Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2427: 3-10, 2022.
Article in English | MEDLINE | ID: mdl-35619020

ABSTRACT

Gene alteration/deletion by allelic exchange is the preferred strategy for gene manipulation in bacteria. Here we present the fundamentals for an efficient allelic exchange gene deletion method in the bacterial pathogen Listeria monocytogenes. Combining vector generation by Gibson assembly with a counterselection system based on the mutated phenylalanine synthetase (pheS*) makes the generation of gene deletion mutants straightforward and time efficient.


Subject(s)
Listeria monocytogenes , Alleles , Gene Deletion , Listeria monocytogenes/genetics , Mutation , Phenylalanine/genetics
2.
Methods Mol Biol ; 2427: 83-93, 2022.
Article in English | MEDLINE | ID: mdl-35619027

ABSTRACT

Listeria monocytogenes is a gram-positive bacterium adapted to life as both an environmental saprophyte and a pathogenic parasite of mammalian hosts, with a transcriptomic program tailored for each niche. Study of the L. monocytogenes pathogenic lifestyle requires conditions that mimic the mammalian niche. Of the myriad experimental models used to achieve such conditions, the bone marrow-derived macrophage (BMDM) is a relatively simple and reliable primary immune cell model for L. monocytogenes infections. Here we describe the extraction, preparation, and storage of BMDMs and their use in L. monocytogenes infection experiments.


Subject(s)
Listeria monocytogenes , Listeriosis , Animals , Listeria monocytogenes/genetics , Listeriosis/microbiology , Macrophages , Mammals
3.
mBio ; 13(3): e0044822, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35435705

ABSTRACT

Listeria monocytogenes is a saprophyte and a human intracellular pathogen. Upon invasion into mammalian cells, it senses multiple metabolic and environmental signals that collectively trigger its transition to the pathogenic state. One of these signals is the tripeptide glutathione, which acts as an allosteric activator of L. monocytogenes's master virulence regulator, PrfA. While glutathione synthesis by L. monocytogenes was shown to be critical for PrfA activation and virulence gene expression, it remains unclear how this tripeptide is synthesized in changing environments, especially in light of the observation that L. monocytogenes is auxotrophic to one of its precursors, cysteine. Here, we show that the ABC transporter TcyKLMN is a cystine/cysteine importer that supplies cysteine for glutathione synthesis, hence mediating the induction of the virulence genes. Further, we demonstrate that this transporter is negatively regulated by three metabolic regulators, CodY, CymR, and CysK, which sense and respond to changing concentrations of branched-chain amino acids (BCAA) and cysteine. The data indicate that under low concentrations of BCAA, TcyKLMN is upregulated, driving the production of glutathione by supplying cysteine, thereby facilitating PrfA activation. These findings provide molecular insight into the coupling of L. monocytogenes metabolism and virulence, connecting BCAA sensing to cysteine uptake and glutathione biosynthesis as a mechanism that controls virulence gene expression. This study exemplifies how bacterial pathogens sense their intracellular environment and exploit essential metabolites as effectors of virulence. IMPORTANCE Bacterial pathogens sense the repertoire of metabolites in the mammalian niche and use this information to shift into the pathogenic state to accomplish a successful infection. Glutathione is a virulence-activating signal that is synthesized by L. monocytogenes during infection of mammalian cells. In this study, we show that cysteine uptake via TcyKLMN drives glutathione synthesis and virulence gene expression. The data emphasize the intimate cross-regulation between metabolism and virulence in bacterial pathogens.


Subject(s)
Listeria monocytogenes , Amino Acids, Branched-Chain/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cysteine/metabolism , Cystine/genetics , Cystine/metabolism , Gene Expression , Gene Expression Regulation, Bacterial , Glutathione/metabolism , Humans , Mammals/genetics , Membrane Transport Proteins/metabolism , Peptide Termination Factors/metabolism , Virulence/genetics
4.
Cell Rep ; 39(3): 110723, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35443160

ABSTRACT

Listeria monocytogenes strain 10403S harbors two phage elements in its chromosome; one produces infective virions and the other tailocins. It was previously demonstrated that induction of the two elements is coordinated, as they are regulated by the same anti-repressor. In this study, we identified AriS as another phage regulator that controls the two elements, bearing the capacity to inhibit their lytic induction under SOS conditions. AriS is a two-domain protein that possesses two distinct activities, one regulating the genes of its encoding phage and the other downregulating the bacterial SOS response. While the first activity associates with the AriS N-terminal AntA/AntB domain, the second associates with its C-terminal ANT/KilAC domain. The ANT/KilAC domain is conserved in many AriS-like proteins of listerial and non-listerial prophages, suggesting that temperate phages acquired such dual-function regulators to align their response with the other phage elements that cohabit the genome.


