced cell death preferentially targeted inner hair cells. Pneumolysin triggered in vitro cell death by an influx of calcium. Extracellular calcium appeared to enter the cell through a pore formed by the toxin. Buffering intracellular calcium with BAPTA improved hair cell survival. The mitochondrial apoptotic pathway involved in pneumolysin-induced cell death was demonstrated by the use of bongkrekic acid. Binding of pneumolysin to the hair cell plasma membrane was required to induce cell death. Increasing external calcium reduced cell toxicity by preventing the binding of pneumolysin to hair cell membranes. These results showed the significant role of calcium both in triggering pneumolysin-induced hair cell apoptosis and in preventing the toxin from binding to its cellular target.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Apoptosis/drug effects
- Bacterial Proteins/genetics
- Bacterial Proteins/toxicity
- Bongkrekic Acid/pharmacology
- Calcium/metabolism
- Calcium/pharmacology
- Calcium Channels/drug effects
- Calcium Channels/metabolism
- Cell Survival/drug effects
- Chelating Agents/pharmacology
- Cochlea/drug effects
- Cochlea/metabolism
- Egtazic Acid/analogs & derivatives
- Egtazic Acid/pharmacology
- Green Fluorescent Proteins/genetics
- Hair Cells, Auditory, Inner/drug effects
- Hair Cells, Auditory, Inner/metabolism
- Hair Cells, Auditory, Inner/ultrastructure
- Hair Cells, Auditory, Outer/drug effects
- Hair Cells, Auditory, Outer/metabolism
- Hair Cells, Auditory, Outer/ultrastructure
- Mitochondria/drug effects
- Mitochondria/metabolism
- Mitochondrial ADP, ATP Translocases/antagonists & inhibitors
- Organ Culture Techniques
- Rats
- Rats, Wistar
- Recombinant Fusion Proteins/metabolism
- Streptolysins/genetics
- Streptolysins/toxicity
- Time Factors
Collapse
Affiliation(s)
- Maryline Beurg
- EA-3665 Université Victor Segalen Bordeaux 2, Laboratoire de Biologie Cellulaire et Moléculaire de l'Audition, Hôpital Pellegrin, Bat PQR, 33076 Bordeaux, France.
| | | | | | | | | | | | | |
Collapse
|
42
|
Fickl H, Cockeran R, Steel HC, Feldman C, Cowan G, Mitchell TJ, Anderson R. Pneumolysin-mediated activation of NFkappaB in human neutrophils is antagonized by docosahexaenoic acid. Clin Exp Immunol 2005; 140:274-81. [PMID: 15807851 PMCID: PMC1809376 DOI: 10.1111/j.1365-2249.2005.02757.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
This study was designed to investigate the relationship between influx of extracellular Ca(2+), activation of NFkappaB and synthesis of interleukin-8 (IL-8) following exposure of human neutrophils to subcytolytic concentrations (8.37 and 41.75 ng/ml) of the pneumococcal toxin, pneumolysin, as well as the potential of the omega-3 polyunsaturated fatty acid, docosahexaenoic acid, to antagonize these events. Activation and translocation of NFkappaB were measured using a radiometric electrophoretic mobility shift assay, while influx of extracellular Ca(2+) and synthesis of IL-8 were determined using a radioassay and an ELISA procedure, respectively. Exposure of neutrophils to pneumolysin was accompanied by influx of Ca(2+), activation of NFkappaB, and synthesis of IL-8, all of which were eliminated by inclusion of the Ca(2+)-chelating agent, EGTA (10 m m), in the cell-suspending medium, as well as by pretreatment of the cells with docosahexaenoic acid (5 and 10 microg/ml). The antagonistic effects of docosahexaenoic acid on these pro-inflammatory interactions of pneumolysin with neutrophils were not attributable to inactivation of the toxin, and required the continuous presence of the fatty acid. These observations demonstrate that activation of NFkappaB and synthesis of IL-8, following exposure of neutrophils to pneumolysin are dependent on toxin-mediated influx of Ca(2+) and that these potentially harmful activities of the toxin are antagonized by docosahexaenoic acid.
