Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Nature ; 572(7769): 387-391, 2019 08.
Article in English | MEDLINE | ID: mdl-31330531

ABSTRACT

The bacterial pathogen Legionella pneumophila creates an intracellular niche permissive for its replication by extensively modulating host-cell functions using hundreds of effector proteins delivered by its Dot/Icm secretion system1. Among these, members of the SidE family (SidEs) regulate several cellular processes through a unique phosphoribosyl ubiquitination mechanism that bypasses the canonical ubiquitination machinery2-4. The activity of SidEs is regulated by another Dot/Icm effector known as SidJ5; however, the mechanism of this regulation is not completely understood6,7. Here we demonstrate that SidJ inhibits the activity of SidEs by inducing the covalent attachment of glutamate moieties to SdeA-a member of the SidE family-at E860, one of the catalytic residues that is required for the mono-ADP-ribosyltransferase activity involved in ubiquitin activation2. This inhibition by SidJ is spatially restricted in host cells because its activity requires the eukaryote-specific protein calmodulin (CaM). We solved a structure of SidJ-CaM in complex with AMP and found that the ATP used in this reaction is cleaved at the α-phosphate position by SidJ, which-in the absence of glutamate or modifiable SdeA-undergoes self-AMPylation. Our results reveal a mechanism of regulation in bacterial pathogenicity in which a glutamylation reaction that inhibits the activity of virulence factors is activated by host-factor-dependent acyl-adenylation.


Subject(s)
Calmodulin/metabolism , Glutamic Acid/metabolism , Legionella pneumophila/enzymology , Legionella pneumophila/metabolism , Ubiquitination , ADP-Ribosylation , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Catalysis , Catalytic Domain , Coenzymes/metabolism , HEK293 Cells , Humans , Legionella pneumophila/cytology , Models, Molecular , Ubiquitin/chemistry , Ubiquitin/metabolism
2.
EMBO J ; 38(14): e100957, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31304634

ABSTRACT

The self-assembly of cellular macromolecular machines such as the bacterial flagellar motor requires the spatio-temporal synchronization of gene expression with proper protein localization and association of dozens of protein components. In Salmonella and Escherichia coli, a sequential, outward assembly mechanism has been proposed for the flagellar motor starting from the inner membrane, with the addition of each new component stabilizing the previous one. However, very little is known about flagellar disassembly. Here, using electron cryo-tomography and sub-tomogram averaging of intact Legionella pneumophila, Pseudomonas aeruginosa, and Shewanella oneidensis cells, we study flagellar motor disassembly and assembly in situ. We first show that motor disassembly results in stable outer membrane-embedded sub-complexes. These sub-complexes consist of the periplasmic embellished P- and L-rings, and bend the membrane inward while it remains apparently sealed. Additionally, we also observe various intermediates of the assembly process including an inner-membrane sub-complex consisting of the C-ring, MS-ring, and export apparatus. Finally, we show that the L-ring is responsible for reshaping the outer membrane, a crucial step in the flagellar assembly process.


Subject(s)
Bacteria/cytology , Bacterial Proteins/metabolism , Flagella/ultrastructure , Bacteria/metabolism , Bacteria/ultrastructure , Bacterial Outer Membrane/metabolism , Electron Microscope Tomography , Escherichia coli/cytology , Escherichia coli/metabolism , Escherichia coli/ultrastructure , Flagella/metabolism , Legionella pneumophila/cytology , Legionella pneumophila/metabolism , Legionella pneumophila/ultrastructure , Pseudomonas aeruginosa/cytology , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/ultrastructure , Shewanella/cytology , Shewanella/metabolism , Shewanella/ultrastructure
3.
Nature ; 533(7601): 120-4, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27049943

ABSTRACT

Signalling by ubiquitination regulates virtually every cellular process in eukaryotes. Covalent attachment of ubiquitin to a substrate is catalysed by the E1, E2 and E3 three-enzyme cascade, which links the carboxy terminus of ubiquitin to the ε-amino group of, in most cases, a lysine of the substrate via an isopeptide bond. Given the essential roles of ubiquitination in the regulation of the immune system, it is not surprising that the ubiquitination network is a common target for diverse infectious agents. For example, many bacterial pathogens exploit ubiquitin signalling using virulence factors that function as E3 ligases, deubiquitinases or as enzymes that directly attack ubiquitin. The bacterial pathogen Legionella pneumophila utilizes approximately 300 effectors that modulate diverse host processes to create a permissive niche for its replication in phagocytes. Here we demonstrate that members of the SidE effector family of L. pneumophila ubiquitinate multiple Rab small GTPases associated with the endoplasmic reticulum. Moreover, we show that these proteins are capable of catalysing ubiquitination without the need for the E1 and E2 enzymes. A putative mono-ADP-ribosyltransferase motif critical for the ubiquitination activity is also essential for the role of the SidE family in intracellular bacterial replication in a protozoan host. The E1/E2-independent ubiquitination catalysed by these enzymes is energized by nicotinamide adenine dinucleotide, which activates ubiquitin by the formation of ADP-ribosylated ubiquitin. These results establish that ubiquitination can be catalysed by a single enzyme, the activity of which does not require ATP.