Subject(s)
Bacteriophages , Listeria monocytogenes , Bacteriophages/genetics , Listeria monocytogenes/genetics , Lysogeny , Prophages/genetics , SOS Response, Genetics
5.
Microorganisms ; 9(6)2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34207021

ABSTRACT

Infection of mammalian cells by Listeria monocytogenes (Lm) was shown to be facilitated by its phage elements. In a search for additional phage remnants that play a role in Lm's lifecycle, we identified a conserved locus containing two XRE regulators and a pair of genes encoding a secreted metzincin protease and a lipoprotein structurally similar to a TIMP-family metzincin inhibitor. We found that the XRE regulators act as a classic CI/Cro regulatory switch that regulates the expression of the metzincin and TIMP-like genes under intracellular growth conditions. We established that when these genes are expressed, their products alter Lm morphology and increase its sensitivity to phage mediated lysis, thereby enhancing virion release. Expression of these proteins also sensitized the bacteria to cell wall targeting compounds, implying that they modulate the cell wall structure. Our data indicate that these effects are mediated by the cleavage of the TIMP-like protein by the metzincin, and its subsequent release to the extracellular milieu. While the importance of this locus to Lm pathogenicity remains unclear, the observation that this phage-associated protein pair act upon the bacterial cell wall may hold promise in the field of antibiotic potentiation to combat antibiotic resistant bacterial pathogens.

6.
Cell Rep ; 32(4): 107956, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32726621

ABSTRACT

Some Listeria monocytogenes (Lm) strains harbor a prophage within the comK gene, which renders it inactive. During Lm infection of macrophage cells, the prophage turns into a molecular switch, promoting comK gene expression and therefore Lm intracellular growth. During this process, the prophage does not produce infective phages or cause bacterial lysis, suggesting it has acquired an adaptive behavior suited to the pathogenic lifestyle of its host. In this study, we demonstrate that this non-classical phage behavior, named active lysogeny, relies on a transcriptional response that is specific to the intracellular niche. While the prophage undergoes lytic induction, the process is arrested midway, preventing the transcription of the late genes. Further, we demonstrate key phage factors, such as LlgA transcription regulator and a DNA replicase, that support the phage adaptive behavior. This study provides molecular insights into the adaptation of phages to their pathogenic hosts, uncovering unusual cooperative interactions.


Subject(s)
Bacterial Proteins/genetics , Listeria monocytogenes/metabolism , Lysogeny/physiology , Transcription Factors/genetics , Animals , Bacterial Proteins/metabolism , Bacteriophages/genetics , Female , Listeriosis/metabolism , Mice , Mice, Inbred C57BL , Prophages/genetics , Transcription Factors/metabolism , Virus Activation/physiology
7.
Nat Commun ; 10(1): 5288, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31754112

ABSTRACT

Bacterial pathogens often carry multiple prophages and other phage-derived elements within their genome, some of which can produce viral particles in response to stress. Listeria monocytogenes 10403S harbors two phage elements in its chromosome, both of which can trigger bacterial lysis under stress: an active prophage (ϕ10403S) that promotes the virulence of its host and can produce infective virions, and a locus encoding phage tail-like bacteriocins. Here, we show that the two phage elements are co-regulated, with the bacteriocin locus controlling the induction of the prophage and thus its activity as a virulence-associated molecular switch. More specifically, a metalloprotease encoded in the bacteriocin locus is upregulated in response to stress and acts as an anti-repressor for CI-like repressors encoded in each phage element. Our results provide molecular insight into the phenomenon of polylysogeny and its intricate adaptation to complex environments.


Subject(s)
Bacteriophages/immunology , Chromosomes, Bacterial/immunology , Listeria monocytogenes/immunology , Prophages/immunology , Amino Acid Sequence , Bacteriocins/genetics , Bacteriocins/immunology , Bacteriolysis/immunology , Bacteriophages/genetics , Bacteriophages/physiology , Chromosomes, Bacterial/genetics , Chromosomes, Bacterial/virology , Genome, Bacterial/genetics , Genome, Bacterial/immunology , Genome, Viral/genetics , Genome, Viral/immunology , Host-Pathogen Interactions/immunology , Listeria monocytogenes/genetics , Listeria monocytogenes/virology , Lysogeny/genetics , Lysogeny/immunology , Metalloproteases/genetics , Metalloproteases/immunology , Prophages/genetics , Prophages/physiology , Sequence Homology, Amino Acid , Virus Activation/genetics , Virus Activation/immunology
8.
Microbiol Spectr ; 7(4)2019 07.
Article in English | MEDLINE | ID: mdl-31418407

ABSTRACT

Bacterial metabolism represents the biochemical space that bacteria can manipulate to produce energy, reducing equivalents and building blocks for replication. Gram-positive pathogens, such as Listeria monocytogenes, show remarkable flexibility, which allows for exploitation of diverse biological niches from the soil to the intracytosolic space. Although the human host represents a potentially rich source for nutrient acquisition, competition for nutrients with the host and hostile host defenses can constrain bacterial metabolism by various mechanisms, including nutrient sequestration. Here, we review metabolism in the model Gram-positive bacterium, L. monocytogenes, and highlight pathways that enable the replication, survival, and virulence of this bacterial pathogen.


Subject(s)
Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Humans , Listeria monocytogenes/genetics , Virulence
9.
PLoS Genet ; 14(3): e1007283, 2018 03.
Article in English | MEDLINE | ID: mdl-29529043

ABSTRACT

Listeria monocytogenes (Lm) is a saprophyte and intracellular pathogen. Transition to the pathogenic state relies on sensing of host-derived metabolites, yet it remains unclear how these are recognized and how they mediate virulence gene regulation. We previously found that low availability of isoleucine signals Lm to activate the virulent state. This response is dependent on CodY, a global regulator and isoleucine sensor. Isoleucine-bound CodY represses metabolic pathways including branched-chain amino acids (BCAA) biosynthesis, however under BCAA depletion, as occurs during infection, BCAA biosynthesis is upregulated and isoleucine-unbound CodY activates virulence genes. While isoleucine was revealed as an important input signal, it was not identified how internal levels are controlled during infection. Here we show that Lm regulates BCAA biosynthesis via CodY and via a riboregulator located upstream to the BCAA biosynthesis genes, named Rli60. rli60 is transcribed when BCAA levels drop, forming a ribosome-mediated attenuator that cis-regulates the downstream genes according to BCAA supply. Notably, we found that Rli60 restricts BCAA production, essentially starving Lm, a mechanism that is directly linked to virulence, as it controls the internal isoleucine pool and thereby CodY activity. This controlled BCAA auxotrophy likely evolved to enable isoleucine to serve as a host signal and virulence effector.


Subject(s)
Amino Acids, Branched-Chain/biosynthesis , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Amino Acids, Branched-Chain/genetics , Genes, Bacterial , Isoleucine/biosynthesis , Isoleucine/genetics , Listeria monocytogenes/genetics , Transcription, Genetic , Virulence
10.
Curr Opin Microbiol ; 38: 81-87, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28544996

ABSTRACT

Bacteriophages are ubiquitous and affect most facets of life, from evolution of bacteria, through ecology and global biochemical cycling to human health. The interactions between phages and bacteria often lead to biological novelty and an important milestone in this process is the ability of phages to regulate their host's behavior. In this review article, we will focus on newly reported cases that demonstrate how temperate phages regulate bacterial gene expression and behavior in a variety of bacterial species, pathogenic and environmental. This regulation is mediated by diverse mechanisms such as transcription factors, sRNAs, DNA rearrangements, and even controlled bacterial lysis. The outcome is mutualistic relationships that enable adaptively enhanced communal phage-host fitness under specific conditions.


