Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Publication year range
1.
Nucleic Acids Res ; 51(14): 7631-7648, 2023 08 11.
Article in English | MEDLINE | ID: mdl-37326020

ABSTRACT

Virulence factors enable pathogenic bacteria to infect host cells, establish infection, and contribute to disease progressions. In Gram-positive pathogens such as Staphylococcus aureus (Sa) and Enterococcus faecalis (Ef), the pleiotropic transcription factor CodY plays a key role in integrating metabolism and virulence factor expression. However, to date, the structural mechanisms of CodY activation and DNA recognition are not understood. Here, we report the crystal structures of CodY from Sa and Ef in their ligand-free form and their ligand-bound form complexed with DNA. Binding of the ligands-branched chain amino acids and GTP-induces conformational changes in the form of helical shifts that propagate to the homodimer interface and reorient the linker helices and DNA binding domains. DNA binding is mediated by a non-canonical recognition mechanism dictated by DNA shape readout. Furthermore, two CodY dimers bind to two overlapping binding sites in a highly cooperative manner facilitated by cross-dimer interactions and minor groove deformation. Our structural and biochemical data explain how CodY can bind a wide range of substrates, a hallmark of many pleiotropic transcription factors. These data contribute to a better understanding of the mechanisms underlying virulence activation in important human pathogens.


Subject(s)
Bacterial Proteins , Enterococcus faecalis , Repressor Proteins , Staphylococcus aureus , Humans , Bacterial Proteins/metabolism , DNA/chemistry , Gene Expression Regulation, Bacterial , Repressor Proteins/metabolism , Transcription Factors/metabolism , Virulence , Virulence Factors , Staphylococcus aureus/chemistry , Enterococcus faecalis/chemistry
2.
Proc Natl Acad Sci U S A ; 119(43): e2210912119, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36252016

ABSTRACT

The alarming rise of multidrug-resistant Gram-positive bacteria has precipitated a healthcare crisis, necessitating the development of new antimicrobial therapies. Here we describe a new class of antibiotics based on a ring-fused 2-pyridone backbone, which are active against vancomycin-resistant enterococci (VRE), a serious threat as classified by the Centers for Disease Control and Prevention, and other multidrug-resistant Gram-positive bacteria. Ring-fused 2-pyridone antibiotics have bacteriostatic activity against actively dividing exponential phase enterococcal cells and bactericidal activity against nondividing stationary phase enterococcal cells. The molecular mechanism of drug-induced killing of stationary phase cells mimics aspects of fratricide observed in enterococcal biofilms, where both are mediated by the Atn autolysin and the GelE protease. In addition, combinations of sublethal concentrations of ring-fused 2-pyridones and standard-of-care antibiotics, such as vancomycin, were found to synergize to kill clinical strains of VRE. Furthermore, a broad range of antibiotic resistant Gram-positive pathogens, including those responsible for the increasing incidence of antibiotic resistant healthcare-associated infections, are susceptible to this new class of 2-pyridone antibiotics. Given the broad antibacterial activities of ring-fused 2-pyridone compounds against Gram-positive (GmP) bacteria we term these compounds GmPcides, which hold promise in combating the rising tide of antibiotic resistant Gram-positive pathogens.


Subject(s)
Gram-Positive Bacteria , Pyridones , Vancomycin-Resistant Enterococci , Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial , Gram-Positive Bacteria/drug effects , Microbial Sensitivity Tests , N-Acetylmuramoyl-L-alanine Amidase/pharmacology , Pyridones/pharmacology , Vancomycin/pharmacology , Vancomycin-Resistant Enterococci/drug effects
3.
PLoS Pathog ; 18(3): e1010213, 2022 03.
Article in English | MEDLINE | ID: mdl-35275969