Subject(s)
Bacterial Proteins/metabolism , Legionella pneumophila/chemistry , Ubiquitination , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/metabolism , Adenosine Diphosphate Ribose/metabolism , Adenosine Triphosphate , Amino Acid Motifs , Amino Acid Sequence , Bacterial Load , Biocatalysis , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Legionella pneumophila/cytology , Legionella pneumophila/enzymology , Legionella pneumophila/pathogenicity , Membrane Proteins/metabolism , Molecular Sequence Data , NAD/metabolism , Ubiquitin/chemistry , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes , Ubiquitin-Conjugating Enzymes , Virulence Factors/metabolism , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/metabolism
4.
BMC Microbiol ; 16(1): 174, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27484084

ABSTRACT

BACKGROUND: The opportunistic bacterial pathogen Legionella pneumophila uses substrate effectors of Dot/Icm type IVB secretion system (T4BSS) to accomplish survival and replication in amoebae cells and mammalian alveolar macrophages. During the conversion between its highly resistant, infectious dormant form and vigorously growing, uninfectious replicative form, L. pneumophila utilizes a complicated regulatory network in which proteolysis may play a significant role. As a highly conserved core protease, ClpP is involved in various cellular processes as well as virulence in bacteria, and has been proved to be required for the expression of transmission traits and cell division of L. pneumophila. RESULTS: The clpP-deficient L. pneumophila strain failed to replicate and was digested in the first 3 h post-infection in mammalian cells J774A.1. Further investigation demonstrates that the clpP deficient mutant strain was unable to escape the endosome-lysosomal pathway in host cells. We also found that the clpP deficient mutant strain still expresses T4BSS components, induces contact-dependent cytotoxicity and translocate effector proteins RalF and LegK2, indicating that its T4BSS was overall functional. Interestingly, we further found that the translocation of several effector proteins is significantly reduced without ClpP. CONCLUSIONS: The data indicate that ClpP plays an important role in regulating the virulence and effector translocation of Legionella pneumophila.


Subject(s)
Bacterial Proteins/genetics , Endopeptidase Clp/genetics , Legionella pneumophila/genetics , Legionella pneumophila/pathogenicity , Animals , Bacterial Proteins/metabolism , Bacterial Translocation/drug effects , Cell Line , Endocytosis/physiology , Endopeptidase Clp/deficiency , Endopeptidase Clp/metabolism , Endosomes/metabolism , Endosomes/microbiology , Guanine Nucleotide Exchange Factors/metabolism , Legionella pneumophila/cytology , Legionella pneumophila/enzymology , Lysosomes/metabolism , Lysosomes/microbiology , Macrophages/microbiology , Mice , Mutation , Phagocytosis , Sequence Deletion , Virulence
5.
Cell Microbiol ; 17(5): 607-20, 2015 May.
Article in English | MEDLINE | ID: mdl-25363599

ABSTRACT

The formation and release of outer membrane vesicles (OMVs) is a phenomenon observed in many bacteria, including Legionella pneumophila. During infection, this human pathogen primarily invades alveolar macrophages and replicates within a unique membrane-bound compartment termed Legionella-containing vacuole. In the current study, we analysed the membrane architecture of L. pneumophila OMVs by small-angle X-ray scattering and biophysically characterized OMV membranes. We investigated the interaction of L. pneumophila OMVs with model membranes by Förster resonance energy transfer and Fourier transform infrared spectroscopy. These experiments demonstrated the incorporation of OMV membrane material into liposomes composed of different eukaryotic phospholipids, revealing an endogenous property of OMVs to fuse with eukaryotic membranes. Cellular co-incubation experiments showed a dose- and time-dependent binding of fluorophore-labelled OMVs to macrophages. Trypan blue quenching experiments disclosed a rapid internalization of OMVs into macrophages at 37 and 4 °C. Purified OMVs induced tumour necrosis factor-α production in human macrophages at concentrations starting at 300 ng ml(-1). Experiments on HEK293-TLR2 and TLR4/MD-2 cell lines demonstrated a dominance of TLR2-dependent signalling pathways. In summary, we demonstrate binding, internalization and biological activity of L. pneumophila OMVs on human macrophages. Our data support OMV membrane fusion as a mechanism for the remote delivery of virulence factors to host cells.


