Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Microbiol Resour Announc ; 13(1): e0089623, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38088573

ABSTRACT

The presence of thermophilic spore-forming bacteria is challenging in industrial food processing. The presented genome sequences of Aeribacillus pallidus, isolated from raw milk and cocoa powder, provide insights into how to prevent damage to minimally processed foods and products with extended shelf life, such as milk products.

2.
Microb Genom ; 9(11)2023 Nov.
Article in English | MEDLINE | ID: mdl-38031909

ABSTRACT

Horizontal gene transfer by plasmids can confer metabolic capabilities that expand a host cell's niche. Yet, it is less understood whether the coalescence of specialized catabolic functions, antibiotic resistances and metal resistances on plasmids provides synergistic benefits. In this study, we report whole-genome assembly and phenotypic analysis of five Salmonella enterica strains isolated in the 1980s from milk powder in Munich, Germany. All strains exhibited the unusual phenotype of lactose-fermentation and encoded either of two variants of the lac operon. Surprisingly, all strains encoded the mobilized colistin resistance gene 9 (mcr-9), long before the first report of this gene in the literature. In two cases, the mcr-9 gene and the lac locus were linked within a large gene island that formed an IncHI2A-type plasmid in one strain but was chromosomally integrated in the other strain. In two other strains, the mcr-9 gene was found on a large IncHI1B/IncP-type plasmid, whereas the lac locus was encoded on a separate chromosomally integrated plasmidic island. The mcr-9 sequences were identical and genomic contexts could not explain the wide range of colistin resistances exhibited by the Salmonella strains. Nucleotide variants did explain phenotypic differences in motility and exopolysaccharide production. The observed linkage of mcr-9 to lactose metabolism, an array of heavy-metal detoxification systems, and other antibiotic resistance genes may reflect a coalescence of specialized phenotypes that improve the spread of colistin resistance in dairy facilities, much earlier than previously suspected.


Subject(s)
Colistin , Salmonella enterica , Colistin/pharmacology , Salmonella enterica/genetics , Lactose , Serogroup , Drug Resistance, Bacterial/genetics , Plasmids/genetics
3.
Toxins (Basel) ; 12(11)2020 10 24.
Article in English | MEDLINE | ID: mdl-33114414

ABSTRACT

Bacillus cereus Hemolysin BL is a tripartite toxin responsible for a diarrheal type of food poisoning. Open questions remain regarding its mode of action, including the extent to which complex formation prior to cell binding contributes to pore-forming activity, how these complexes are composed, and the properties of the pores formed in the target cell membrane. Distinct complexes of up to 600 kDa were found on native gels, whose structure and size were primarily defined by Hbl B. Hbl L1 and L2 were also identified in these complexes using Western blotting and an LC-MS approach. LC-MS also revealed that many other proteins secreted by B. cereus exist in complexes. Further, a decrease of toxic activity at temperatures ≥60 °C was shown, which was unexpectedly restored at higher temperatures. This could be attributed to a release of Hbl B monomers from tight complexation, resulting in enhanced cell binding. In contrast, Hbl L1 was rather susceptible to heat, while heat treatment of Hbl L2 seemed not to be crucial. Furthermore, Hbl-induced pores had a rather small single-channel conductance of around 200 pS and a probable channel diameter of at least 1 nm on planar lipid bilayers. These were highly instable and had a limited lifetime, and were also slightly cation-selective. Altogether, this study provides astonishing new insights into the complex mechanism of Hbl pore formation, as well as the properties of the pores.


Subject(s)
Bacillus cereus , Bacterial Proteins , Hemolysin Proteins , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/toxicity , Cell Survival , Chlorocebus aethiops , Hemolysin Proteins/chemistry , Hemolysin Proteins/toxicity , Lipid Bilayers , Porosity , Vero Cells
4.
Article in English | MEDLINE | ID: mdl-30863829

ABSTRACT

We report here the draft genome sequence of Acinetobacter junii MHI21018, isolated in 2009 from bovine colostrum. The draft genome sequence is composed of 3,267,995 bp, has a GC content of 38.54%, and was assembled into 114 contigs (contig size, >500 bp) with an N 50 value of 72,566 bp.