ABSTRACT

The general stress response (GSR) in Listeria monocytogenes plays a critical role in the survival of this pathogen in the host gastrointestinal tract. The GSR is regulated by the alternative sigma factor B (σB), whose role in protection against acid stress is well established. Here, we investigated the involvement of the stressosome, a sensory hub, in transducing low pH signals to induce the GSR. Mild acid shock (15 min at pH 5.0) activated σB and conferred protection against a subsequent lethal pH challenge. A mutant strain where the stressosome subunit RsbR1 was solely present retained the ability to induce σB activity at pH 5.0. The role of stressosome phosphorylation in signal transduction was investigated by mutating the putative phosphorylation sites in the core stressosome proteins RsbR1 (rsbR1-T175A, -T209A, -T241A) and RsbS (rsbS-S56A), or the stressosome kinase RsbT (rsbT-N49A). The rsbS S56A and rsbT N49A mutations abolished the response to low pH. The rsbR1-T209A and rsbR1-T241A mutants displayed constitutive σB activity. Mild acid shock upregulates invasion genes inlAB and stimulates epithelial cell invasion, effects that were abolished in mutants with an inactive or overactive stressosome. Overall, the results show that the stressosome is required for acid-induced activation of σB in L. monocytogenes. Furthermore, they show that RsbR1 can function independently of its paralogues and signal transduction requires RsbT-mediated phosphorylation of RsbS on S56 and RsbR1 on T209 but not T175. These insights shed light on the mechanisms of signal transduction that activate the GSR in L. monocytogenes in response to acidic environments, and highlight the role this sensory process in the early stages of the infectious cycle.


Subject(s)
Listeria monocytogenes , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Listeria monocytogenes/metabolism , Phosphorylation , Sigma Factor/genetics , Sigma Factor/metabolism , Signal Transduction/physiology
4.
PLoS Pathog ; 18(1): e1010166, 2022 01.
Article in English | MEDLINE | ID: mdl-35007292

ABSTRACT

A hallmark of Listeria (L.) monocytogenes pathogenesis is bacterial escape from maturing entry vacuoles, which is required for rapid bacterial replication in the host cell cytoplasm and cell-to-cell spread. The bacterial transcriptional activator PrfA controls expression of key virulence factors that enable exploitation of this intracellular niche. The transcriptional activity of PrfA within infected host cells is controlled by allosteric coactivation. Inhibitory occupation of the coactivator site has been shown to impair PrfA functions, but consequences of PrfA inhibition for L. monocytogenes infection and pathogenesis are unknown. Here we report the crystal structure of PrfA with a small molecule inhibitor occupying the coactivator site at 2.0 Å resolution. Using molecular imaging and infection studies in macrophages, we demonstrate that PrfA inhibition prevents the vacuolar escape of L. monocytogenes and enables extensive bacterial replication inside spacious vacuoles. In contrast to previously described spacious Listeria-containing vacuoles, which have been implicated in supporting chronic infection, PrfA inhibition facilitated progressive clearance of intracellular L. monocytogenes from spacious vacuoles through lysosomal degradation. Thus, inhibitory occupation of the PrfA coactivator site facilitates formation of a transient intravacuolar L. monocytogenes replication niche that licenses macrophages to effectively eliminate intracellular bacteria. Our findings encourage further exploration of PrfA as a potential target for antimicrobials and highlight that intra-vacuolar residence of L. monocytogenes in macrophages is not inevitably tied to bacterial persistence.


Subject(s)
Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Macrophages/microbiology , Vacuoles/microbiology , Virulence/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL
5.
Cell ; 139(4): 770-9, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19914169

ABSTRACT

Riboswitches are RNA elements acting in cis, controlling expression of their downstream genes through a metabolite-induced alteration of their secondary structure. Here, we demonstrate that two S-adenosylmethionine (SAM) riboswitches, SreA and SreB, can also function in trans and act as noncoding RNAs in Listeria monocytogenes. SreA and SreB control expression of the virulence regulator PrfA by binding to the 5'-untranslated region of its mRNA. Absence of the SAM riboswitches SreA and SreB increases the level of PrfA and virulence gene expression in L. monocytogenes. Thus, the impact of the SAM riboswitches on PrfA expression highlights a link between bacterial virulence and nutrient availability. Together, our results uncover an unexpected role for riboswitches and a distinct class of regulatory noncoding RNAs in bacteria.