Collapse
Affiliation(s)
- H Fickl
- Medical Research Council Unit for Inflammation and Immunity, Department of Immunology, University of PretoriaPretoria
| | - R Cockeran
- Medical Research Council Unit for Inflammation and Immunity, Department of Immunology, University of PretoriaPretoria
| | - H C Steel
- Medical Research Council Unit for Inflammation and Immunity, Department of Immunology, University of PretoriaPretoria
| | - C Feldman
- Division of Pulmonology, Department of Medicine, Johannesburg Hospital and University of the WitwatersrandJohannesburg, South Africa
| | - G Cowan
- Division of Infection and Immunity, Institute of Biomedical and Life Sciences, University of GlasgowGlasgow, UK
| | - T J Mitchell
- Division of Infection and Immunity, Institute of Biomedical and Life Sciences, University of GlasgowGlasgow, UK
| | - R Anderson
- Medical Research Council Unit for Inflammation and Immunity, Department of Immunology, University of PretoriaPretoria
| |
Collapse
|
43
|
Mueller KJ, Freitag NE. Pleiotropic enhancement of bacterial pathogenesis resulting from the constitutive activation of the Listeria monocytogenes regulatory factor PrfA. Infect Immun 2005; 73:1917-26. [PMID: 15784531 PMCID: PMC1087396 DOI: 10.1128/iai.73.4.1917-1926.2005] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Listeria monocytogenes is a facultative intracellular bacterial pathogen that causes serious disease in immunocompromised individuals, pregnant women, and neonates. Bacterial virulence is mediated by the expression of specific gene products that facilitate entry into host cells and enable bacterial replication; the majority of these gene products are regulated by a transcriptional activator known as PrfA. L. monocytogenes strains containing prfA E77K or prfA G155S mutations exhibit increased expression of virulence genes in broth culture and are hypervirulent in mice. To define the scope of the influences of the prfA E77K and prfA G155S mutations on L. monocytogenes pathogenesis, multiple aspects of bacterial invasion and intracellular growth were examined. Enhanced bacterial invasion of host epithelial cells was dependent on the expression of a number of surface proteins previously associated with invasion, including InlA, InlB, and ActA. In addition to these surface proteins, increased production of the hly-encoded secreted hemolysin listeriolysin O (LLO) was also found to significantly enhance bacterial invasion into epithelial cell lines for both prfA mutant strains. Although prfA E77K and prfA G155S strains were similar in their invasive phenotypes, the infection of epithelial cells with prfA E77K strains resulted in host cell plasma membrane damage, whereas prfA G155S strains did not alter plasma membrane integrity. Bacterial infection of human epithelial cells, in which the production of LLO is not required for bacterial entry into the cytosol, indicated that prfA E77K cytotoxic effects were mediated via LLO. Both prfA E77K and prfA G155S strains were more efficient than wild-type bacteria in gaining access to the host cell cytosol and in initiating the polymerization of host cell actin, and both were capable of mediating LLO-independent lysis of host cell vacuoles in cell lines for which L. monocytogenes vacuole disruption normally requires LLO activity. These experiments illuminate the diverse facets of L. monocytogenes pathogenesis that are significantly enhanced by the constitutive activation of PrfA via prfA mutations and underscore the critical role of this protein in promoting L. monocytogenes virulence.
Collapse
Affiliation(s)
- Kimberly J Mueller
- Seattle Biomedical Research Institute, 307 Westlake Ave N., Ste. 500, Seattle, WA 98109-5219, USA
| | | |
Collapse
|
44
|
Schwab U, Bowen B, Nadon C, Wiedmann M, Boor KJ. The Listeria monocytogenes prfAP2 promoter is regulated by sigma B in a growth phase dependent manner. FEMS Microbiol Lett 2005; 245:329-36. [PMID: 15837390 DOI: 10.1016/j.femsle.2005.03.025] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2005] [Revised: 03/15/2005] [Accepted: 03/15/2005] [Indexed: 11/25/2022] Open
Abstract
Listeria monocytogenes prfA, encoding positive regulatory factor A, is transcribed from three promoters (prfAP1, prfAP2, and PplcA). The prfAP2 promoter was previously proposed to be sigma B (sigma(B))-dependent. This hypothesis was tested by creating prfA promoter-gus transcriptional fusions in both L. monocytogenes wild-type (wt) and DeltasigB backgrounds and then measuring (i) beta-glucuronidase (GUS) activities; (ii) gus mRNA transcript levels; and (iii) the presence or absence of GUS in cells by immunofluorescence staining. prfAP2-directed expression increased as the wt L. monocytogenes strain entered stationary phase, whereas prfAP2-directed expression was greatly reduced in the DeltasigB strain, confirming both growth phase- and sigma(B)-dependent transcription of prfAP2. We conclude that prfAP2 is directly regulated by sigma(B).
Collapse
Affiliation(s)
- Ute Schwab
- Department of Food Science, Cornell University, 415 Stocking Hall, Ithaca, NY 14853, USA.
| | | | | | | | | |
Collapse
|
45
|
Noah CW, Shaw CI, Ikeda JS, Kreuzer KS, Sofos JN. Development of green fluorescent protein-expressing bacterial strains and evaluation for potential use as positive controls in sample analyses. J Food Prot 2005; 68:680-6. [PMID: 15830656 DOI: 10.4315/0362-028x-68.4.680] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Strains of enterohemorrhagic Escherichia coli O157:H7 and Salmonella Typhimurium were engineered to express the gene for a modified green fluorescent protein (GFP) and were evaluated for potential use as positive controls in sample analyses. The strains fluoresced when observed as colonies with a handheld UV lamp or as individual cells under a fluorescent microscope. The strains maintained their fluorescence following growth in three series of transfer experiments including 8 to 11 passages from broth to broth and twice for 15 consecutive transfers from broth onto Trypticase soy agar plates. Cultures also maintained stability in the ability to fluoresce when agar plates were refrigerated (4 degrees C) for up to 12 days. Growth characteristics of the GFP-positive strains were comparable to those of corresponding control strains. The GFP-positive strains were successfully identified using rapid diagnostic methods and were differentiated from their corresponding non-GFP strains by pulsed-field gel electrophoresis but not by repetitive extragenic palindromic PCR. The GFP-positive and the control strains were recovered successfully from individually inoculated food samples (Feta cheese, raw shrimp, cooked shrimp, and cooked crawfish). However, in one Feta cheese sample and one raw shrimp sample inoculated with combined GFP-positive and GFP-negative cultures, colonies of the GFP-positive strains were not observed under UV light; fluorescing cells in one of the inoculated samples (raw shrimp) were revealed by microscopy. In general, the isolates from the inoculated foods were GFP positive by microscopic examination; the pure isolates could also be restreaked onto Trypticase soy agar, and colonies could be visually examined under UV light. Because GFP strains are not known to occur naturally in the environment, the use of the Salmonella GFP-positive strain may offer advantages as a positive control even when distinct and rare serotypes are available. The GFP-positive E. coli O157:H7 strain may also prove beneficial for use as a positive control strain for sample analyses.
Collapse
Affiliation(s)
- Charles W Noah
- Denver District Office, Food and Drug Administration, Denver, Colorado 80225, USA.
| | | | | | | | | |
Collapse
|
46
|
Wong KKY, Freitag NE. A novel mutation within the central Listeria monocytogenes regulator PrfA that results in constitutive expression of virulence gene products. J Bacteriol 2004; 186:6265-76. [PMID: 15342597 PMCID: PMC515134 DOI: 10.1128/jb.186.18.6265-6276.2004] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The PrfA protein of Listeria monocytogenes functions as a key regulatory factor for the coordinated expression of many virulence genes during bacterial infection of host cells. PrfA activity is controlled by multiple regulatory mechanisms, including an apparent requirement for either the presence of a cofactor or some form of posttranslational modification that regulates the activation of PrfA. In this study, we describe the identification and characterization of a novel PrfA mutation that results in constitutive activation of the PrfA protein. The PrfA L140F mutation was found to confer high-level expression of PrfA-regulated genes and to be functionally dominant over the wild-type allele. The presence of the PrfA L140F mutation resulted in the aggregation of L. monocytogenes in broth culture and, unlike previously described prfA mutations, appeared to be slightly toxic to the bacteria. High-level PrfA-dependent gene expression showed no additional increase in L. monocytogenes strains containing an additional copy of prfA L140F despite a >4-fold increase in PrfA protein levels. In contrast, the introduction of multiple copies of the wild-type prfA allele to L. monocytogenes resulted in a corresponding increase in PrfA-dependent gene expression, although overall expression levels remained far below those observed for PrfA L140F strains. These results suggest a hierarchy of PrfA regulation, such that the relative levels of PrfA protein present within the cell correlate with the levels of PrfA-dependent gene expression when the protein is not in its fully activated state; however, saturating levels of the protein are then quickly reached when PrfA is converted to its active form. Regulation of the PrfA activation status must be an important facet of L. monocytogenes survival, as mutations that result in constitutive PrfA activation may have deleterious consequences for bacterial physiology.
Collapse
Affiliation(s)
- Kendy K Y Wong
- Seattle Biomedical Research Institute, 307 Westlake Ave. N., Suite 500, Seattle, WA 98109-5219, USA
| | | |
Collapse
|
47
|
Ermolaeva S, Novella S, Vega Y, Ripio MT, Scortti M, Vázquez-Boland JA. Negative control of Listeria monocytogenes virulence genes by a diffusible autorepressor. Mol Microbiol 2004; 52:601-11. [PMID: 15066044 DOI: 10.1111/j.1365-2958.2004.04003.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Virulence genes from the facultative intracellular pathogen Listeria monocytogenes are controlled by the transcriptional regulator PrfA. Although PrfA synthesis is activated at 37 degrees C, PrfA-dependent expression remains low in rich medium. However, a strong induction of the PrfA regulon is observed when L. monocytogenes is cultured in the presence of activated charcoal. Here, we show that the 'charcoal effect' results from the adsorption of a diffusible autorepressor substance released by L. monocytogenes during exponential growth. Analyses using an L. monocytogenes strain in which the prfA gene is expressed constitutively at 37 degrees C from a plasmid indicate that the autoregulatory substance represses PrfA-dependent expression by inhibiting PrfA activity. PrfA presumably functions via an allosteric activation mechanism. The inhibitory effect is bypassed by a PrfA* mutation that locks PrfA in fully active conformation, suggesting that the autorepressor interferes with the allosteric shift of PrfA. Our data indicate that the listerial autorepressor is a low-molecular-weight hydrophobic substance. We suggest that this diffusible substance mediates a quorum-sensing mechanism by which L. monocytogenes restricts the expression of its PrfA virulence regulon. This autoregulatory pathway could serve L. monocytogenes to ensure the silencing of virulence genes during extracellular growth at 37 degrees C. It may also play a role during intracellular infection, by limiting the damage to the host cell caused by an excess production of cytotoxic PrfA-dependent virulence factors in the PrfA-activating cytosolic compartment.
Collapse
Affiliation(s)
- Svetlana Ermolaeva
- Grupo de Patogénesis Molecular Bacteriana, Unidad de Microbiología e Inmunología, Facultad de Veterinaria, Universidad Complutense de Madrid, Spain
| | | | | | | | | | | |
Collapse
|
48
|
Vialette M, Jandos-Rudnik AM, Guyard C, Legeay O, Pinon A, Lange M. Validating the use of green fluorescent-marked Escherichia coli O157:H7 for assessing the organism behaviour in foods. J Appl Microbiol 2004; 96:1097-104. [PMID: 15078527 DOI: 10.1111/j.1365-2672.2004.02245.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
AIMS Monitoring bacterial kinetics in food is of great importance in food safety. The targeted micro-organism has to be identified accurately among competitive flora. Using green fluorescent protein (GFP)-transformed strains is a possible answer to such issues. However, quantitative studies require that this transformation does not alter the micro-organism behaviour: parent and transformed organisms were thus compared. METHODS AND RESULTS Three Escherichia coli O157:H7 strains were transformed using a GFP-plasmid expressing. Parent and transformed strains were compared according to their genetic characteristics and serotypes. Growth ability was also assessed in constant and fluctuating temperature profiles. Cardinal values of pH, water activity and temperature were computed. No differences were observed between parent and transformed strains for all these experiments. The plasmid was satisfactorily maintained within transformed strains throughout the studies. Growth was eventually monitored in beef meat. CONCLUSIONS Using the GFP marker is of great value, as it allows easier enumeration of E. coli O157:H7 in food in the presence of natural microflora. Using transformed strains is legitimate: their behaviour does not differ from that of their parent strains. SIGNIFICANCE AND IMPACT OF THE STUDY GFP transformation appears to be a valuable and reliable tool for challenge testing studies and predictive microbiology.
Collapse
Affiliation(s)
- M Vialette
- Institut Pasteur de Lille, Lille, France.
| | | | | | | | | | | |
Collapse
|
49
|
Drevets DA, Leenen PJM, Greenfield RA. Invasion of the central nervous system by intracellular bacteria. Clin Microbiol Rev 2004; 17:323-47. [PMID: 15084504 PMCID: PMC387409 DOI: 10.1128/cmr.17.2.323-347.2004] [Citation(s) in RCA: 144] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Infection of the central nervous system (CNS) is a severe and frequently fatal event during the course of many diseases caused by microbes with predominantly intracellular life cycles. Examples of these include the facultative intracellular bacteria Listeria monocytogenes, Mycobacterium tuberculosis, and Brucella and Salmonella spp. and obligate intracellular microbes of the Rickettsiaceae family and Tropheryma whipplei. Unfortunately, the mechanisms used by intracellular bacterial pathogens to enter the CNS are less well known than those used by bacterial pathogens with an extracellular life cycle. The goal of this review is to elaborate on the means by which intracellular bacterial pathogens establish infection within the CNS. This review encompasses the clinical and pathological findings that pertain to the CNS infection in humans and includes experimental data from animal models that illuminate how these microbes enter the CNS. Recent experimental data showing that L. monocytogenes can invade the CNS by more than one mechanism make it a useful model for discussing the various routes for neuroinvasion used by intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Douglas A Drevets
- Department of Medicine, Oklahoma University Health Sciences Center, Oklahoma City, Oklahoma 73104, USA.
| | | | | |
Collapse
|
50
|
Wong KKY, Bouwer HGA, Freitag NE. Evidence implicating the 5' untranslated region of Listeria monocytogenes actA in the regulation of bacterial actin-based motility. Cell Microbiol 2004; 6:155-66. [PMID: 14706101 DOI: 10.1046/j.1462-5822.2003.00348.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The ActA protein of Listeria monocytogenes is a major virulence factor, essential for the recruitment and polymerization of host actin filaments that lead to intracellular motility and cell-to-cell spread of bacteria within the infected host. The expression of actA is tightly regulated and is strongly induced only when L. monocytogenes is within the host cytosol. Intracellular induction of actA expression is mediated through a single promoter element that directs the expression of a messenger RNA with a long (150 bp) 5' untranslated region (UTR). Deletion of the actA+3 to +130 upstream region was found to result in bacterial mutants that were no longer capable of intracellular actin recruitment or cell-to-cell spread, thus indicating that this region is important for actA expression. L. monocytogenes strains that contained smaller deletions (21-23 bp) within the actA upstream region demonstrated a range of actA expression levels that coincided with the amount of bacterial cell-to-cell spread observed within infected monolayers. A correlation appeared to exist between levels of actA expression and the ability of L. monocytogenes to transition from uniform actin accumulation surrounding individual bacteria (actin clouds) to directional assembly and the formation of actin tails. Bacterial mutants containing deletions that most significantly altered the predicted secondary structure of the actA mRNA 5' UTR had the largest reductions in actA expression. These results suggest that the actA 5' UTR is required for maximal ActA synthesis and that a threshold level of ActA synthesis must be achieved to promote the transition from bacteria-associated actin clouds to directional actin assembly and movement.
Collapse
Affiliation(s)
- Kendy K Y Wong
- Seattle Biomedical Research Institute, Department of Pathobiology and Department of Microbiology, University of Washington, 4 Nickerson St., Seattle, WA 98109-1651, USA
| | | | | |
Collapse
|