Subject(s)
Cell Membrane/metabolism , Exosomes/metabolism , Host-Pathogen Interactions , Legionella pneumophila/physiology , Virulence Factors/metabolism , Biophysical Phenomena , Cells, Cultured , Endocytosis , Epithelial Cells/metabolism , Exosomes/chemistry , Fluorescence Resonance Energy Transfer , Humans , Legionella pneumophila/cytology , Macrophages/immunology , Macrophages/metabolism , Scattering, Small Angle , Spectroscopy, Fourier Transform Infrared , Temperature , Tumor Necrosis Factor-alpha/metabolism
6.
Environ Microbiol ; 16(2): 382-95, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23968544

ABSTRACT

Legionella pneumophila is an intracellular bacterial parasite of freshwater protozoa and an accidental waterborne human pathogen. L. pneumophila is highly pleomorphic showing several forms that differentiate within its developmental cycle. In water, L. pneumophila produces viable but non-culturable cells (VBNCCs), which remain largely uncharacterized. We produced VBNCCs from two developmental forms of L. pneumophila [stationary phase forms (SPFs) and mature infectious forms (MIFs)] in two water microcosms [double-deionized (dd) and tap water] at 45°C. In contrast with SPFs, MIFs upheld a robust ultrastructure and high viability in the two water microcosms. In dd-water, MIFs and SPFs lost their culturability faster than in tap water and did not consume their poly-ß-hydroxybutyrate inclusions. Resuscitation in Acanthamoeba castellani was only possible for VBNCCs produced from SPFs in tap water. Addition of salts to dd-water prolonged L. pneumophila culturability to tap water levels, suggesting that L. pneumophila requires ions to maintain its readiness to resume growth. VBNCCs resisted detergent lysis and digestion in the ciliate Tetrahymena, except for VBNCCs produced from SPFs in dd-water. L. pneumophila VBNCCs thus show distinct traits according to its originating developmental form and the surrounding water microcosm.


Subject(s)
Fresh Water/chemistry , Legionella pneumophila/cytology , Microbial Viability , Water Microbiology , Drinking Water/chemistry , Hydrogen-Ion Concentration , Legionella pneumophila/growth & development , Legionella pneumophila/ultrastructure , Microscopy, Electron, Transmission , Salts/chemistry , Temperature
7.
Sci Total Environ ; 927: 172410, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38608884

ABSTRACT

There is little evidence of the long-term consequences of maintaining sanitary hot water at high temperatures on the persistence of Legionella in the plumbing system. The aims of this study were to describe the persistence and genotypic variability of L. pneumophila in a hospital building with two entirely independent hot water distribution systems, and to estimate the thermotolerance of the genotypic variants by studying the quantity of VBNC L. pneumophila. Eighty isolates from 55 water samples obtained between the years 2012-2017 were analyzed. All isolates correspond to L. pneumophila serogroup 6. The isolates were discriminated in four restriction patterns by pulsed-field gel electrophoresis. In one installation, pattern A + Aa predominated, accounting for 75.8 % of samples, while the other installation exhibited pattern B as the most frequent (81.8 % of samples; p < 0.001). The mean temperature of the isolates was: 52.6 °C (pattern A + Aa) and 55.0 °C (pattern B), being significantly different. Nine strains were selected as representative among patterns to study their thermotolerance by flow-cytometry after 24 h of thermic treatment. VBNC bacteria were detected in all samples. After thermic treatment at 50 °C, 52.0 % of bacteria had an intact membrane, and after 55 °C this percentage decreased to 23.1 %. Each pattern exhibited varying levels of thermotolerance. These findings indicate that the same hospital building can be colonized with different predominant types of Legionella if it has independent hot water installations. Maintaining a minimum temperature of 50 °C at distal points of the system would allow the survival of replicative L. pneumophila. However, the presence of Legionella in hospital water networks is underestimated if culture is considered as the standard method for Legionella detection, because VBNC do not grow on culture plates. This phenomenon can carry implications for the Legionella risk management plans in hospitals that adjust their control measures based on the microbiological surveillance of water.