Subject(s)
Bacterial Proteins/genetics , Listeria monocytogenes/genetics , Peptide Termination Factors/genetics , Regulatory Sequences, Ribonucleic Acid , 5' Untranslated Regions , Base Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Molecular Sequence Data , Temperature , Virulence
6.
Nucleic Acids Res ; 50(19): 11285-11300, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36300626

ABSTRACT

HflX is a ubiquitous bacterial GTPase that splits and recycles stressed ribosomes. In addition to HflX, Listeria monocytogenes contains a second HflX homolog, HflXr. Unlike HflX, HflXr confers resistance to macrolide and lincosamide antibiotics by an experimentally unexplored mechanism. Here, we have determined cryo-EM structures of L. monocytogenes HflXr-50S and HflX-50S complexes as well as L. monocytogenes 70S ribosomes in the presence and absence of the lincosamide lincomycin. While the overall geometry of HflXr on the 50S subunit is similar to that of HflX, a loop within the N-terminal domain of HflXr, which is two amino acids longer than in HflX, reaches deeper into the peptidyltransferase center. Moreover, unlike HflX, the binding of HflXr induces conformational changes within adjacent rRNA nucleotides that would be incompatible with drug binding. These findings suggest that HflXr confers resistance using an allosteric ribosome protection mechanism, rather than by simply splitting and recycling antibiotic-stalled ribosomes.


Subject(s)
Listeria monocytogenes , Listeria monocytogenes/genetics , GTP-Binding Proteins/genetics , Drug Resistance, Microbial , Ribosomes/genetics , Ribosomes/metabolism , Lincosamides/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism
7.
Infect Immun ; 91(6): e0057122, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37125941

ABSTRACT

Listeria monocytogenes is a bacterial pathogen capable of causing severe infections but also thriving outside the host. To respond to different stress conditions, L. monocytogenes mainly utilizes the general stress response regulon, which largely is controlled by the alternative sigma factor Sigma B (SigB). In addition, SigB is important for virulence gene expression and infectivity. Upon encountering stress, a large multicomponent protein complex known as the stressosome becomes activated, ultimately leading to SigB activation. RsbX is a protein needed to reset a "stressed" stressosome and prevent unnecessary SigB activation in nonstressed conditions. Consequently, absence of RsbX leads to constitutive activation of SigB even without prevailing stress stimulus. To further examine the involvement of SigB in the virulence of this pathogen, we investigated whether a strain with constitutively active SigB would be affected in virulence factor expression and/or infectivity in cultured cells and in a chicken embryo infection model. Our results suggest that increased SigB activity does not substantially alter virulence gene expression compared with the wild-type (WT) strain at transcript and protein levels. Bacteria lacking RsbX were taken up by phagocytic and nonphagocytic cells at a similar frequency to WT bacteria, both in stressed and nonstressed conditions. Finally, the absence of RsbX only marginally affected the ability of bacteria to infect chicken embryos. Our results suggest only a minor role of RsbX in controlling virulence factor expression and infectivity under these conditions.


Subject(s)
Listeria monocytogenes , Chick Embryo , Animals , Virulence , Bacterial Proteins/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Sigma Factor/genetics , Gene Expression Regulation, Bacterial
8.
J Bacteriol ; 204(1): e0048621, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34694900

ABSTRACT

The survival of microbial cells under changing environmental conditions requires an efficient reprogramming of transcription, often mediated by alternative sigma factors. The Gram-positive human pathogen Listeria monocytogenes senses and responds to environmental stress mainly through the alternative sigma factor σB (SigB), which controls expression of the general stress response regulon. SigB activation is achieved through a complex series of phosphorylation/dephosphorylation events culminating in the release of SigB from its anti-sigma factor RsbW. At the top of the signal transduction pathway lies a large multiprotein complex known as the stressosome that is believed to act as a sensory hub for stresses. Following signal detection, stressosome proteins become phosphorylated. Resetting of the stressosome is hypothesized to be exerted by a putative phosphatase, RsbX, which presumably removes phosphate groups from stressosome proteins poststress. We addressed the role of the RsbX protein in modulating the activity of the stressosome and consequently regulating SigB activity in L. monocytogenes. We show that RsbX is required to reduce SigB activation levels under nonstress conditions and that it is required for appropriate SigB-mediated stress adaptation. A strain lacking RsbX displayed impaired motility and biofilm formation and also an increased survival at low pH. Our results could suggest that absence of RsbX alters the multiprotein composition of the stressosome without dramatically affecting its phosphorylation status. Overall, the data show that RsbX plays a critical role in modulating the signal transduction pathway by blocking SigB activation under nonstressed conditions. IMPORTANCE Pathogenic bacteria need to sense and respond to stresses to survive harsh environments and also to turn off the response when no longer facing stress. Activity of the stress sigma factor SigB in the human pathogen Listeria monocytogenes is controlled by a hierarchic system having a large stress-sensing multiprotein complex known as the stressosome at the top. Following stress exposure, proteins in the stressosome become phosphorylated, leading to SigB activation. We have studied the role of a putative phosphatase, RsbX, which is hypothesized to dephosphorylate stressosome proteins. RsbX is critical not only to switch off the stress response poststress but also to keep the activity of SigB low at nonstressed conditions to prevent unnecessary gene expression and save energy.