Subject(s)
Bacteria/genetics , Bacteria/virology , Gene Expression Regulation, Bacterial , Host-Parasite Interactions , Lysogeny , Prophages/genetics
11.
Infect Immun ; 85(6)2017 06.
Article in English | MEDLINE | ID: mdl-28396325

ABSTRACT

Listeria monocytogenes is an environmental saprophyte and intracellular bacterial pathogen. Upon invading mammalian cells, the bacterium senses abrupt changes in its metabolic environment, which are rapidly transduced to regulation of virulence gene expression. To explore the relationship between L. monocytogenes metabolism and virulence, we monitored virulence gene expression dynamics across a library of genetic mutants grown under two metabolic conditions known to activate the virulent state: charcoal-treated rich medium containing glucose-1-phosphate and minimal defined medium containing limiting concentrations of branched-chain amino acids (BCAAs). We identified over 100 distinct mutants that exhibit aberrant virulence gene expression profiles, the majority of which mapped to nonessential metabolic genes. Mutants displayed enhanced, decreased, and early and late virulence gene expression profiles, as well as persistent levels, demonstrating a high plasticity in virulence gene regulation. Among the mutants, one was noteworthy for its particularly low virulence gene expression level and mapped to an X-prolyl aminopeptidase (PepP). We show that this peptidase plays a role in posttranslational activation of the major virulence regulator, PrfA. Specifically, PepP mediates recruitment of PrfA to the cytoplasmic membrane, a step identified as critical for PrfA protein activation. This study establishes a novel step in the complex mechanism of PrfA activation and further highlights the cross regulation of metabolism and virulence.


Subject(s)
Aminopeptidases/metabolism , Bacterial Proteins/genetics , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Macrophages/microbiology , Peptide Termination Factors/genetics , Virulence Factors/genetics , Animals , Female , Gene Expression Regulation, Bacterial , Glucosephosphates/metabolism , Listeria monocytogenes/metabolism , Listeriosis/microbiology , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mutation , RNA, Bacterial/genetics , Virulence/genetics
12.
PLoS Pathog ; 13(1): e1006161, 2017 01.
Article in English | MEDLINE | ID: mdl-28114430

ABSTRACT

The high environmental adaptability of bacteria is contingent upon their ability to sense changes in their surroundings. Bacterial pathogen entry into host poses an abrupt and dramatic environmental change, during which successful pathogens gauge multiple parameters that signal host localization. The facultative human pathogen Listeria monocytogenes flourishes in soil, water and food, and in ~50 different animals, and serves as a model for intracellular infection. L. monocytogenes identifies host entry by sensing both physical (e.g., temperature) and chemical (e.g., metabolite concentrations) factors. We report here that L-glutamine, an abundant nitrogen source in host serum and cells, serves as an environmental indicator and inducer of virulence gene expression. In contrast, ammonia, which is the most abundant nitrogen source in soil and water, fully supports growth, but fails to activate virulence gene transcription. We demonstrate that induction of virulence genes only occurs when the Listerial intracellular concentration of L-glutamine crosses a certain threshold, acting as an on/off switch: off when L-glutamine concentrations are below the threshold, and fully on when the threshold is crossed. To turn on the switch, L-glutamine must be present, and the L-glutamine high affinity ABC transporter, GlnPQ, must be active. Inactivation of GlnPQ led to complete arrest of L-glutamine uptake, reduced type I interferon response in infected macrophages, dramatic reduction in expression of virulence genes, and attenuated virulence in a mouse infection model. These results may explain observations made with other pathogens correlating nitrogen metabolism and virulence, and suggest that gauging of L-glutamine as a means of ascertaining host localization may be a general mechanism.


Subject(s)
Gene Expression Regulation, Bacterial/physiology , Glutamine/metabolism , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Virulence/physiology , Animals , Blotting, Western , Humans , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , Polymerase Chain Reaction
13.
Appl Environ Microbiol ; 83(6)2017 03 15.
Article in English | MEDLINE | ID: mdl-28039138

ABSTRACT

Construction of Listeria monocytogenes mutants by allelic exchange has been laborious and time-consuming due to lack of proficient selection markers for the final recombination event, that is, a marker conveying substance sensitivity to the bacteria bearing it, enabling the exclusion of merodiploids and selection for plasmid loss. In order to address this issue, we engineered a counterselection marker based on a mutated phenylalanyl-tRNA synthetase gene (pheS*). This mutation renders the phenylalanine-binding site of the enzyme more promiscuous and allows the binding of the toxic p-chloro-phenylalanine analog (p-Cl-phe) as a substrate. When pheS* is introduced into L. monocytogenes and highly expressed under control of a constitutively active promoter, the bacteria become sensitive to p-Cl-phe supplemented in the medium. This enabled us to utilize pheS* as a negative selection marker and generate a novel, efficient suicide vector for allelic exchange in L. monocytogenes We used this vector to investigate the monocin genomic region in L. monocytogenes strain 10403S by constructing deletion mutants of the region. We have found this region to be active and to cause bacterial lysis upon mitomycin C treatment. The future applications of such an effective counterselection system, which does not require any background genomic alterations, are vast, as it can be modularly used in various selection systems (e.g., genetic screens). We expect this counterselection marker to be a valuable genetic tool in research on L. monocytogenesIMPORTANCEL. monocytogenes is an opportunistic intracellular pathogen and a widely studied model organism. An efficient counterselection marker is a long-standing need in Listeria research for improving the ability to design and perform various genetic manipulations and screening systems for different purposes. We report the construction and utilization of an efficient suicide vector for allelic exchange which can be conjugated, leaves no marker in the bacterial chromosome, and does not require the use of sometimes leaky inducible promoters. This highly efficient genome editing tool for L. monocytogenes will allow for rapid sequential mutagenesis, introduction of point mutations, and design of screening systems. We anticipate that it will be extensively used by the research community and yield novel insights into the diverse fields studied using this model organism.


Subject(s)
Bacteriocins/genetics , Listeria monocytogenes/genetics , Mitomycin/pharmacology , Phenylalanine-tRNA Ligase/genetics , Phenylalanine/analogs & derivatives , Binding Sites/genetics , Binding Sites/physiology , Genetic Markers/genetics , Listeria monocytogenes/growth & development , Phenylalanine/metabolism , Promoter Regions, Genetic/genetics , Selection, Genetic/genetics , Sequence Deletion/genetics
14.
J Vis Exp ; (112)2016 06 04.
Article in English | MEDLINE | ID: mdl-27341521

ABSTRACT

Analysis of the transcriptome of bacterial pathogens during mammalian infection is a valuable tool for studying genes and factors that mediate infection. However, isolating bacterial RNA from infected cells or tissues is a challenging task, since mammalian RNA mostly dominates the lysates of infected cells. Here we describe an optimized method for RNA isolation of Listeria monocytogenes bacteria growing within bone marrow derived macrophage cells. Upon infection, cells are mildly lysed and rapidly filtered to discard most of the host proteins and RNA, while retaining intact bacteria. Next, bacterial RNA is isolated using hot phenol-SDS extraction followed by DNase treatment. The extracted RNA is suitable for gene transcription analysis by multiple techniques. This method is successfully employed in our studies of Listeria monocytogenes gene regulation during infection of macrophage cells (1-4). The protocol can be easily modified to study other bacterial pathogens and cell types.


Subject(s)
Macrophages , Animals , Gene Expression Regulation , Listeria monocytogenes , RNA, Bacterial
15.
PLoS Genet ; 12(2): e1005870, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26895237

ABSTRACT

Bacteria sense and respond to many environmental cues, rewiring their regulatory network to facilitate adaptation to new conditions/niches. Global transcription factors that co-regulate multiple pathways simultaneously are essential to this regulatory rewiring. CodY is one such global regulator, controlling expression of both metabolic and virulence genes in Gram-positive bacteria. Branch chained amino acids (BCAAs) serve as a ligand for CodY and modulate its activity. Classically, CodY was considered to function primarily as a repressor under rich growth conditions. However, our previous studies of the bacterial pathogen Listeria monocytogenes revealed that CodY is active also when the bacteria are starved for BCAAs. Under these conditions, CodY loses the ability to repress genes (e.g., metabolic genes) and functions as a direct activator of the master virulence regulator gene, prfA. This observation raised the possibility that CodY possesses multiple functions that allow it to coordinate gene expression across a wide spectrum of metabolic growth conditions, and thus better adapt bacteria to the mammalian niche. To gain a deeper understanding of CodY's regulatory repertoire and identify direct target genes, we performed a genome wide analysis of the CodY regulon and DNA binding under both rich and minimal growth conditions, using RNA-Seq and ChIP-Seq techniques. We demonstrate here that CodY is indeed active (i.e., binds DNA) under both conditions, serving as a repressor and activator of different genes. Further, we identified new genes and pathways that are directly regulated by CodY (e.g., sigB, arg, his, actA, glpF, gadG, gdhA, poxB, glnR and fla genes), integrating metabolism, stress responses, motility and virulence in L. monocytogenes. This study establishes CodY as a multifaceted factor regulating L. monocytogenes physiology in a highly versatile manner.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Systems Biology/methods , Bacterial Adhesion , Bacterial Proteins/genetics , Binding Sites , Caco-2 Cells , Cell Adhesion , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Flagella/metabolism , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Humans , Listeria monocytogenes/genetics , Models, Biological , Movement , Protein Binding , Regulon/genetics , Repressor Proteins/metabolism , Reproducibility of Results , Sequence Analysis, RNA , Transcription, Genetic , Virulence
16.
Nat Rev Microbiol ; 13(10): 641-50, 2015 10.
Article in English | MEDLINE | ID: mdl-26373372

ABSTRACT

Unlike lytic phages, temperate phages that enter lysogeny maintain a long-term association with their bacterial host. In this context, mutually beneficial interactions can evolve that support efficient reproduction of both phages and bacteria. Temperate phages are integrated into the bacterial chromosome as large DNA insertions that can disrupt gene expression, and they may pose a fitness burden on the cell. However, they have also been shown to benefit their bacterial hosts by providing new functions in a bacterium-phage symbiotic interaction termed lysogenic conversion. In this Opinion article, we discuss another type of bacterium-phage interaction, active lysogeny, in which phages or phage-like elements are integrated into the bacterial chromosome within critical genes or operons and serve as switches that regulate bacterial genes via genome excision.


Subject(s)
Gene Expression Regulation, Bacterial/physiology , Lysogeny/physiology , Prophages/physiology , Bacterial Physiological Phenomena , DNA Transformation Competence/physiology , Gene Expression Regulation, Viral/physiology , Nitrogen Fixation/genetics , Nitrogen Fixation/physiology , Phagosomes/physiology , Symbiosis/physiology , Virus Replication/physiology
17.
Infect Immun ; 83(6): 2358-68, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25824830

ABSTRACT

Human multidrug efflux transporters are known for their ability to extrude antibiotics and toxic compounds out of cells, yet accumulating data indicate they have additional functions in diverse physiological processes not related to drug efflux. Here, we show that the human multidrug transporter P-glycoprotein (P-gp) (also named MDR1 and ABCB1) is transcriptionally induced in the monocytic cell line THP-1 upon infection with the human intracellular bacterial pathogen Listeria monocytogenes. Notably, we found that P-gp is important for full activation of the type I interferon response elicited against L. monocytogenes bacteria. Both inhibition of P-gp function by verapamil and inhibition of its transcription using mRNA silencing led to a reduction in the magnitude of the type I response in infected cells. This function of P-gp was specific to type I interferon cytokines elicited against cytosolic replicating bacteria and was not observed in response to cyclic di-AMP (c-di-AMP), a molecule that was shown to be secreted by L. monocytogenes during infection and to trigger type I interferons. Moreover, P-gp was not involved in activation of other proinflammatory cytokines, such as those triggered by vacuolar-restricted L. monocytogenes or lipopolysaccharide (LPS). Taken together, these findings demonstrate a role for P-gp in proper development of an innate immune response against intracellular pathogens, highlighting the complexity in employing therapeutic strategies that involve inhibition of multidrug resistance (MDR) efflux pumps.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Interferon Type I/metabolism , Listeria monocytogenes/physiology , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Cell Line , Gene Expression Regulation , Humans , Interferon Type I/genetics , Interferon-beta , Macrophages , Mice , Mice, Knockout , Verapamil/pharmacology
18.
Mol Microbiol ; 95(4): 624-44, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25430920

ABSTRACT

Metabolic adaptations are critical to the ability of bacterial pathogens to grow within host cells and are normally preceded by sensing of host-specific metabolic signals, which in turn can influence the pathogen's virulence state. Previously, we reported that the intracellular bacterial pathogen Listeria monocytogenes responds to low availability of branched-chain amino acids (BCAAs) within mammalian cells by up-regulating both BCAA biosynthesis and virulence genes. The induction of virulence genes required the BCAA-responsive transcription regulator, CodY, but the molecular mechanism governing this mode of regulation was unclear. In this report, we demonstrate that CodY directly binds the coding sequence of the L. monocytogenes master virulence activator gene, prfA, 15 nt downstream of its start codon, and that this binding results in up-regulation of prfA transcription specifically under low concentrations of BCAA. Mutating this site abolished CodY binding and reduced prfA transcription in macrophages, and attenuated bacterial virulence in mice. Notably, the mutated binding site did not alter prfA transcription or PrfA activity under other conditions that are known to activate PrfA, such as during growth in the presence of glucose-1-phosphate. This study highlights the tight crosstalk between L. monocytogenes metabolism and virulence, while revealing novel features of CodY-mediated regulation.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Peptide Termination Factors/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Bacterial Proteins/metabolism , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Genes, Regulator , Glucosephosphates/metabolism , Listeria monocytogenes/growth & development , Listeria monocytogenes/pathogenicity , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mutation , Operon , Peptide Termination Factors/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Up-Regulation , Virulence/genetics
19.
Article in English | MEDLINE | ID: mdl-24611134

ABSTRACT

Multi-drug resistance (MDR) transporters are known eponymously for their ability to confer resistance to various antimicrobial drugs. However, it is likely that this is not their primary function and that MDR transporters evolved originally to play additional roles in bacterial physiology. In Listeria monocytogenes a set of MDR transporters was identified to mediate activation of innate immune responses during mammalian cell infection. This phenotype was shown to be dependent on c-di-AMP secretion, but the physiological processes underlying this phenomenon were not completely resolved. Here we describe a genetic approach taken to screen for L. monocytogenes genes or physiological pathways involved in MDR transporter-dependent triggering of the type I interferon response. We found that disruption of L. monocytogenes lipoteichoic acid (LTA) synthesis results in enhanced triggering of type I interferon responses in infected macrophage cells yet does not impact bacterial intracellular growth. This innate immune response required the MDR transporters and could be recapitulated by exposing macrophage cells to culture supernatants derived from LTA mutant bacteria. Notably, we found that the MDR transporters themselves are required for full production of LTA, an observation that links MDR transporters to LTA synthesis for the first time. In light of our findings, we propose that the MDR transporters play a role in regulating LTA synthesis, possibly via c-di-AMP efflux, a physiological function in cell wall maintenance that triggers the host innate immune system.


Subject(s)
Interferon Type I/immunology , Lipopolysaccharides/metabolism , Listeria monocytogenes/enzymology , Listeria monocytogenes/metabolism , Listeriosis/immunology , Membrane Transport Proteins/immunology , Membrane Transport Proteins/metabolism , Teichoic Acids/metabolism , Animals , Cells, Cultured , Dinucleoside Phosphates/metabolism , Immunity, Innate , Interferon Type I/metabolism , Lipopolysaccharides/immunology , Listeria monocytogenes/growth & development , Listeria monocytogenes/immunology , Listeriosis/microbiology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Teichoic Acids/immunology
20.
J Bacteriol ; 195(23): 5262-72, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24056100

ABSTRACT

Listeria monocytogenes is a Gram-positive human intracellular pathogen that infects diverse mammalian cells. Upon invasion, L. monocytogenes secretes multiple virulence factors that target host cellular processes and promote infection. It has been presumed, but was not empirically established, that the Sec translocation system is the primary mediator of this secretion. Here, we validate an important role for SecDF, a component of the Sec system, in the secretion of several critical L. monocytogenes virulence factors. A ΔsecDF mutant is demonstrated to exhibit impaired membrane translocation of listeriolysin O (LLO), PlcA, PlcB, and ActA, factors that mediate L. monocytogenes phagosomal escape and spread from cell to cell. This impaired translocation was monitored by accumulation of the factors on the bacterial membrane and by reduced activity upon secretion. This defect in secretion is shown to be associated with a severe intracellular growth defect of the ΔsecDF mutant in macrophages and a less virulent phenotype in mice, despite normal growth in laboratory medium. We further show that SecDF is upregulated when the bacteria reside in macrophage phagosomes and that it is necessary for efficient phagosomal escape. Taken together, these data support the premise that SecDF plays a role as a chaperone that facilitates the translocation of L. monocytogenes virulence factors during infection.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/metabolism , Molecular Chaperones/metabolism , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial/physiology , Listeria monocytogenes/genetics , Listeriosis/microbiology , Liver/microbiology , Mice , Spleen/microbiology , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...