Subject(s)
Cross Infection , Hospitals , Legionella pneumophila , Legionnaires' Disease , Microbial Viability , Water Supply , Cross Infection/microbiology , Hot Temperature , Legionella pneumophila/classification , Legionella pneumophila/cytology , Legionella pneumophila/genetics , Legionella pneumophila/growth & development , Legionella pneumophila/isolation & purification , Thermotolerance , Time Factors , Legionnaires' Disease/microbiology , Legionnaires' Disease/transmission , Colony Count, Microbial , Humans
8.
Cell Microbiol ; 14(10): 1632-55, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22727141

ABSTRACT

Legionella, the aetiological agent responsible for Legionellosis, is an opportunistic pathogen that infects humans upon the inhalation of contaminated aerosolized water droplets. Legionella is pleomorphic and its different morphotypes exhibit varying degrees of virulence. While the filamentous forms of Legionella pneumophila (Lp) have been reported in patient samples since the first description of legionellosis, their role in disease has not been studied. Our results show that both E-cadherin and ß1 integrin receptors mediate filamentous Lp (FLp) attachment to lung epithelial cells (LECs). The activation of these receptors induces the formation of actin enriched membrane surface structures that we designated 'hooks' and 'membrane wraps'. These structures entrap the filaments on the cell surface leading to their gradual internalization through a zipper mechanism of phagocytosis dependent on actomyosin activity. The supply of E-cadherin receptors from the recycling pathway and ß1 integrins released from focal adhesion turnover are required to sustain this process. Intracellular FLp inhabits a vacuolar compartment where filaments differentiate into short rods and replicate to produce infective progeny. Here we are reporting a first description of the invasion mechanism used by FLp to invade LECs. Therefore, filamentous morphotype of Lp can induce its own uptake by LECs and has the potential ability to cause disease.


Subject(s)
Epithelial Cells/microbiology , Legionella pneumophila/pathogenicity , Phagocytosis , Actomyosin/metabolism , Bacterial Adhesion , Cadherins/metabolism , Cell Line , Humans , Integrin beta1/metabolism , Legionella pneumophila/cytology , Protein Binding
9.
BMC Infect Dis ; 13: 371, 2013 Aug 10.
Article in English | MEDLINE | ID: mdl-23937571

ABSTRACT

BACKGROUND: Legionella pneumophila (Lp) flagellin activates signaling pathways in murine macrophages that control Lp replication. Nucleotide-binding oligomerization domain (NOD) containing-like receptor (NLR) family, caspase recruitment domain (CARD) containing 4 (NLRC4) and Toll-like Receptor (TLR5) both recognize Lp flagellin in vitro, but whether these two receptors play redundant or separate functional roles in vivo is unknown. METHODS: The immune response of Nlrc4-/-, Nlrc4-/-/Tlr5-/-, and wild type C57Bl/6 mice was analyzed after in vivo infection with aerosolized Lp. RESULTS: Lp clearance from the lungs was delayed in Nlrc4-/- mice over seven days in comparison to wild type controls. Nlrc4-/-/Tlr5-/- mice had no additional defect. In contrast to TLR5, NLRC4 did not regulate recruitment of neutrophils to the lung. Although there were no differences among the mouse strains in the lung transcriptome at 4 hours, Nlrc4-/- and Nlrc4-/-Tlr5-/- mice had increased lung inflammation at 72 hours in comparison to WT. Nlrc4-/-/Tlr5-/- mice also had altered cytokine production at both 4 and 24 hours post infection when compared to wild-type (WT) and Nlrc4-/- mice. Lp replication in murine alveolar macrophages was NLRC4-dependent and TLR5-independent. CONCLUSION: These studies reveal that NLRC4 and TLR5 mediate different roles in the inflammatory response to Lp flagellin in an aerosolized infection model and NLRC4 regulates replication in both lungs and alveolar macrophages.


Subject(s)
Apoptosis Regulatory Proteins/pharmacology , Calcium-Binding Proteins/pharmacology , Legionella pneumophila/cytology , Legionnaires' Disease/microbiology , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Bacterial Proteins/metabolism , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Female , Flagellin/metabolism , Host-Pathogen Interactions , Legionella pneumophila/immunology , Legionella pneumophila/metabolism , Legionnaires' Disease/metabolism , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction , Toll-Like Receptors/genetics
10.
Appl Environ Microbiol ; 77(17): 6225-32, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21742913

ABSTRACT

A new method was developed for the rapid and sensitive detection of viable Legionella pneumophila. The method combines specific immunofluorescence (IF) staining using monoclonal antibodies with a bacterial viability marker (ChemChrome V6 cellular esterase activity marker) by means of solid-phase cytometry (SPC). IF methods were applied to the detection and enumeration of both the total and viable L. pneumophila cells in water samples. The sensitivity of the IF methods coupled to SPC was 34 cells liter(-1), and the reproducibility was good, with the coefficient of variation generally falling below 30%. IF methods were applied to the enumeration of total and viable L. pneumophila cells in 46 domestic hot water samples as well as in cooling tower water and natural water samples, such as thermal spring water and freshwater samples. Comparison with standard plate counts showed that (i) the total direct counts were always higher than the plate counts and (ii) the viable counts were higher than or close to the plate counts. With domestic hot waters, when the IF assay was combined with the viability test, SPC detected up to 3.4 × 10(3) viable but nonculturable L. pneumophila cells per liter. These direct IF methods could be a powerful tool for high-frequency monitoring of domestic hot waters or for investigating the occurrence of viable L. pneumophila in both man-made water systems and environmental water samples.