Subject(s)
Gene Expression Regulation, Bacterial/physiology , Listeria monocytogenes/metabolism , Sigma Factor/metabolism , Stress, Physiological/physiology , Biofilms , Listeria monocytogenes/genetics , Mutation , Sigma Factor/genetics
9.
Mol Microbiol ; 113(3): 588-592, 2020 03.
Article in English | MEDLINE | ID: mdl-31971637

ABSTRACT

An outstanding question regards the ability of organisms to sense their environments and respond in a suitable way. Pathogenic bacteria in particular exploit host-temperature sensing as a cue for triggering virulence gene expression. This micro-review does not attempt to fully cover the field of bacterial thermosensors and in detail describe each identified case. Instead, the review focus on the time-period at the end of the 1990's and beginning of the 2000's when several key discoveries were made, identifying protein, DNA and RNA as potential thermosensors controlling gene expression in several different bacterial pathogens in general and on the prfA thermosensor of Listeria monocytogenes in particular.


Subject(s)
Bacteria/metabolism , Host Microbial Interactions/physiology , Thermoreceptors/physiology , Bacteria/pathogenicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics , Hot Temperature , Listeria monocytogenes/metabolism , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , RNA/genetics , RNA/metabolism , Thermoreceptors/metabolism , Thermosensing/genetics , Thermosensing/physiology , Trans-Activators/metabolism , Virulence/genetics , Virulence Factors/genetics
10.
J Bacteriol ; 202(11)2020 05 11.
Article in English | MEDLINE | ID: mdl-32179627

ABSTRACT

Listeria monocytogenes is a Gram-positive pathogen able to cause severe human infections. Its major virulence regulator is the transcriptional activator PrfA, a member of the Crp/Fnr family of transcriptional regulators. To establish a successful L. monocytogenes infection, the PrfA protein needs to be in an active conformation, either by binding the cognate inducer glutathione (GSH) or by possessing amino acid substitutions rendering the protein constitutively active (PrfA*). By a yet unknown mechanism, phosphotransferase system (PTS) sugars repress the activity of PrfA. We therefore took a transposon-based approach to identify the mechanism by which PTS sugars repress PrfA activity. For this, we screened a transposon mutant bank to identify clones able to grow in the presence of glucose-6-phosphate as the sole carbon source. Surprisingly, most of the isolated transposon mutants also carried amino acid substitutions in PrfA. In transposon-free strains, the PrfA amino acid substitution mutants displayed growth, virulence factor expression, infectivity, and DNA binding, agreeing with previously identified PrfA* mutants. Hence, the initial growth phenotype observed in the isolated clone was due to the amino acid substitution in PrfA and unrelated to the loci inactivated by the transposon mutant. Finally, we provide structural evidence for the existence of an intermediately activated PrfA state, which gives new insights into PrfA protein activation.IMPORTANCE The Gram-positive bacterium Listeria monocytogenes is a human pathogen affecting mainly the elderly, immunocompromised people, and pregnant women. It can lead to meningoencephalitis, septicemia, and abortion. The major virulence regulator in L. monocytogenes is the PrfA protein, a transcriptional activator. Using a growth-based selection strategy, we identified mutations in the PrfA protein leading to constitutively active virulence factor expression. We provide structural evidence for the existence of an intermediately activated PrfA state, which gives new insights into PrfA protein activation.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/growth & development , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Peptide Termination Factors/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Humans , Mutation , Peptide Termination Factors/genetics , Virulence
11.
J Bacteriol ; 202(9)2020 04 09.
Article in English | MEDLINE | ID: mdl-32094160