5.
Nat Microbiol ; 4(2): 362-374, 2019 02.
Article in English | MEDLINE | ID: mdl-30531979

ABSTRACT

Host recognition of microbial components is essential in mediating an effective immune response. Cytosolic bacteria must secure entry into the host cytoplasm to facilitate replication and, in doing so, liberate microbial ligands that activate cytosolic innate immune sensors and the inflammasome. Here, we identified a multicomponent enterotoxin, haemolysin BL (HBL), that engages activation of the inflammasome. This toxin is highly conserved among the human pathogen Bacillus cereus. The three subunits of HBL bind to the cell membrane in a linear order, forming a lytic pore and inducing activation of the NLRP3 inflammasome, secretion of interleukin-1ß and interleukin-18, and pyroptosis. Mechanistically, the HBL-induced pore results in the efflux of potassium and triggers the activation of the NLRP3 inflammasome. Furthermore, HBL-producing B. cereus induces rapid inflammasome-mediated mortality. Pharmacological inhibition of the NLRP3 inflammasome using MCC950 prevents B. cereus-induced lethality. Overall, our results reveal that cytosolic sensing of a toxin is central to the innate immune recognition of infection. Therapeutic modulation of this pathway enhances host protection against deadly bacterial infections.


Subject(s)
Bacillus cereus/immunology , Bacterial Proteins/immunology , Enterotoxins/immunology , Hemolysin Proteins/immunology , Inflammasomes/metabolism , Inflammation , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Bacterial Proteins/metabolism , Cell Membrane/metabolism , Cell Membrane/pathology , Cells, Cultured , Culture Media, Conditioned , Enterotoxins/chemistry , Enterotoxins/metabolism , Female , Hemolysin Proteins/metabolism , Immunity, Innate , Macrophages/immunology , Macrophages/pathology , Macrophages/ultrastructure , Male , Mice , Mice, Mutant Strains , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Potassium/metabolism , Protein Multimerization , Pyroptosis , Survival Analysis
6.
Sci Rep ; 7(1): 17821, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29259308

ABSTRACT

The human pathogen L. monocytogenes and the animal pathogen L. ivanovii, together with four other species isolated from symptom-free animals, form the "Listeria sensu stricto" clade. The members of the second clade, "Listeria sensu lato", are believed to be solely environmental bacteria without the ability to colonize mammalian hosts. To identify novel determinants that contribute to infection by L. monocytogenes, the causative agent of the foodborne disease listeriosis, we performed a genome comparison of the two clades and found 151 candidate genes that are conserved in the Listeria sensu stricto species. Two factors were investigated further in vitro and in vivo. A mutant lacking an ATP-binding cassette transporter exhibited defective adhesion and invasion of human Caco-2 cells. Using a mouse model of foodborne L. monocytogenes infection, a reduced number of the mutant strain compared to the parental strain was observed in the small intestine and the liver. Another mutant with a defective 1,2-propanediol degradation pathway showed reduced persistence in the stool of infected mice, suggesting a role of 1,2-propanediol as a carbon and energy source of listeriae during infection. These findings reveal the relevance of novel factors for the colonization process of L. monocytogenes.


Subject(s)
Listeria monocytogenes/genetics , Listeriosis/microbiology , ATP-Binding Cassette Transporters/genetics , Animals , Caco-2 Cells , Cell Line, Tumor , Female , Foodborne Diseases/genetics , Foodborne Diseases/microbiology , Humans , Listeriosis/genetics , Mice , Mice, Inbred BALB C , Virulence/genetics
7.
Front Microbiol ; 8: 1826, 2017.
Article in English | MEDLINE | ID: mdl-28979257

ABSTRACT

The ubiquitous and opportunistic pathogen Cronobacter sakazakii is responsible for severe meningitis, sepsis, and necrotizing enterocolitis in neonates and infants associated with ingestion of contaminated powdered infant formula (PIF). The current ISO method for isolation and detection of Cronobacter spp. is laborious, time-consuming and expensive. In this study, a multiplexed lateral flow test strip was developed to rapidly detect and simultaneously serotype O1 and O2 C. sakazakii serotypes. The assay is based on two monoclonal antibodies (MAb) that specifically bind to the lipopolysaccharides (LPS) of these pathogens. The test strip provides results very quickly; C. sakazakii could be detected in pure culture within 15 min with a sensitivity of 107 CFU/ml. After non-selective enrichment for 18 h as low as one Cronobacter cell per g PIF could be detected. Moreover, the established lateral flow assay (LFA) offers excellent specificity showing no cross-reactivity with other C. sakazakii serotypes, Cronobacter species or Enterobacteriaceae tested. These characteristics, together with several advantages such as speed, simplicity in performance, low analysis cost, and no requirement of specialized skills or sophisticated equipment make the developed multiplexed LFA suitable for reliable detection and serotyping of C. sakazakii serotypes O1 and O2.

8.
Toxins (Basel) ; 9(9)2017 09 16.
Article in English | MEDLINE | ID: mdl-28926954

ABSTRACT

Haemolysin BL is an important virulence factor regarding the diarrheal type of food poisoning caused by Bacillus cereus. However, the pathogenic importance of this three-component enterotoxin is difficult to access, as nearly all natural B. cereus culture supernatants additionally contain the highly cytotoxic Nhe, the second three-component toxin involved in the aetiology of B. cereus-induced food-borne diseases. To better address the toxic properties of the Hbl complex, a system for overexpression and purification of functional, cytotoxic, recombinant (r)Hbl components L2, L1 and B from E. coli was established and an nheABC deletion mutant was constructed from B. cereus reference strain F837/76. Furthermore, 35 hybridoma cell lines producing monoclonal antibodies (mAbs) against Hbl L2, L1 and B were generated. While mAbs 1H9 and 1D8 neutralized Hbl toxicity and thus, represent important tools for future investigations of the mode-of-action of Hbl on the target cell surface, mAb 1D7, in contrast, even enhanced Hbl toxicity by supporting the binding of Hbl B to the cell surface. By using the specific mAbs in Dot blots, indirect and hybrid sandwich enzyme immuno assays (EIAs), complex formation between Hbl L1 and B, as well as L1 and L2 in solution could be shown for the first time. Surface plasmon resonance experiments with the rHbl components confirmed these results with KD values of 4.7 × 10-7 M and 1.5 × 10-7 M, respectively. These findings together with the newly created tools lay the foundation for the detailed elucidation of the molecular mode-of-action of the highly complex three-component Hbl toxin.


Subject(s)
Bacterial Proteins , Hemolysin Proteins , Recombinant Proteins , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Bacillus cereus/genetics , Bacillus cereus/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Proteins/toxicity , Cell Line , Cell Survival/drug effects , Chlorocebus aethiops , Escherichia coli/genetics , Female , Hemolysin Proteins/genetics , Hemolysin Proteins/immunology , Hemolysin Proteins/metabolism , Hemolysin Proteins/toxicity , Mice, Inbred BALB C , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/toxicity , Solutions , Vero Cells
9.
Genes (Basel) ; 8(1)2016 Dec 28.
Article in English | MEDLINE | ID: mdl-28036056

ABSTRACT

Multidrug resistant microorganisms are forecast to become the single biggest challenge to medical care in the 21st century. Over the last decades, members of the genus Acinetobacter have emerged as bacterial opportunistic pathogens, in particular as challenging nosocomial pathogens because of the rapid evolution of antimicrobial resistances. Although we lack fundamental biological insight into virulence mechanisms, an increasing number of researchers are working to identify virulence factors and to study antibiotic resistance. Here, we review current knowledge regarding the regulation of virulence genes and antibiotic resistance in Acinetobacter baumannii. A survey of the two-component systems AdeRS, BaeSR, GacSA and PmrAB explains how each contributes to antibiotic resistance and virulence gene expression, while BfmRS regulates cell envelope structures important for pathogen persistence. A. baumannii uses the transcription factors Fur and Zur to sense iron or zinc depletion and upregulate genes for metal scavenging as a critical survival tool in an animal host. Quorum sensing, nucleoid-associated proteins, and non-classical transcription factors such as AtfA and small regulatory RNAs are discussed in the context of virulence and antibiotic resistance.

10.
Appl Environ Microbiol ; 82(8): 2300-2311, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26850303

ABSTRACT

Cronobacter sakazakii is a foodborne pathogen associated with rare but often lethal infections in neonates. Powdered infant formula (PIF) represents the most frequent source of infection. Out of the identified serotypes (O1 to O7), O1, O2, and O3 are often isolated from clinical and PIF samples. Serotype-specific monoclonal antibodies (MAbs) suitable for application in enzyme immunoassays (EIAs) for the rapid detection of C. sakazakii have not yet been developed. In this study, we created specific MAbs with the ability to bind toC. sakazakii of serotypes O1, O2, and O3. Characterization by indirect EIAs, immunofluorescence, motility assays, and immunoblotting identified lipopolysaccharide (LPS) and exopolysaccharide (EPS) as the antigenic determinants of the MAbs. The established sandwich EIAs were highly sensitive and were able to detect between 2 × 10(3)and 9 × 10(6)CFU/ml. Inclusivity tests confirmed that 93% of serotype O1 strains, 100% of O2 strains, and 87% of O3 strains were detected at low cell counts. No cross-reactivity with >100 strains of Cronobacter spp. and other Enterobacter iaceae was observed, except for that with C. sakazakii serotype O3 and Cronobacter muytjensii serotype O1. Moreover, the sandwich EIAs detected C. sakazakii in PIF samples artificially contaminated with 1 to 10 bacterial cells per 10 g of sample after 15 h of preenrichment. The use of these serotype-specific MAbs not only allows the reliable detection of C. sakazakii strains but also enables simultaneous serotyping in a simple sandwich EIA method.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Cronobacter sakazakii/classification , Cronobacter sakazakii/isolation & purification , Serogroup , Serotyping/methods , Humans , Immunoenzyme Techniques/methods , Sensitivity and Specificity , Time Factors
11.
Infect Immun ; 83(3): 876-87, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25534937

ABSTRACT

Cronobacter turicensis is an opportunistic foodborne pathogen that can cause a rare but sometimes lethal infection in neonates. Little is known about the virulence mechanisms and intracellular lifestyle of this pathogen. In this study, we developed an IgG monoclonal antibody (MAb; MAb 2G4) that specifically recognizes the O1 antigen of C. turicensis cells. The antilipopolysaccharide antibody bound predominantly monovalently to the O antigen and reduced bacterial growth without causing cell agglutination. Furthermore, binding of the antibody to the O1 antigen of C. turicensis cells caused a significant reduction of the membrane potential which is required to energize flagellar rotation, accompanied by a decreased flagellum-based motility. These results indicate that binding of IgG to the O antigen of C. turicensis causes a direct antimicrobial effect. In addition, this feature of the antibody enabled new insight into the pathogenicity of C. turicensis. In a tissue culture infection model, pretreatment of C. turicensis with MAb 2G4 showed no difference in adhesion to human epithelial cells, whereas invasion of bacteria into Caco-2 cells was significantly inhibited.


Subject(s)
Antibodies, Bacterial/biosynthesis , Antibodies, Monoclonal/biosynthesis , Cronobacter/drug effects , Immunoglobulin G/biosynthesis , O Antigens/immunology , Animals , Antibodies, Bacterial/isolation & purification , Antibodies, Bacterial/pharmacology , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Caco-2 Cells , Cell Adhesion/drug effects , Cronobacter/chemistry , Cronobacter/immunology , Cronobacter/pathogenicity , Female , Flagella/drug effects , Flow Cytometry , Humans , Hybridomas/chemistry , Hybridomas/immunology , Immunoglobulin G/isolation & purification , Immunoglobulin G/pharmacology , Membrane Potentials/drug effects , Mice , Mice, Inbred BALB C , Movement/drug effects , O Antigens/chemistry
12.
Article in English | MEDLINE | ID: mdl-25405102

ABSTRACT

Intracellular bacterial pathogens (IBPs) are dependent on various nutrients provided by the host cells. Different strategies may therefore be necessary to adapt the intracellular metabolism of IBPs to the host cells. The specific carbon sources, the catabolic pathways participating in their degradation, and the biosynthetic performances of IBPs are still poorly understood. In this report, we have exploited the technique of (13)C-isotopologue profiling to further study the carbon metabolism of Listeria monocytogenes by using the EGDe wild-type strain and mutants (defective in the uptake and/or catabolism of various carbon compounds) replicating in J774A.1 macrophages. For this goal, the infected macrophages were cultivated in the presence of [1,2-(13)C2]glucose, [U-(13)C3]glycerol, [U-(13)C3]pyruvate, [U-(13)C3]lactate, or a mix of [U-(13)C]amino acids. GC/MS-based isotopologue profiling showed efficient utilization of amino acids, glucose 6-phosphate, glycerol, and (at a low extent) also of lactate but not of pyruvate by the IBPs. Most amino acids imported from the host cells were directly used for bacterial protein biosynthesis and hardly catabolized. However, Asp was de novo synthesized by the IBPs and not imported from the host cell. As expected, glycerol was catabolized via the ATP-generating lower part of the glycolytic pathway, but apparently not used for gluconeogenesis. The intermediates generated from glucose 6-phosphate in the upper part of the glycolytic pathway and the pentose phosphate shunt likely serve primarily for anabolic purposes (probably for the biosynthesis of cell wall components and nucleotides). This bipartite bacterial metabolism which involves at least two major carbon substrates-glycerol mainly for energy supply and glucose 6-phosphate mainly for indispensible anabolic performances-may put less nutritional stress on the infected host cells, thereby extending the lifespan of the host cells to the benefit of the IBPs.


Subject(s)
Carbon/metabolism , Listeria monocytogenes/metabolism , Macrophages/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalysis , Cell Line , Gluconeogenesis , Glucose-6-Phosphate/metabolism , Glycerol/metabolism , Host-Pathogen Interactions , Lactic Acid/metabolism , Listeria monocytogenes/genetics , Listeria monocytogenes/immunology , Macrophages/immunology , Mice , Mutation , Pyruvic Acid/metabolism
13.
BMC Genomics ; 14: 285, 2013 Apr 27.
Article in English | MEDLINE | ID: mdl-23622257

ABSTRACT

BACKGROUND: The saprophytic pathogen Listeria monocytogenes has to cope with a variety of acidic habitats during its life cycle. The impact of low-temperature coupled with pH decrease for global gene expression and subsequent virulence properties, however, has not been elucidated. RESULTS: qRT-PCR revealed for the first time a transient, acid triggered prfA induction of approximately 4-fold, 5.7-fold, 7-fold and 9.3-fold 60 to 90 min after acid shock of L. monocytogenes at 37°C, 25°C, 18°C, and 10°C, respectively. Comparable data were obtained for seven different L. monocytogenes strains, demonstrating that prfA induction under these conditions is a general response of L. monocytogenes. Transcriptome analysis revealed that the in vivo-relevant genes bsh, clpP, glpD, hfq, inlA, inlB, inlE, lisR, and lplA1 as well as many other genes with a putative role during infection are transiently induced upon acid shock conducted at 25°C and 37°C. Twenty-five genes repressed upon acid shock are known to be down regulated during intracellular growth or by virulence regulators. These data were confirmed by qRT-PCR of twelve differentially regulated genes and by the identification of acid shock-induced genes influenced by σB. To test if up regulation of virulence genes at temperatures below 37°C correlates with pathogenicity, the capacity of L. monocytogenes to invade epithelial cells after acid shock at 25°C was measured. A 12-fold increased number of intracellular bacteria was observed (acid shock, t = 60 min) that was reduced after adaptation to the level of the unshocked control. This increased invasiveness was shown to be in line with the induction of inlAB. Using a nematode infection assay, we demonstrated that Caenorhabditis elegans fed with acid-shocked L. monocytogenes exhibits a shorter time to death of 50% (TD50) of the worms (6.4 days) compared to infection with unshocked bacteria (TD50 = 10.2 days). CONCLUSIONS: PrfA and other listerial virulence genes are induced by an inorganic acid in a temperature-dependent manner. The data presented here suggest that low pH serves as a trigger for listerial pathogenicity at environmental temperatures.


Subject(s)
Bacterial Proteins/biosynthesis , Listeria monocytogenes/pathogenicity , Peptide Termination Factors/biosynthesis , Virulence/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Caenorhabditis elegans/microbiology , Cold Temperature , Epithelial Cells/microbiology , Gene Expression Regulation, Bacterial , Hydrogen-Ion Concentration , Listeria monocytogenes/genetics , Membrane Proteins/biosynthesis , Peptide Termination Factors/genetics , Sigma Factor/physiology , Temperature
14.
BMC Genomics ; 11: 573, 2010 Oct 18.
Article in English | MEDLINE | ID: mdl-20955543

ABSTRACT

BACKGROUND: The human pathogen Listeria monocytogenes resides and proliferates within the cytoplasm of epithelial cells. While the virulence factors essentially contributing to this step of the infection cycle are well characterized, the set of listerial genes contributing to intracellular replication remains to be defined on a genome-wide level. RESULTS: A comprehensive library of L. monocytogenes strain EGD knockout mutants was constructed upon insertion-duplication mutagenesis, and 1491 mutants were tested for their phenotypes in rich medium and in a Caco-2 cell culture assay. Following sequencing of the plasmid insertion site, 141 different genes required for invasion of and replication in Caco-2 cells were identified. Ten in-frame deletion mutants were constructed that confirmed the data. The genes with known functions are mainly involved in cellular processes including transport, in the intermediary metabolism of sugars, nucleotides and lipids, and in information pathways such as regulatory functions. No function could be ascribed to 18 genes, and a counterpart of eight genes is missing in the apathogenic species L. innocua. Mice infection studies revealed the in vivo requirement of IspE (Lmo0190) involved in mevalonate synthesis, and of the novel ABC transporter Lmo0135-0137 associated with cysteine transport. Based on the data of this genome-scale screening, an extreme pathway and elementary mode analysis was applied that demonstrates the critical role of glycerol and purine metabolism, of fucose utilization, and of the synthesis of glutathione, aspartate semialdehyde, serine and branched chain amino acids during intracellular replication of L. monocytogenes. CONCLUSION: The combination of a genetic screening and a modelling approach revealed that a series of transporters help L. monocytogenes to overcome a putative lack of nutrients within cells, and that a high metabolic flexibility contributes to the intracellular replication of this pathogen.


Subject(s)
Genetic Testing/methods , Intracellular Space/metabolism , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Models, Biological , Mutation/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Caco-2 Cells , DNA Replication/genetics , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Genes, Bacterial/genetics , Humans , Listeria monocytogenes/pathogenicity , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Reading Frames/genetics , Reproducibility of Results , Sequence Deletion/genetics , Virulence/genetics
15.
J Bacteriol ; 192(7): 1774-84, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20097852

ABSTRACT

The human pathogen L. monocytogenes is a facultatively intracellular bacterium that survives and replicates in the cytosol of many mammalian cells. The listerial metabolism, especially under intracellular conditions, is still poorly understood. Recent studies analyzed the carbon metabolism of L. monocytogenes by the (13)C isotopologue perturbation method in a defined minimal medium containing [U-(13)C(6)]glucose. It was shown that these bacteria produce oxaloacetate mainly by carboxylation of pyruvate due to an incomplete tricarboxylic acid cycle. Here, we report that a pycA insertion mutant defective in pyruvate carboxylase (PYC) still grows, albeit at a reduced rate, in brain heart infusion (BHI) medium but is unable to multiply in a defined minimal medium with glucose or glycerol as a carbon source. Aspartate and glutamate of the pycA mutant, in contrast to the wild-type strain, remain unlabeled when [U-(13)C(6)]glucose is added to BHI, indicating that the PYC-catalyzed carboxylation of pyruvate is the predominant reaction leading to oxaloacetate in L. monocytogenes. The pycA mutant is also unable to replicate in mammalian cells and exhibits high virulence attenuation in the mouse sepsis model.


Subject(s)
Bacterial Proteins/metabolism , Carbon/metabolism , Listeria monocytogenes/enzymology , Listeria monocytogenes/metabolism , Pyruvate Carboxylase/metabolism , Animals , Aspartic Acid/metabolism , Bacterial Proteins/genetics , Carbon Isotopes/metabolism , Cell Line , Culture Media/chemistry , Epithelial Cells/microbiology , Female , Gene Deletion , Glucose/metabolism , Glutamic Acid/metabolism , Humans , Listeria monocytogenes/growth & development , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mutagenesis, Insertional , Oxaloacetic Acid/metabolism , Pyruvate Carboxylase/genetics , Pyruvic Acid/metabolism , Sepsis/microbiology , Virulence
16.
J Bacteriol ; 191(7): 2218-27, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19181806

ABSTRACT

Thiamine pyrophosphate is an essential cofactor involved in central metabolism and amino acid biosynthesis and is derived from thiamine (vitamin B(1)). The extent to which this metabolite is available to bacterial pathogens replicating within host cells is still little understood. Growth studies using modified minimal Welshimer's broth (mMWB) supplemented with thiamine or the thiamine precursor hydroxymethylpyrimidine (HMP) showed that Listeria monocytogenes, in agreement with bioinformatic prediction, is able to synthesize thiamine only in the presence of HMP. This appears to be due to a lack of ThiC, which is involved in HMP synthesis. The knockout of thiD (lmo0317), which probably catalyzes the phosphorylation of HMP, inhibited growth in mMWB supplemented with HMP and reduced the replication rate of L. monocytogenes in epithelial cells. Mutation of a predicted thiamine transporter gene, lmo1429, led to reduced proliferation of L. monocytogenes in mMWB containing thiamine or thiamine phosphates and also within epithelial cells but had no influence on the expression of the virulence factors Hly and ActA. The toxic thiamine analogue pyrithiamine inhibited growth of wild-type strain EGD but not of the transporter mutant EGDDeltathiT. We also demonstrated that ThiT binds thiamine, a finding compatible with ThiT acting as the substrate-binding component of a multimeric thiamine transporter complex. These data provide experimental evidence that Lmo1429 homologs including Bacillus YuaJ are necessary for thiamine transport in gram-positive bacteria and are therefore proposed to be annotated "ThiT." Taken together, these data indicate that concurrent thiamine uptake and biosynthesis of thiamine precursors is a strategy of L. monocytogenes and possibly other facultative intracellular pathogens to enable proliferation within the cytoplasm.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/growth & development , Listeriosis/microbiology , Membrane Transport Proteins/metabolism , Thiamine/biosynthesis , Bacterial Proteins/genetics , Biological Transport , Caco-2 Cells , Epithelial Cells/microbiology , Humans , Imino Furanoses/metabolism , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Membrane Transport Proteins/genetics , Protein Binding
17.
J Bacteriol ; 188(2): 556-68, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16385046

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.


Subject(s)
Genes, Bacterial , Listeria monocytogenes/physiology , Animals , Carbon/metabolism , Cytosol/microbiology , Epithelial Cells/microbiology , Ethanolamine/metabolism , Gene Expression Profiling , Genomics , Glucose/metabolism , Glycerol/metabolism , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Mutagenesis , Nitrogen/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...