Subject(s)
Bacterial Load/methods , Legionella pneumophila/isolation & purification , Legionella pneumophila/physiology , Water Microbiology , Fluorescent Antibody Technique/methods , Legionella pneumophila/cytology , Microbial Viability , Reproducibility of Results , Sensitivity and Specificity , Staining and Labeling/methods
11.
Proc Natl Acad Sci U S A ; 105(27): 9385-90, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18599442

ABSTRACT

Analysis of phenotypes associated with specific mutants has been instrumental in determining the roles of a bacterial gene in a biological process. However, this technique does not allow one to address whether a specific gene or gene set is necessary to maintain such a process once it has been established. In the study of microbial pathogenesis, it is important but difficult to determine the temporal requirement of essential pathogenic determinants in the entire infection cycle. Here we report a Cre/loxP-based genetic system that allowed inducible deletion of specific bacterial genes after the pathogen had been phagocytosed by host cells. Using this system, we have examined the temporal requirement of the Dot/Icm type IV protein transporter of Legionella pneumophila during infection. We found that deletion of single essential dot/icm genes did not prevent the internalized bacteria from completing one cycle of intracellular replication. Further analyses indicate that the observed phenotypes were due to the high stability of the examined Dot/Icm protein. However, postinfection deletion within 8 h of the gene coding for the Dot/Icm substrate, SdhA, abolishes intracellular bacterial growth. This result indicates that the Dot/Icm transporter is important for intracellular bacterial growth after the initial biogenesis of the vacuole. Our study has provided a technical concept for analyzing the temporal requirement of specific bacterial proteins or protein complexes in infection or development.


Subject(s)
Gene Deletion , Legionella pneumophila/pathogenicity , Virulence Factors/genetics , Bacterial Infections/microbiology , Bacterial Proteins/metabolism , Cell Death/drug effects , Chromosomes, Bacterial/metabolism , Genes, Bacterial , Intracellular Space/drug effects , Intracellular Space/microbiology , Isopropyl Thiogalactoside/pharmacology , Legionella pneumophila/cytology , Legionella pneumophila/genetics , Legionella pneumophila/growth & development , Macrophages/drug effects , Macrophages/microbiology , Membrane Transport Proteins/metabolism , Phagocytosis/drug effects , Time Factors
12.
mBio ; 12(5): e0218021, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34634944

ABSTRACT

Legionella pneumophila, the causative agent of Legionnaires' disease, is a facultative intracellular pathogen that survives inside phagocytic host cells by establishing a protected replication niche, termed the "Legionella-containing vacuole" (LCV). To form an LCV and subvert pivotal host pathways, L. pneumophila employs a type IV secretion system (T4SS), which translocates more than 300 different effector proteins into the host cell. The L. pneumophila T4SS complex has been shown to span the bacterial cell envelope at the bacterial poles. However, the interactions between the T4SS and the LCV membrane are not understood. Using cryo-focused ion beam milling, cryo-electron tomography, and confocal laser scanning fluorescence microscopy, we show that up to half of the intravacuolar L. pneumophila bacteria tether their cell pole to the LCV membrane. Tethering coincides with the presence and function of T4SSs and likely promotes the establishment of distinct contact sites between T4SSs and the LCV membrane. Contact sites are characterized by indentations in the limiting LCV membrane and localize juxtaposed to T4SS machineries. The data are in agreement with the notion that effector translocation occurs by close membrane contact rather than by an extended pilus. Our findings provide novel insights into the interactions of the L. pneumophila T4SS with the LCV membrane in situ. IMPORTANCE Legionnaires' disease is a life-threatening pneumonia, which is characterized by high fever, coughing, shortness of breath, muscle pain, and headache. The disease is caused by the amoeba-resistant bacterium L. pneumophila found in various soil and aquatic environments and is transmitted to humans via the inhalation of small bacteria-containing droplets. An essential virulence factor of L. pneumophila is a so-called "type IV secretion system" (T4SS), which, by injecting a plethora of "effector proteins" into the host cell, determines pathogen-host interactions and the formation of a distinct intracellular compartment, the "Legionella-containing vacuole" (LCV). It is unknown how the T4SS makes contact to the LCV membrane to deliver the effectors. In this study, we identify indentations in the host cell membrane in close proximity to functional T4SSs localizing at the bacterial poles. Our work reveals first insights into the architecture of Legionella-LCV contact sites.


Subject(s)
Bacterial Proteins/metabolism , Legionella pneumophila/metabolism , Legionnaires' Disease/microbiology , Type IV Secretion Systems/metabolism , Vacuoles/microbiology , Bacterial Proteins/genetics , Cell Polarity , Humans , Legionella pneumophila/cytology , Legionella pneumophila/genetics , Protein Transport , Type IV Secretion Systems/genetics
13.
Clin Exp Immunol ; 162(2): 289-97, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20819093

ABSTRACT

CCL20 is a chemokine that attracts immature dendritic cells. We show that monocytes, cells characteristic of the innate immune response, infected with Mycobacterium tuberculosis express the CCL20 gene at a much higher level than the same cells infected with non-tuberculous mycobacteria. Interferon (IFN)-γ, a fundamental cytokine in the immune response to tuberculosis, strongly inhibits both the transcription and the translation of CCL20. We have also confirmed that dendritic cells are a suitable host for mycobacteria proliferation, although CCL20 does not seem to influence their intracellular multiplication rate. The chemokine, however, down-regulates the characteristic production of reactive oxygen species (ROS) induced by M. tuberculosis in monocytes, which may affect the activity of the cells. Apoptosis mediated by the mycobacteria, possibly ROS-dependent, was also inhibited by CCL20.


Subject(s)
Chemokine CCL20/metabolism , Monocytes/metabolism , Monocytes/microbiology , Mycobacterium tuberculosis/immunology , Reactive Oxygen Species/metabolism , Antibodies/immunology , Antibodies/pharmacology , Apoptosis/drug effects , Apoptosis/immunology , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Chemokine CCL20/genetics , Chemokine CCL20/immunology , Chemokine CCL20/pharmacology , Chemokines, CC/genetics , Chemotaxis/drug effects , Chemotaxis/immunology , Colony Count, Microbial , Culture Media, Conditioned/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/microbiology , Gene Expression/genetics , Humans , Interferon-gamma/pharmacology , Legionella pneumophila/cytology , Legionella pneumophila/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/microbiology , Monocytes/drug effects , Monocytes/immunology , Mycobacterium avium/cytology , Mycobacterium avium/immunology , Mycobacterium kansasii/cytology , Mycobacterium kansasii/immunology , Mycobacterium tuberculosis/cytology
14.
BMC Microbiol ; 10: 54, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20167127

ABSTRACT

BACKGROUND: Legionella pneumophila, the intracellular bacterial pathogen that causes Legionnaires' disease, exhibit characteristic transmission traits such as elevated stress tolerance, shortened length and virulence during the transition from the replication phase to the transmission phase. ClpP, the catalytic core of the Clp proteolytic complex, is widely involved in many cellular processes via the regulation of intracellular protein quality. RESULTS: In this study, we showed that ClpP was required for optimal growth of L. pneumophila at high temperatures and under several other stress conditions. We also observed that cells devoid of clpP exhibited cell elongation, incomplete cell division and compromised colony formation. Furthermore, we found that the clpP-deleted mutant was more resistant to sodium stress and failed to proliferate in the amoebae host Acanthamoeba castellanii. CONCLUSIONS: The data present in this study illustrate that the ClpP protease homologue plays an important role in the expression of transmission traits and cell division of L. pneumophila, and further suggest a putative role of ClpP in virulence regulation.


Subject(s)
Endopeptidase Clp/physiology , Legionella pneumophila/physiology , Acanthamoeba castellanii/microbiology , Amino Acid Sequence , Cell Division/genetics , Endopeptidase Clp/genetics , Hot Temperature , Legionella pneumophila/cytology , Legionella pneumophila/genetics , Legionella pneumophila/pathogenicity , Molecular Sequence Data , Mutation , Sequence Alignment , Sodium Chloride/pharmacology , Stress, Physiological/drug effects , Stress, Physiological/genetics
15.
Nat Microbiol ; 4(7): 1173-1182, 2019 07.
Article in English | MEDLINE | ID: mdl-31011165

ABSTRACT

Legionella pneumophila survives and replicates inside host cells by secreting ~300 effectors through the defective in organelle trafficking (Dot)/intracellular multiplication (Icm) type IVB secretion system (T4BSS). Here, we used complementary electron cryotomography and immunofluorescence microscopy to investigate the molecular architecture and biogenesis of the Dot/Icm secretion apparatus. Electron cryotomography mapped the location of the core and accessory components of the Legionella core transmembrane subcomplex, revealing a well-ordered central channel that opens into a large, windowed secretion chamber with an unusual 13-fold symmetry. Immunofluorescence microscopy deciphered an early-stage assembly process that begins with the targeting of Dot/Icm components to the bacterial poles. Polar targeting of this T4BSS is mediated by two Dot/Icm proteins, DotU and IcmF, that, interestingly, are homologues of the T6SS membrane complex components TssL and TssM, suggesting that the Dot/Icm T4BSS is a hybrid system. Together, these results revealed that the Dot/Icm complex assembles in an 'axial-to-peripheral' pattern.