ABSTRACT

In Listeria monocytogenes, the full details of how stress signals are integrated into the σB regulatory pathway are not yet available. To help shed light on this question, we investigated a collection of transposon mutants that were predicted to have compromised activity of the alternative sigma factor B (σB). These mutants were tested for acid tolerance, a trait that is known to be under σB regulation, and they were found to display increased acid sensitivity, similar to a mutant lacking σB (ΔsigB). The transposon insertions were confirmed by whole-genome sequencing, but in each case, the strains were also found to carry a frameshift mutation in the sigB operon. The changes were predicted to result in premature stop codons, with negative consequences for σB activation, independently of the transposon location. Reduced σB activation in these mutants was confirmed. Growth measurements under conditions similar to those used during the construction of the transposon library revealed that the frameshifted sigB operon alleles conferred a growth advantage at higher temperatures, during late exponential phase. Mixed-culture experiments at 42°C demonstrated that the loss of σB activity allowed mutants to take over a population of parental bacteria. Together, our results suggest that mutations affecting σB activity can arise during laboratory culture because of the growth advantage conferred by these mutations under mild stress conditions. The data highlight the significant cost of stress protection in this foodborne pathogen and emphasize the need for whole-genome sequence analysis of newly constructed strains to confirm the expected genotype.IMPORTANCE In the present study, we investigated a collection of Listeria monocytogenes strains that all carried sigB operon mutations. The mutants all had reduced σB activity and were found to have a growth advantage under conditions of mild heat stress (42°C). In mixed cultures, these mutants outcompeted the wild type when mild heat stress was present but not at an optimal growth temperature. An analysis of 22,340 published L. monocytogenes genome sequences found a high rate of premature stop codons present in genes positively regulating σB activity. Together, these findings suggest that the occurrence of mutations that attenuate σB activity can be favored under conditions of mild stress, probably highlighting the burden on cellular resources that stems from deploying the general stress response.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/physiology , Sigma Factor/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Kinetics , Listeria monocytogenes/chemistry , Listeria monocytogenes/genetics , Listeria monocytogenes/growth & development , Mutation , Operon , Sigma Factor/genetics , Stress, Physiological
12.
Microbiology (Reading) ; 165(8): 819-833, 2019 08.
Article in English | MEDLINE | ID: mdl-31107205

ABSTRACT

In contrast to obligate intracellular pathogens that can remain in relatively stable host-associated environments, the soil-living bacterial pathogen Listeria monocytogenes has to sense and respond to physical and chemical cues in a variety of quite different niches. In particular, the bacterium has to survive the dramatic transition from its saprophytic existence to life within the host where nutritional stress, increased temperature, acidity, osmotic stress and the host defences present a new and challenging landscape. This review focuses on the σB and PrfA regulatory systems used by L. monocytogenes to sense the changing environment and implement survival mechanisms that help to overcome the disparate conditions within the host, but also to switch from a harmless saprophyte to an impressively effective pathogen.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/pathogenicity , Peptide Termination Factors/metabolism , Sigma Factor/metabolism , Virulence Factors/genetics , Virulence/genetics , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Light Signal Transduction/genetics , Listeria monocytogenes/genetics , Listeriosis , Peptide Termination Factors/genetics , Sigma Factor/genetics , Virulence Factors/metabolism
13.
RNA Biol ; 16(10): 1424-1437, 2019 10.
Article in English | MEDLINE | ID: mdl-31242083

ABSTRACT

The facultative intracellular pathogen Listeria monocytogenes can persist and grow in a diverse range of environmental conditions, both outside and within its mammalian host. The alternative sigma factor Sigma B (σB) plays an important role in this adaptability and is critical for the transition into the host. While some of the functions of the σB regulon in facilitating this transition are understood the role of σB-dependent small regulatory RNAs (sRNAs) remain poorly characterized. In this study, we focused on elucidating the function of Rli47, a σB-dependent sRNA that is highly induced in the intestine and in macrophages. Using a combination of in silico and in vivo approaches, a binding interaction was predicted with the Shine-Dalgarno region of the ilvA mRNA, which encodes threonine deaminase, an enzyme required for branched-chain amino acid biosynthesis. Both ilvA transcript levels and threonine deaminase activity were increased in a deletion mutant lacking the rli47 gene. The Δrli47 mutant displayed a shorter growth lag in isoleucine-depleted growth media relative to the wild-type, and a similar phenotype was also observed in a mutant lacking σB. The impact of the Δrli47 on the global transcription profile of the cell was investigated using RNA-seq, and a significant role for Rli47 in modulating amino acid metabolism was uncovered. Taken together, the data point to a model where Rli47 is responsible for specifically repressing isoleucine biosynthesis as a way to restrict growth under harsh conditions, potentially contributing to the survival of L. monocytogenes in niches both outside and within the mammalian host.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Isoleucine/biosynthesis , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , RNA, Small Untranslated/genetics , Sigma Factor/metabolism , Amino Acids/metabolism , Bacterial Proteins/metabolism , Biological Transport , Gene Expression Profiling , Mutation , Nucleic Acid Conformation , Protein Binding , RNA, Messenger/genetics , RNA, Small Untranslated/chemistry , Transcription, Genetic
14.
BMC Infect Dis ; 19(1): 164, 2019 Feb 14.
Article in English | MEDLINE | ID: mdl-30764767

ABSTRACT

BACKGROUND: Herpes simplex virus type 1 (HSV1), establishes life-long latency and can cause symptoms during both first-time infection and later reactivation. The aim of the present study was to describe a protocol to generate a reliable and discriminative avidity index (AI) for anti-HSV1 IgG content in human sera. METHODS: Human serum from two distinct cohorts; one a biobank collection (Betula) (n = 28), and one from a clinical diagnostics laboratory at Northern Sweden University Hospital (NUS) (n = 18), were assessed for presence of IgG antibodies against HSV1 by a commercially available ELISA-kit. Addition of urea at the incubation step reduces effective binding, and the ratio between urea treated sample and non-treated sample was used to express an avidity index (AI) for individual samples. RESULTS: AI score ranged between 43.2 and 73.4% among anti-HSV1 positive biobank sera. Clinical samples ranged between 36.3 and 74.9%. Reproducibility expressed as an intraclass correlation coefficient (ICC) was estimated at 0.948 (95% CI: 0.900-0.979) and 0.989 (95% CI 0.969-0.996) in the biobank and clinical samples, respectively. CONCLUSION: The method allows for AI scoring of anti-HSV1 IgG from individual human sera with a single measurement. The least significant change between two measurements at the p < 0.05 level was estimated at 5.4 and 3.2 points, respectively, for the two assessed cohorts.


Subject(s)
Antibodies, Viral/analysis , Antibody Affinity/drug effects , Herpesvirus 1, Human/immunology , Immunoglobulin G/analysis , Serologic Tests/methods , Urea/pharmacology , Adult , Aged , Aged, 80 and over , Antibodies, Viral/blood , Antibodies, Viral/drug effects , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Immunoglobulin G/metabolism , Immunoglobulin M/blood , Indicator Dilution Techniques , Male , Middle Aged , Reagent Kits, Diagnostic , Reproducibility of Results
15.
Nucleic Acids Res ; 45(4): 1914-1924, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28062853

ABSTRACT

The emerging development of antibiotic resistant bacteria calls for novel types of antibacterial agents. In this work we examined the putative antibacterial effect of purine analogs in Listeria monocytogenes. We show that, among several tested purine analogs, only 6-N-hydroxylaminopurine (6-N-HAP) reduces the viability of the Gram-positive pathogen Listeria monocytogenes. As in Bacillus subtilis, 6-N-HAP terminates expression at guanine riboswitches in L. monocytogenes hence preventing expression of their downstream genes. However, we show that the bacteriocidal effect of the compound was unlinked to the terminated expression at the guanine riboswitches. When further examining the antimicrobial effect, we observed that 6-N-HAP acts as a potent mutagen in L. monocytogenes, by increasing the mutation rate and inducing the SOS-response. Also, addition of 6-N-HAP decreased virulence gene expression by reducing both the levels and activity of the virulence regulator PrfA.


Subject(s)
Adenine/analogs & derivatives , Anti-Bacterial Agents/pharmacology , Listeria monocytogenes/drug effects , Listeria monocytogenes/physiology , Virulence/drug effects , Adenine/pharmacology , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial/drug effects , Listeria monocytogenes/pathogenicity , Microbial Viability/drug effects , Microbial Viability/genetics , Mutagens/pharmacology , Mutation , Mutation Rate , Riboswitch/drug effects , Rifamycins/pharmacology , SOS Response, Genetics/drug effects , SOS Response, Genetics/genetics , Virulence/genetics
16.
Proc Natl Acad Sci U S A ; 113(51): 14733-14738, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27930316

ABSTRACT

Infection by the human bacterial pathogen Listeria monocytogenes is mainly controlled by the positive regulatory factor A (PrfA), a member of the Crp/Fnr family of transcriptional activators. Published data suggest that PrfA requires the binding of a cofactor for full activity, and it was recently proposed that glutathione (GSH) could fulfill this function. Here we report the crystal structures of PrfA in complex with GSH and in complex with GSH and its cognate DNA, the hly operator PrfA box motif. These structures reveal the structural basis for a GSH-mediated allosteric mode of activation of PrfA in the cytosol of the host cell. The crystal structure of PrfAWT in complex only with DNA confirms that PrfAWT can adopt a DNA binding-compatible structure without binding the GSH activator molecule. By binding to PrfA in the cytosol of the host cell, GSH induces the correct fold of the HTH motifs, thus priming the PrfA protein for DNA interaction.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/metabolism , Peptide Termination Factors/metabolism , Amino Acid Motifs , Crystallography, X-Ray , DNA, Bacterial/chemistry , Gene Expression Regulation, Bacterial , Glutathione/chemistry , Glycine/chemistry , Protein Binding , Protein Multimerization , Trans-Activators/metabolism , Transcription Factors/metabolism , Virulence
17.
Vet Res ; 49(1): 13, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29409521

ABSTRACT

Most human listeriosis outbreaks are caused by Listeria monocytogenes evolutionary lineage I strains which possess four exotoxins: a phosphatidylinositol-specific phospholipase C (PlcA), a broad-range phospholipase C (PlcB), listeriolysin O (LLO) and listeriolysin S (LLS). The simultaneous contribution of these molecules to virulence has never been explored. Here, the importance of these four exotoxins of an epidemic lineage I L. monocytogenes strain (F2365) in virulence was assessed in chicken embryos infected in the allantoic cavity. We show that LLS does not play a role in virulence while LLO is required to infect and kill chicken embryos both in wild type transcriptional regulator of virulence PrfA (PrfAWT) and constitutively active PrfA (PrfA*) backgrounds. We demonstrate that PlcA, a toxin previously considered as a minor virulence factor, played a major role in virulence in a PrfA* background. Interestingly, GFP transcriptional fusions show that the plcA promoter is less active than the hly promoter in vitro, explaining why the contribution of PlcA to virulence could be observed more importantly in a PrfA* background. Together, our results suggest that PlcA might play a more important role in the infectious lifecycle of L. monocytogenes than previously thought, explaining why all the strains of L. monocytogenes have conserved an intact copy of plcA in their genomes.


Subject(s)
Bacterial Proteins/genetics , Bacterial Toxins/pharmacology , Exotoxins/pharmacology , Heat-Shock Proteins/pharmacology , Hemolysin Proteins/pharmacology , Listeria monocytogenes/pathogenicity , Phospholipases/toxicity , Virulence Factors/pharmacology , Animals , Bacterial Proteins/metabolism , Chick Embryo , Listeria monocytogenes/enzymology , Virulence , Virulence Factors/genetics
18.
Genes Dev ; 24(13): 1345-50, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20595230

ABSTRACT

In Escherichia coli, the major nucleoid protein H-NS limits transcription by acting as a repressor or transcriptional silencer, presumably by its ability to close the looped chromosome domains in the nucleoid through DNA-protein-DNA bridging. Here, we demonstrate the direct involvement of H-NS as a positive factor stimulating translation of the malT mRNA. In vitro studies showed that H-NS facilitates a repositioning of the 30S preinitiation complex on the malT mRNA. H-NS stimulation of translation depended on the AU-rich -35 to -40 region of the mRNA. Several additional examples were found demonstrating a novel function for H-NS in translation of genes with suboptimal ribosome-binding sequences.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Fimbriae Proteins/metabolism , Protein Biosynthesis , Ribosomes/metabolism , Transcriptional Activation , Binding Sites , Protein Binding
19.
J Bacteriol ; 198(23): 3233-3243, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27672192

ABSTRACT

The riboflavin analogs roseoflavin (RoF) and 8-demethyl-8-aminoriboflavin (AF) are produced by the bacteria Streptomyces davawensis and Streptomyces cinnabarinus Riboflavin analogs have the potential to be used as broad-spectrum antibiotics, and we therefore studied the metabolism of riboflavin (vitamin B2), RoF, and AF in the human pathogen Listeria monocytogenes, a bacterium which is a riboflavin auxotroph. We show that the L. monocytogenes protein Lmo1945 is responsible for the uptake of riboflavin, RoF, and AF. Following import, these flavins are phosphorylated/adenylylated by the bifunctional flavokinase/flavin adenine dinucleotide (FAD) synthetase Lmo1329 and adenylylated by the unique FAD synthetase Lmo0728, the first monofunctional FAD synthetase to be described in bacteria. Lmo1329 generates the cofactors flavin mononucleotide (FMN) and FAD, whereas Lmo0728 produces FAD only. The combined activities of Lmo1329 and Lmo0728 are responsible for the intracellular formation of the toxic cofactor analogs roseoflavin mononucleotide (RoFMN), roseoflavin adenine dinucleotide (RoFAD), 8-demethyl-8-aminoriboflavin mononucleotide (AFMN), and 8-demethyl-8-aminoriboflavin adenine dinucleotide (AFAD). In vivo reporter gene assays and in vitro transcription/translation experiments show that the L. monocytogenes FMN riboswitch Rli96, which controls expression of the riboflavin transport gene lmo1945, is negatively affected by riboflavin/FMN and RoF/RoFMN but not by AF/AFMN. Treatment of L. monocytogenes with RoF or AF leads to drastically reduced FMN/FAD levels. We suggest that the reduced flavin cofactor levels in combination with concomitant synthesis of inactive cofactor analogs (RoFMN, RoFAD, AFMN, and AFAD) explain why RoF and AF contribute to antibiotic activity in L. monocytogenes IMPORTANCE: The riboflavin analogs roseoflavin (RoF) and 8-demethyl-8-aminoriboflavin (AF) are small molecules which are produced by Streptomyces davawensis and Streptomyces cinnabarinus RoF and AF were reported to have antibacterial activity, and we studied how these compounds are metabolized by the human bacterial pathogen Listeria monocytogenes We found that the L. monocytogenes protein Lmo1945 mediates uptake of AF and RoF and that the combined activities of the enzymes Lmo1329 and Lmo0728 are responsible for the conversion of AF and RoF to toxic cofactor analogs. Comparative studies with RoF and AF (a weaker antibiotic) suggest that the reduction in FMN/FAD levels and the formation of inactive FMN/FAD analogs explain to a large extent the antibiotic activity of AF and RoF.


Subject(s)
Anti-Bacterial Agents/metabolism , Listeria monocytogenes/metabolism , Riboflavin/analogs & derivatives , Autotrophic Processes , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Listeria monocytogenes/genetics , Listeria monocytogenes/growth & development , Riboflavin/metabolism
20.
Infect Immun ; 84(1): 67-76, 2016 01.
Article in English | MEDLINE | ID: mdl-26483402

ABSTRACT

RNA helicases have been shown to be important for the function of RNA molecules at several levels, although their putative involvement in microbial pathogenesis has remained elusive. We have previously shown that Listeria monocytogenes DExD-box RNA helicases are important for bacterial growth, motility, ribosomal maturation, and rRNA processing. We assessed the importance of the RNA helicase Lmo0866 (here named CshA) for expression of virulence traits. We observed a reduction in hemolytic activity in a strain lacking CshA compared to the wild type. This phenomenon was less evident in strains lacking other RNA helicases. The reduced hemolysis was accompanied by lower expression of major listerial virulence factors in the ΔcshA strain, mainly listeriolysin O, but also to some degree the actin polymerizing factor ActA. Reduced expression of these virulence factors in the strain lacking CshA did not, however, correlate with a decreased level of the virulence regulator PrfA. When combining the ΔcshA knockout with a mutation creating a constitutively active PrfA protein (PrfA*), the effect of the ΔcshA knockout on LLO expression was negated. These data suggest a role for the RNA helicase CshA in posttranslational activation of PrfA. Surprisingly, although the expression of several virulence factors was reduced, the ΔcshA strain did not demonstrate any reduced ability to infect nonphagocytic cells compared to the wild-type strain.


Subject(s)
Gene Expression Regulation, Bacterial/genetics , Hemolysis/genetics , Listeria monocytogenes/pathogenicity , RNA Helicases/genetics , Virulence Factors/genetics , Animals , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/biosynthesis , Bacterial Toxins/genetics , Caco-2 Cells , Cell Line , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Hemolysin Proteins/biosynthesis , Hemolysin Proteins/genetics , Humans , Listeria monocytogenes/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Protein Processing, Post-Translational/genetics , Virulence Factors/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL