Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters










Publication year range
1.
Infect Immun ; 92(4): e0034523, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38591895

ABSTRACT

Listeria monocytogenes is well recognized for both its broad resistance to stress conditions and its ability to transition from a soil bacterium to an intracellular pathogen of mammalian hosts. The bacterium's impressive ability to adapt to changing environments and conditions requires the rapid sensing of environmental cues and the coordinated response of gene products that enable bacterial growth and survival. Two-component signaling systems (TCSs) have been long recognized for their ability to detect environmental stimuli and transmit those signals into transcriptional responses; however, often the precise nature of the stimulus triggering TCS responses can be challenging to define. L. monocytogenes has up to 16 TCSs that have been recognized based on homology and included in this list are several whose functions remain poorly described. This review highlights the current understanding of the breadth and scope of L. monocytogenes TCS as relates to stress resistance and pathogenesis. Precise signals still often remain elusive, but the gene networks associated with TCSs are providing clues into possible functions.


Subject(s)
Listeria monocytogenes , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Listeria monocytogenes/genetics , Mammals , Signal Transduction
2.
Microbiol Spectr ; 11(3): e0331722, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37199604

ABSTRACT

The soil-dwelling bacterium Listeria monocytogenes survives a multitude of conditions when residing in the outside environment and as a pathogen within host cells. Key to survival within the infected mammalian host is the expression of bacterial gene products necessary for nutrient acquisition. Similar to many bacteria, L. monocytogenes uses peptide import to acquire amino acids. Peptide transport systems play an important role in nutrient uptake as well as in additional functions that include bacterial quorum sensing and signal transduction, recycling of peptidoglycan fragments, adherence to eukaryotic cells, and alterations in antibiotic susceptibility. It has been previously described that CtaP, encoded by lmo0135, is a multifunctional protein associated with activities that include cysteine transport, resistance to acid, membrane integrity, and bacterial adherence to host cells. ctaP is located next to two genes predicted to encode membrane-bound permeases lmo0136 and lmo0137, termed CtpP1 and CtpP2, respectively. Here, we show that CtpP1 and CtpP2 are required for bacterial growth in the presence of low concentrations of cysteine and for virulence in mouse infection models. Taken together, the data identify distinct nonoverlapping roles for two related permeases that are important for the growth and survival of L. monocytogenes within host cells. IMPORTANCE Bacterial peptide transport systems are important for nutrient uptake and may additionally function in a variety of other roles, including bacterial communication, signal transduction, and bacterial adherence to eukaryotic cells. Peptide transport systems often consist of a substrate-binding protein associated with a membrane-spanning permease. The environmental bacterial pathogen Listeria monocytogenes uses the substrate-binding protein CtaP not only for cysteine transport but also for resistance to acid, maintenance of membrane integrity, and bacterial adherence to host cells. In this study, we demonstrate complementary yet distinct functional roles for two membrane permeases, CtpP1 and CtpP2, that are encoded by genes linked to ctaP and that contribute to bacterial growth, invasion, and pathogenicity.


Subject(s)
Listeria monocytogenes , Animals , Mice , Listeria monocytogenes/genetics , Cysteine/metabolism , Virulence , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Virulence Factors/genetics , Bacterial Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation, Bacterial , Mammals
3.
Cells ; 13(1)2023 12 31.
Article in English | MEDLINE | ID: mdl-38201292

ABSTRACT

Vertically transmitted infections are a significant cause of fetal morbidity and mortality during pregnancy and pose substantial risks to fetal development. These infections are primarily transmitted to the fetus through two routes: (1) direct invasion and crossing the placenta which separates maternal and fetal circulation, or (2) ascending the maternal genitourinary tact and entering the uterus. Only two bacterial species are commonly found to cross the placenta and infect the fetus: Listeria monocytogenes and Treponema pallidum subsp. pallidum. L. monocytogenes is a Gram-positive, foodborne pathogen found in soil that acutely infects a wide variety of mammalian species. T. pallidum is a sexually transmitted spirochete that causes a chronic infection exclusively in humans. We briefly review the pathogenesis of these two very distinct bacteria that have managed to overcome the placental barrier and the role placental immunity plays in resisting infection. Both organisms share characteristics which contribute to their transplacental transmission. These include the ability to disseminate broadly within the host, evade immune phagocytosis, and the need for a strong T cell response for their elimination.


Subject(s)
Listeria monocytogenes , Treponema pallidum , Pregnancy , Humans , Animals , Female , Placenta , Treponema , Mammals
4.
Mol Microbiol ; 118(3): 278-293, 2022 09.
Article in English | MEDLINE | ID: mdl-35943959

ABSTRACT

Listeria monocytogenes (Lm) is a widespread environmental Gram-positive bacterium that can transition into a pathogen following ingestion by a susceptible host. To cross host barriers and establish infection, Lm is dependent upon the regulated secretion and activity of many proteins including PrsA2, a peptidyl-prolyl cis-trans isomerase with foldase activity. PrsA2 contributes to the stability and activity of a number of secreted virulence factors that are required for Lm invasion, replication, and cell-to-cell spread within the infected host. In contrast, a second related secretion chaperone, PrsA1, has thus far no identified contributions to Lm pathogenesis. Here we describe the characterization of a two-component signal transduction system PieRS that regulates the expression of a regulon that includes the secretion chaperones PrsA1 and PrsA2. PieRS regulated gene products are required for bacterial resistance to ethanol exposure and are important for bacterial survival during transit through the gastrointestinal tract. PrsA1 was also found to make a unique contribution to Lm survival in the GI tract, revealing for the first time a non-overlapping requirement for both secretion chaperones PrsA1 and PrsA2 during the process of intra-gastric infection.


Subject(s)
Listeria monocytogenes , Listeriosis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Translocation , Humans , Intestines , Listeria monocytogenes/genetics , Listeriosis/microbiology , Molecular Chaperones/metabolism , Virulence Factors/metabolism
5.
Infect Immun ; 89(4)2021 03 17.
Article in English | MEDLINE | ID: mdl-33495274

ABSTRACT

The Gram-positive bacterium Listeria monocytogenes survives in environments ranging from the soil to the cytosol of infected host cells. Key to L. monocytogenes intracellular survival is the activation of PrfA, a transcriptional regulator that is required for the expression of multiple bacterial virulence factors. Mutations that constitutively activate prfA (prfA* mutations) result in high-level expression of multiple bacterial virulence factors as well as the physiological adaptation of L. monocytogenes for optimal replication within host cells. Here, we demonstrate that L. monocytogenesprfA* mutants exhibit significantly enhanced resistance to oxidative stress in comparison to that of wild-type strains. Transposon mutagenesis of L. monocytogenesprfA* strains resulted in the identification of three novel gene targets required for full oxidative stress resistance only in the context of PrfA activation. One gene, lmo0779, predicted to encode an uncharacterized protein, and two additional genes known as cbpA and ygbB, encoding a cyclic di-AMP binding protein and a 2-C-methyl-d-erythritol 2,4-cyclodiphosphate synthase, respectively, contribute to the enhanced oxidative stress resistance of prfA* strains while exhibiting no significant contribution in wild-type L. monocytogenes Transposon inactivation of cbpA and lmo0779 in a prfA* background led to reduced virulence in the liver of infected mice. These results indicate that L. monocytogenes calls upon specific bacterial factors for stress resistance in the context of PrfA activation and thus under conditions favorable for bacterial replication within infected mammalian cells.


Subject(s)
Host-Pathogen Interactions , Listeria monocytogenes/genetics , Listeriosis/metabolism , Listeriosis/microbiology , Oxidative Stress , Virulence Factors/genetics , Animals , Bacterial Proteins/genetics , Disease Models, Animal , Female , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Listeria monocytogenes/drug effects , Listeria monocytogenes/pathogenicity , Mice , Mutagenesis, Insertional , Organ Specificity
6.
Infect Immun ; 89(2)2021 01 19.
Article in English | MEDLINE | ID: mdl-33139387

ABSTRACT

Listeria monocytogenes is a facultative Gram-positive intracellular bacterium that is capable of causing serious invasive infections in pregnant women, resulting in abortion, still-birth, and disseminated fetal infection. Previously, a clinical L. monocytogenes isolate, 07PF0776, was identified as having an enhanced ability to target cardiac tissue. This tissue tropism appeared to correlate with amino acid variations found within internalin B (InlB), a bacterial surface protein associated with host cell invasion. Given that the mammalian receptor bound by InlB, Met, is abundantly expressed by placental tissue, we assessed isolate 07PF0776 for its ability to be transmitted from mother to fetus. Pregnant Swiss Webster mice were infected on gestational day E13 via tail vein injection with the standard isolate 10403S, a noncardiotropic strain, or 07PF0776, the cardiac isolate. Pregnant mice infected with 07PF0776 exhibited significantly enhanced transmission of L. monocytogenes to placentas and fetuses compared to 10403S. Both bacterial burdens and the frequency of placental and fetal infection were increased in mice infected with the cardiac isolate. Strain 07PF0776 also exhibited an enhanced ability to invade Jar human trophoblast tissue culture cells in comparison to 10403S, and was found to have increased levels of InlB associated with the bacterial cell surface. Overexpression of surface InlB via genetic manipulation was sufficient to confer enhanced invasion of the placenta and fetus to both 10403S and 07PF0776. These data support a central role for surface InlB in promoting vertical transmission of L. monocytogenes.


Subject(s)
Bacterial Proteins/physiology , Fetus/physiopathology , Heart/physiopathology , Listeria monocytogenes/pathogenicity , Listeriosis/transmission , Membrane Proteins/physiology , Virulence/physiology , Adult , Female , Fetus/microbiology , Heart/microbiology , Humans , Infectious Disease Transmission, Vertical , Male , Pregnancy
8.
Microbiol Spectr ; 7(4)2019 07.
Article in English | MEDLINE | ID: mdl-31441398

ABSTRACT

Whereas obligate human and animal bacterial pathogens may be able to depend upon the warmth and relative stability of their chosen replication niche, environmental bacteria such as Listeria monocytogenes that harbor the ability to replicate both within animal cells and in the outside environment must maintain the capability to manage life under a variety of disparate conditions. Bacterial life in the outside environment requires adaptation to wide ranges of temperature, available nutrients, and physical stresses such as changes in pH and osmolarity as well as desiccation. Following ingestion by a susceptible animal host, the bacterium must adapt to similar changes during transit through the gastrointestinal tract and overcome a variety of barriers associated with host innate immune responses. Rapid alteration of patterns of gene expression and protein synthesis represent one strategy for quickly adapting to a dynamic host landscape. Here, we provide an overview of the impressive variety of strategies employed by the soil-dwelling, foodborne, mammalian pathogen L. monocytogenes to straddle diverse environments and optimize bacterial fitness both inside and outside host cells.


Subject(s)
Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Humans , Immunity, Innate , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Listeriosis/immunology , Osmolar Concentration , Virulence
9.
Biochim Biophys Acta Gen Subj ; 1863(8): 1283-1291, 2019 08.
Article in English | MEDLINE | ID: mdl-31059750

ABSTRACT

BACKGROUND: The scope of the present work was to characterize the activity of class IIa bacteriocins in Listeria (L.) monocytogenes cells that constitutively express an activated form of PrfA, the virulence master regulator, since bacteriocin sensitivity was only characterized in saprophytic cells so far. The mannose phosphotransferase system (Man-PTS) has been shown to be the class IIa bacteriocin receptor in Listeria; hence, special attention was paid to its expression in virulent bacteria. METHODS: L. monocytogenes FBprfA* cells were obtained by transconjugation. Bacterial growth was studied in TSB and glucose containing-minimal medium. Sensitivity to antimicrobial peptides was assessed by killing curves. Membranes of L. monocytogenes FBprfA* cells were characterized using proteomic and lipidomic approaches. RESULTS: The mannose phosphotransferase system (Man-PTS) was downregulated upon expression of PrfA*, and these cells turned out to be more sensitive to enterocin CRL35 and pediocin PA-1, while not to nisin. Proteomic and lipidomic analysis showed differences between wild type (WT) and PrfA* strains. For instance, phosphatidic acid was only detected in PrfA* cells, whereas, there was a significant decline of plasmalogen-phosphatidylglycerol in the same strain. CONCLUSIONS: Our results support a model in which Man-PTS acts just as a docking molecule that brings class IIa bacteriocins to the plasma membrane. Furthermore, our results suggest that lipids play a crucial role in the mechanism of action of bacteriocins. GENERAL SIGNIFICANCE: This is the first demonstration of the link between L. monocytogenes virulence and the bacterial sensitivity toward pediocin-like peptides.


Subject(s)
Bacterial Proteins/metabolism , Bacteriocins/metabolism , Listeria monocytogenes/metabolism , Peptide Termination Factors/metabolism , Receptors, Cell Surface/metabolism , Culture Media , Glucose/metabolism , Listeria monocytogenes/growth & development
10.
J Vis Exp ; (144)2019 02 22.
Article in English | MEDLINE | ID: mdl-30855571

ABSTRACT

There are a variety of strategies bacterial pathogens employ to survive and proliferate once inside the eukaryotic cell. The so-called 'cytosolic' pathogens (Listeria monocytogenes, Shigella flexneri, Burkholderia pseudomallei, Francisella tularensis, and Rickettsia spp.) gain access to the infected cell cytosol by physically and enzymatically degrading the primary vacuolar membrane. Once in the cytosol, these pathogens both proliferate as well as generate sufficient mechanical forces to penetrate the plasma membrane of the host cell in order to infect new cells. Here, we show how this terminal step of the cellular infection cycle of L. monocytogenes (Lm) can be quantified by both colony-forming unit assays and flow cytometry and give examples of how both pathogen- and host-encoded factors impact this process. We also show a close correspondence of Lm infection dynamics of cultured cells infected in vitro and those of hepatic cells derived from mice infected in vivo. These function-based assays are relatively simple and can be readily scaled up for discovery-based high-throughput screens for modulators of eukaryotic cell function.


Subject(s)
Bacterial Physiological Phenomena , Host-Pathogen Interactions , Animals , Cell Membrane/metabolism , Cell Membrane/microbiology , Cytosol/metabolism , Cytosol/microbiology , Humans , Mice
11.
mBio ; 9(6)2018 11 27.
Article in English | MEDLINE | ID: mdl-30482826

ABSTRACT

Bacillus subtilis has the capacity to choose between two mutually exclusive lifestyles: biofilm formation and flagellum-mediated swimming motility. Interestingly, this choice is made at the individual cell level, with bacterial cells in a population expressing genes required for biofilm formation or genes required for swimming motility but not both. A bistable switch controls the biofilm-versus-swimming decision, resulting in an evolutionarily favorable strategy known as "bet hedging" that ensures that subpopulations of bacteria continue to grow as conditions change and/or become unfavorable. In a recent issue of mBio, J. Kampf and colleagues (mBio 9:e01464-18, 2018, https://doi.org/10.1128/mBio.01464-18) reported the use of a combination of genetics and microfluidics to reveal that the interplay that occurs between the SinR and YmdB proteins underlies the B. subtilis choice between biofilm formation and swimming motility. Their report suggests that B. subtilis experiences selective pressure to form biofilms while maintaining reserve cell subpopulations with the capacity to swim away.


Subject(s)
Bacillus subtilis , Gene Expression Regulation, Bacterial , Bacterial Proteins/genetics , Biofilms , Mutation
12.
Nature ; 562(7725): 43-44, 2018 10.
Article in English | MEDLINE | ID: mdl-30275549
13.
Pathogens ; 7(2)2018 May 25.
Article in English | MEDLINE | ID: mdl-29799503

ABSTRACT

Protection of the developing fetus from pathogens is one of the many critical roles of the placenta. Listeria monocytogenes is one of a select number of pathogens that can cross the placental barrier and cause significant harm to the fetus, leading to spontaneous abortion, stillbirth, preterm labor, and disseminated neonate infection despite antibiotic treatment. Such severe outcomes serve to highlight the importance of understanding how L. monocytogenes mediates infiltration of the placental barrier. Here, we review what is currently known regarding vertical transmission of L. monocytogenes as a result of cell culture and animal models of infection. In vitro cell culture and organ models have been useful for the identification of L. monocytogenes virulence factors that contribute to placental invasion. Examples include members of the Internalin family of bacterial surface proteins such as Interalin (Inl)A, InlB, and InlP that promote invasion of cells at the maternal-fetal interface. A number of animal models have been used to interrogate L. monocytogenes vertical transmission, including mice, guinea pigs, gerbils, and non-human primates; each of these models has advantages while still not providing a comprehensive understanding of L. monocytogenes invasion of the human placenta and/or fetus. These models do, however, allow for the molecular investigation of the balance between fetal tolerance and immune protection from L. monocytogenes during pregnancy.

14.
Article in English | MEDLINE | ID: mdl-28702378

ABSTRACT

Microbes employ the thioredoxin system to defend against oxidative stress and ensure correct disulfide bonding to maintain protein function. Listeria monocytogenes has been shown to encode a putative thioredoxin, TrxA, but its biological roles and underlying mechanisms remain unknown. Here, we showed that expression of L. monocytogenes TrxA is significantly induced in bacteria treated with the thiol-specific oxidizing agent, diamide. Deletion of trxA markedly compromised tolerance of the pathogen to diamide, and mainly impaired early stages of infection in human intestinal epithelial Caco-2 cells. In addition, most trxA mutant bacteria were not associated with polymerized actin, and the rare bacteria that were associated with polymerized actin displayed very short tails or clouds during infection. Deletion or constitutive overexpression of TrxA, which was regulated by SigH, severely attenuated the virulence of the pathogen. Transcriptome analysis of L. monocytogenes revealed over 270 genes that were differentially transcribed in the ΔtrxA mutant compared to the wild-type, especially for the virulence-associated genes plcA, mpl, hly, actA, and plcB. Particularly, deletion of TrxA completely reduced LLO expression, and thereby led to a thoroughly impaired hemolytic activity. Expression of these virulence factors are positively regulated by the master regulator PrfA that was found here to use TrxA to maintain its reduced forms for activation. Interestingly, the trxA deletion mutant completely lacked flagella and was non-motile. We further confirmed that this deficiency is attributable to TrxA in maintaining the reduced intracellular monomer status of MogR, the key regulator for flagellar formation, to ensure correct dimerization. In summary, we demonstrated for the first time that L. monocytogenes thioredoxin A as a vital cellular reductase is essential for maintaining a highly reducing environment in the bacterial cytosol, which provides a favorable condition for protein folding and activation, and therefore contributes to bacterial virulence and motility.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/cytology , Listeria monocytogenes/metabolism , Listeriosis/microbiology , Thioredoxins/metabolism , Bacterial Proteins/genetics , Caco-2 Cells , Cytosol/metabolism , Flagella/genetics , Flagella/metabolism , Gene Expression Regulation, Bacterial , Humans , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Listeriosis/metabolism , Oxidation-Reduction , Thioredoxins/genetics , Virulence
15.
Infect Immun ; 85(7)2017 07.
Article in English | MEDLINE | ID: mdl-28461390

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) is responsible for large numbers of postsurgical nosocomial infections across the United States and worldwide. Propofol anesthesia is widely used in surgery and in intensive care units, and recent evidence indicates that even brief exposure to propofol can substantially increase host susceptibility to microbial infection. Here, we delineate the impact of propofol sedation on MRSA bloodstream infections in mice in the presence and absence of prophylactic antibiotic treatment. Consistent with previous reports, brief periods of anesthesia with propofol were sufficient to significantly increase bacterial burdens and kidney pathology in mice infected with MRSA. Propofol exposure increased neutrophilic infiltrates into the kidney and enhanced bacterial dissemination throughout kidney tissue. Propofol sedation reduced populations of effector phagocytes and mature dendritic cells within the kidney and led to the apparent expansion of myeloid-derived suppressor cell-like populations. When propofol was coadministered with vancomycin prophylaxis, it dramatically increased kidney abscess formation and bacterial dissemination throughout kidney tissue at early times post-S. aureus infection compared to antibiotic-treated but nonsedated animals. Taken together, our data indicate that short-term sedation with propofol significantly increases the severity of bloodstream MRSA infection, even when administered in conjunction with vancomycin prophylaxis.


Subject(s)
Bacteremia/pathology , Disease Susceptibility/chemically induced , Hypnotics and Sedatives/adverse effects , Kidney Diseases/chemically induced , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Propofol/adverse effects , Staphylococcal Infections/pathology , Animals , Bacterial Load , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Female , Kidney Diseases/pathology , Macrophages/drug effects , Macrophages/immunology , Mice , Neutrophils/drug effects , Neutrophils/immunology
16.
Microb Pathog ; 105: 218-225, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28242425

ABSTRACT

Infections by pathogenic microorganisms elicit host immune responses, which crucially limit those infections. Pathogens employ various strategies to evade host immunity. We have identified the exploitation of the repertoire of potent immunosuppressive responses elicited normally by apoptotic cells ("Innate Apoptotic Immunity"; IAI) as one of these strategies. In the case of Listeria monocytogenes, an environmentally ubiquitous, foodborne bacterial pathogen capable of causing life-threatening invasive disease in immunocompromised and elderly individuals, the induction of host cell apoptosis appears to play an important role in pathogenesis. Previous studies have documented extensive lymphocyte apoptosis resulting from L. monocytogenes infection and demonstrated paradoxically that lymphocyte-deficient animals exhibit diminished susceptibility to listerial pathogenicity. We speculated that the triggering of IAI following the induction of host cell apoptosis was responsible for enhanced pathogenesis, and that the administration of exogenous apoptotic cells would serve to exert this effect. Importantly, apoptotic cells, which are not susceptible to L. monocytogenes infection, do not provide a niche for bacterial replication. Our experiments confirm that apoptotic cells, including exogenous apoptotic cells induced to die independently of the pathogen, specifically enhance pathogenesis. The recognition of a role of apoptotic cells and Innate Apoptotic Immunity in microbial pathogenesis provides an intriguing and novel insight for therapeutic approaches for the control of pathogenic infections.


Subject(s)
Apoptosis/immunology , Listeria monocytogenes/pathogenicity , Listeriosis/immunology , Listeriosis/pathology , Animals , Cell Line , Cells, Cultured , Cytokines/immunology , Disease Models, Animal , Immunity, Innate , Inflammation/immunology , Inflammation/pathology , Listeria monocytogenes/immunology , Listeriosis/microbiology , Lymphocytes/immunology , Mice, Inbred C57BL , Signal Transduction
17.
Structure ; 25(2): 295-304, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28089449

ABSTRACT

Active in the aqueous cellular environment where a massive excess of water is perpetually present, enzymes that catalyze the transfer of an electrophile to a non-water nucleophile (transferases) require specific strategies to inhibit mechanistically related hydrolysis reactions. To identify principles that confer transferase versus hydrolase reaction specificity, we exploited two enzymes that use highly similar catalytic apparatuses to catalyze the transglycosylation (a transferase reaction) or hydrolysis of α-1,3-glucan linkages in the cyclic tetrasaccharide cycloalternan (CA). We show that substrate binding to non-catalytic domains and a conformationally stable active site promote CA transglycosylation, whereas a distinct pattern of active site conformational change is associated with CA hydrolysis. These findings defy the classic view of induced-fit conformational change and illustrate a mechanism by which a stable hydrophobic binding site can favor transferase activity and disfavor hydrolysis. Application of these principles could facilitate the rational reengineering of transferases with desired catalytic properties.


Subject(s)
Actinomycetales/enzymology , Glucosidases/chemistry , Glycoside Hydrolases/chemistry , Listeria monocytogenes/enzymology , Oligosaccharides/chemistry , Water/chemistry , Actinomycetales/genetics , Amino Acid Motifs , Binding Sites , Biocatalysis , Carbohydrate Conformation , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Glucosidases/genetics , Glucosidases/metabolism , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Glycosylation , Hydrolysis , Hydrophobic and Hydrophilic Interactions , Kinetics , Listeria monocytogenes/genetics , Models, Molecular , Oligosaccharides/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Water/metabolism
18.
Nat Microbiol ; 2: 16202, 2016 Nov 07.
Article in English | MEDLINE | ID: mdl-27819654

ABSTRACT

Here we employ a 'systems structural biology' approach to functionally characterize an unconventional α-glucan metabolic pathway from the food-borne pathogen Listeria monocytogenes (Lm). Crystal structure determination coupled with basic biochemical and biophysical assays allowed for the identification of anabolic, transport, catabolic and regulatory portions of the cycloalternan pathway. These findings provide numerous insights into cycloalternan pathway function and reveal the mechanism of repressor, open reading frame, kinase (ROK) transcription regulators. Moreover, by developing a structural overview we were able to anticipate the cycloalternan pathway's role in the metabolism of partially hydrolysed starch derivatives and demonstrate its involvement in Lm pathogenesis. These findings suggest that the cycloalternan pathway plays a role in interspecies resource competition-potentially within the host gastrointestinal tract-and establish the methodological framework for characterizing bacterial systems of unknown function.


Subject(s)
Enzymes/chemistry , Enzymes/metabolism , Listeria monocytogenes/enzymology , Listeria monocytogenes/metabolism , Metabolic Networks and Pathways , Oligosaccharides/metabolism , Biophysical Phenomena , Crystallography, X-Ray , Protein Conformation
19.
Future Microbiol ; 11: 1447-1459, 2016 10.
Article in English | MEDLINE | ID: mdl-27750441

ABSTRACT

A number of bacterial pathogens are capable of detecting the presence of other bacteria located within their surrounding niche through a process of bacterial signaling and cell-to-cell communication commonly referred to as quorum sensing (QS). QS systems are commonly now described in the context of collective behaviors exhibited by groups of bacteria coordinating diverse arrays of physiological functions to enhance survival of the community. However, QS systems have also been implicated in a variety of processes distinct from the measure of bacterial cell density. This review will highlight noncanonical adaptations of canonical QS systems that have evolved to enable bacteria to detect nonself individuals within a population or to detect occupation of confined spaces.


Subject(s)
Bacteria/metabolism , Bacterial Physiological Phenomena , Quorum Sensing/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Communication/physiology , Cell Count , Enterococcus/genetics , Enterococcus/metabolism , Gene Expression Regulation, Bacterial , Host-Parasite Interactions/physiology , Listeria/genetics , Listeria/metabolism , Signal Transduction/physiology , Staphylococcus/genetics , Staphylococcus/metabolism , Vacuoles/microbiology
20.
Infect Immun ; 84(10): 3034-46, 2016 10.
Article in English | MEDLINE | ID: mdl-27481256

ABSTRACT

The Gram-positive bacterium Listeria monocytogenes transitions from an environmental organism to an intracellular pathogen following its ingestion by susceptible mammalian hosts. Bacterial replication within the cytosol of infected cells requires activation of the central virulence regulator PrfA followed by a PrfA-dependent induction of secreted virulence factors. The PrfA-induced secreted chaperone PrsA2 and the chaperone/protease HtrA contribute to the folding and stability of select proteins translocated across the bacterial membrane. L. monocytogenes strains that lack both prsA2 and htrA exhibit near-normal patterns of growth in broth culture but are severely attenuated in vivo We hypothesized that, in the absence of PrsA2 and HtrA, the increase in PrfA-dependent protein secretion that occurs following bacterial entry into the cytosol results in misfolded proteins accumulating at the bacterial membrane with a subsequent reduction in intracellular bacterial viability. Consistent with this hypothesis, the introduction of a constitutively activated allele of prfA (prfA*) into ΔprsA2 ΔhtrA strains was found to essentially inhibit bacterial growth at 37°C in broth culture. ΔprsA2 ΔhtrA strains were additionally found to be defective for cell invasion and vacuole escape in selected cell types, steps that precede full PrfA activation. These data establish the essential requirement for PrsA2 and HtrA in maintaining bacterial growth under conditions of PrfA activation. In addition, chaperone function is required for efficient bacterial invasion and rapid vacuole lysis within select host cell types, indicating roles for PrsA2/HtrA prior to cytosolic PrfA activation and the subsequent induction of virulence factor secretion.


Subject(s)
Heat-Shock Proteins/physiology , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Molecular Chaperones/physiology , Peptidylprolyl Isomerase/physiology , Serine Endopeptidases/physiology , Animals , Cytoplasm/microbiology , Epithelial Cells/microbiology , Glucuronidase/metabolism , Humans , Listeria monocytogenes/growth & development , Listeria monocytogenes/physiology , Macrophages/microbiology , Mice , Molecular Chaperones/metabolism , Protein Folding , Protein Stability , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...