Subject(s)
Legionella pneumophila/chemistry , Type IV Secretion Systems/metabolism , Type IV Secretion Systems/ultrastructure , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Cell Polarity , Electron Microscope Tomography , Legionella pneumophila/cytology , Legionella pneumophila/genetics , Legionella pneumophila/ultrastructure , Microscopy, Fluorescence , Mutation , Periplasm/chemistry , Periplasm/ultrastructure , Protein Multimerization , Type IV Secretion Systems/chemistry
16.
J Bacteriol ; 190(23): 7728-38, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18805971

ABSTRACT

The intracellular bacterial pathogen Legionella pneumophila follows a developmental cycle in which replicative forms (RFs) differentiate into infectious stationary-phase forms (SPFs) in vitro and in vivo into highly infectious mature intracellular forms (MIFs). The potential relationships between SPFs and MIFs remain uncharacterized. Previously we determined that L. pneumophila survives, but does not replicate, while it transiently resides (for 1 to 2 h) in food vacuoles of the freshwater ciliate Tetrahymena tropicalis before being expelled as legionellae-laden pellets. We report here that SPFs have the ability to rapidly (<1 h) and directly (in the absence of bacterial replication) differentiate into MIFs while in transit through T. tropicalis, indicating that SPFs and MIFs constitute a differentiation continuum. Mutant RFs lacking the sigma factor gene rpoS, or the response regulator gene letA, were unable to produce normal SPFs in vitro and did not fully differentiate into MIFs in vivo, further supporting the existence of a common mechanism of differentiation shared by SPFs and MIFs. Mutants with a defective Dot/Icm system morphologically differentiated into MIFs while in transit through T. tropicalis. Therefore, T. tropicalis has allowed us to unequivocally conclude that SPFs can directly differentiate into MIFs and that the Dot/Icm system is not required for differentiation, two events that could not be experimentally addressed before. The Tetrahymena model can now be exploited to study the signals that trigger MIF development in vivo and is the only replication-independent model reported to date that allows the differentiation of Dot/Icm mutants into MIFs.


Subject(s)
Legionella pneumophila/cytology , Legionella pneumophila/physiology , Tetrahymena/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/physiology , HeLa Cells , Humans , Legionella pneumophila/genetics , Mutation , Tetrahymena/ultrastructure , Vacuoles
17.
Autophagy ; 13(8): 1467-1469, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28598235

ABSTRACT

RavZ, an effector protein of pathogenic Legionella pneumophila, inhibits host macroautophagy/autophagy by deconjugation of lipidated LC3 proteins from phosphatidylethanolamine (PE) on the autophagosome membrane. The mechanism for how RavZ specifically recognizes and deconjugates the lipidated LC3s is not clear. To understand the structure-function relationship of LC3-deconjugation by RavZ, we prepared semisynthetic LC3 proteins modified with different fragments of PE or 1-hexadecanol (C16). We find that RavZ activity is strictly dependent on the conjugated PE structure and RavZ extracts LC3-PE from the membrane before deconjugation. Structural and biophysical analysis of RavZ-LC3 interactions suggest that RavZ initially recognizes LC3-PE on the membrane via its N-terminal LC3-interacting region (LIR) motif. RavZ specifically targets to autophagosome membranes by interaction with phosphatidylinositol 3-phosphate (PtdIns3P) via its C-terminal domain and association with membranes via the hydrophobic α3 helix. The α3 helix is involved in extraction of the PE moiety and docking of the fatty acid chains into the lipid-binding site of RavZ, which is related in structure to that of the phospholipid transfer protein Sec14. The LIR interaction and lipid binding facilitate subsequent proteolytic cleavage of LC3-PE. The findings reveal a novel mode of host-pathogen interaction.


Subject(s)
Autophagy , Legionella pneumophila/cytology , Autophagosomes/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Legionella pneumophila/metabolism , Models, Biological
18.
J Biosci Bioeng ; 101(6): 478-84, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16935249

ABSTRACT

Bacterial growth is influenced by several different culture conditions. Temperature is one of an essential component which regulates bacterial growth and their morphology. The influence of temperature on the length of bacteria was investigated in broth and on agar in a temperature range from 30.0 degrees C to 47.0 degrees C in 0.5 degrees C steps using a newly developed temperature gradient incubator. The incubator is able to reach a set temperature within 2 h and maintain temperature as accurate as +/-0.1 degrees C of the set temperature. Three Legionella pneumophila serotype 1 strains were incubated for 48 h in BCYE-alpha agar at various temperatures ranging from 30.0 degrees C to 48.0 degrees C and length of bacteria grown at each temperature was microscopically measured. Ability of bacteria to multiply at a given temperature was also determined. L. pneumophila serotype 1 strains ATCC 33152, a clinical isolate Okinawa 02-001 were going to elongate to longer than 100 mum when cultured higher than at 39.5 degrees C and at 41.5 degrees C, respectively. Each strain was unable to multiply when cultured higher than at 44.2 degrees C (ATCC 33152) or at 44.0 degrees C (Okinawa 02-001). Those data would provide insights for establishing regulations in terms of maintaining hot water temperature in a facility where a circulating hot water supply-system is available and contamination with Legionella spp. is likely to happen.


Subject(s)
Biofilms/growth & development , Bioreactors/microbiology , Cell Culture Techniques/methods , Legionella pneumophila/cytology , Legionella pneumophila/physiology , Adaptation, Physiological/physiology , Cell Culture Techniques/instrumentation , Cell Proliferation , Temperature
19.
Article in English | MEDLINE | ID: mdl-27734007

ABSTRACT

The water-borne pathogen Legionella pneumophila (Lp) strongly expresses the lpg1659 gene in water. This gene encodes a hypothetical protein predicted to be a membrane protein using in silico analysis. While no conserved domains were identified in Lpg1659, similar proteins are found in many Legionella species and other aquatic bacteria. RT-qPCR showed that lpg1659 is positively regulated by the alternative sigma factor RpoS, which is essential for Lp to survive in water. These observations suggest an important role of this novel protein in the survival of Lp in water. Deletion of lpg1659 did not affect cell morphology, membrane integrity or tolerance to high temperature. Moreover, lpg1659 was dispensable for growth of Lp in rich medium, and during infection of the amoeba Acanthamoeba castellanii and of THP-1 human macrophages. However, deletion of lpg1659 resulted in an early loss of culturability in water, while over-expression of this gene promoted the culturability of Lp. Therefore, these results suggest that lpg1659 is required for Lp to maintain culturability, and possibly long-term survival, in water. Since the loss of culturability observed in the absence of Lpg1659 was complemented by the addition of trace metals into water, this membrane protein is likely a transporter for acquiring essential trace metal for maintaining culturability in water and potentially in other metal-deprived conditions. Given its role in the survival of Lp in water, Lpg1659 was named LasM for Legionella aquatic survival membrane protein.


Subject(s)
Gene Expression Regulation, Bacterial , Legionella pneumophila/growth & development , Legionella pneumophila/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Promoter Regions, Genetic , Water Microbiology , Acanthamoeba castellanii/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line/microbiology , Gene Deletion , Humans , Legionella pneumophila/cytology , Legionella pneumophila/metabolism , Legionnaires' Disease/microbiology , Macrophages/microbiology , Mutation , Sigma Factor/genetics , Sigma Factor/metabolism , Survival , Survival Analysis , Temperature , Thermotolerance , Trace Elements/metabolism , Water
20.
J Colloid Interface Sci ; 459: 175-182, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26291573

ABSTRACT

Magnetic nanoparticles are the next tool in medical diagnoses and treatment in many different biomedical applications, including magnetic hyperthermia as alternative treatment for cancer and bacterial infections, as well as the disruption of biofilms. The colloidal stability of the magnetic nanoparticles in a biological environment is crucial for efficient delivery. A surface that can be easily modifiable can also improve the delivery and imaging properties of the magnetic nanoparticle by adding targeting and imaging moieties, providing a platform for additional modification. The strategy presented in this work includes multiple nitroDOPA anchors for robust binding to the surface tied to the same polymer backbone as multiple poly(ethylene oxide) chains for steric stability. This approach provides biocompatibility and enhanced stability in fetal bovine serum (FBS) and phosphate buffer saline (PBS). As a proof of concept, these polymer-particles complexes were then modified with a near infrared dye and utilized in characterizing the integration of magnetic nanoparticles in biofilms. The work presented in this manuscript describes the synthesis and characterization of a nontoxic platform for the labeling of near IR-dyes for bioimaging.


Subject(s)
Biofilms , Dihydroxyphenylalanine/chemistry , Fluorescent Dyes/chemistry , Legionella pneumophila , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Animals , Cattle , Legionella pneumophila/cytology , Legionella pneumophila/physiology , Mice , Microscopy, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL