Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
PLoS One ; 19(5): e0301624, 2024.
Article in English | MEDLINE | ID: mdl-38713678

ABSTRACT

Salmonella enterica serovar Typhi (S. Typhi) is the causative agent of Typhoid fever. Blood culture is the gold standard for clinical diagnosis, but this is often difficult to employ in resource limited settings. Environmental surveillance of waste-impacted waters is a promising supplement to clinical surveillance, however validating methods is challenging in regions where S. Typhi concentrations are low. To evaluate existing S. Typhi environmental surveillance methods, a novel process control organism (PCO) was created as a biosafe surrogate. Using a previous described qPCR assay, a modified PCR amplicon for the staG gene was cloned into E. coli. We developed a target region that was recognized by the Typhoid primers in addition to a non-coding internal probe sequence. A multiplex qPCR reaction was developed that differentiates between the typhoid and control targets, with no cross-reactivity or inhibition of the two probes. The PCO was shown to mimic S. Typhi in lab-based experiments with concentration methods using primary wastewater: filter cartridge, recirculating Moore swabs, membrane filtration, and differential centrifugation. Across all methods, the PCO seeded at 10 CFU/mL and 100 CFU/mL was detected in 100% of replicates. The PCO is detected at similar quantification cycle (Cq) values across all methods at 10 CFU/mL (Average = 32.4, STDEV = 1.62). The PCO was also seeded into wastewater at collection sites in Vellore (India) and Blantyre (Malawi) where S. Typhi is endemic. All methods tested in both countries were positive for the seeded PCO. The PCO is an effective way to validate performance of environmental surveillance methods targeting S. Typhi in surface water.


Subject(s)
Environmental Monitoring , Escherichia coli , Salmonella typhi , Salmonella typhi/genetics , Salmonella typhi/isolation & purification , Escherichia coli/genetics , Escherichia coli/isolation & purification , Environmental Monitoring/methods , Wastewater/microbiology , Typhoid Fever/microbiology , Typhoid Fever/epidemiology , Typhoid Fever/diagnosis , Typhoid Fever/prevention & control , Humans , Water Microbiology
2.
mBio ; 15(4): e0045424, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38497655

ABSTRACT

Salmonella serovars Typhi and Paratyphi cause a prolonged illness known as enteric fever, whereas other serovars cause acute gastroenteritis. Mechanisms responsible for the divergent clinical manifestations of nontyphoidal and enteric fever Salmonella infections have remained elusive. Here, we show that S. Typhi and S. Paratyphi A can persist within human macrophages, whereas S. Typhimurium rapidly induces apoptotic macrophage cell death that is dependent on Salmonella pathogenicity island 2 (SPI2). S. Typhi and S. Paratyphi A lack 12 specific SPI2 effectors with pro-apoptotic functions, including nine that target nuclear factor κB (NF-κB). Pharmacologic inhibition of NF-κB or heterologous expression of the SPI2 effectors GogA or GtgA restores apoptosis of S. Typhi-infected macrophages. In addition, the absence of the SPI2 effector SarA results in deficient signal transducer and activator of transcription 1 (STAT1) activation and interleukin 12 production, leading to impaired TH1 responses in macrophages and humanized mice. The absence of specific nontyphoidal SPI2 effectors may allow S. Typhi and S. Paratyphi A to cause chronic infections. IMPORTANCE: Salmonella enterica is a common cause of gastrointestinal infections worldwide. The serovars Salmonella Typhi and Salmonella Paratyphi A cause a distinctive systemic illness called enteric fever, whose pathogenesis is incompletely understood. Here, we show that enteric fever Salmonella serovars lack 12 specific virulence factors possessed by nontyphoidal Salmonella serovars, which allow the enteric fever serovars to persist within human macrophages. We propose that this fundamental difference in the interaction of Salmonella with human macrophages is responsible for the chronicity of typhoid and paratyphoid fever, suggesting that targeting the nuclear factor κB (NF-κB) complex responsible for macrophage survival could facilitate the clearance of persistent bacterial infections.


Subject(s)
Salmonella typhi , Salmonella , Typhoid Fever , Humans , Animals , Mice , Salmonella typhi/genetics , Typhoid Fever/microbiology , NF-kappa B , Macrophages/microbiology
3.
Lancet Infect Dis ; 23(6): 740-750, 2023 06.
Article in English | MEDLINE | ID: mdl-36731480

ABSTRACT

BACKGROUND: Shigella spp have been associated with community-wide outbreaks in urban settings. We analysed a sustained shigellosis outbreak in Seattle, WA, USA, to understand its origins and mechanisms of antimicrobial resistance, define ongoing transmission patterns, and optimise strategies for treatment and infection control. METHODS: We did a retrospective study of all Shigella isolates identified from stool samples at the clinical laboratories at Harborview Medical Center and University of Washington Medical Center (Seattle, WA, USA) from May 1, 2017, to Feb 28, 2022. We characterised isolates by species identification, phenotypic susceptibility testing, and whole-genome sequencing. Demographic characteristics and clinical outcomes of the patients were retrospectively examined. FINDINGS: 171 cases of shigellosis were included. 78 (46%) patients were men who have sex with men (MSM), and 88 (52%) were people experiencing homelessness (PEH). Although 84 (51%) isolates were multidrug resistant, 100 (70%) of 143 patients with data on antimicrobial therapy received appropriate empirical therapy. Phylogenomic analysis identified sequential outbreaks of multiple distinct lineages of Shigella flexneri and Shigella sonnei. Discrete clonal lineages (ten in S flexneri and nine in S sonnei) and resistance traits were responsible for infection in different at-risk populations (ie, MSM, PEH), enabling development of effective guidelines for empirical treatment. The most prevalent lineage in Seattle was probably introduced to Washington State via international travel, with subsequent domestic transmission between at-risk groups. INTERPRETATION: An outbreak in Seattle was driven by parallel emergence of multidrug-resistant strains involving international transmission networks and domestic transmission between at-risk populations. Genomic analysis elucidated not only outbreak origin, but directed optimal approaches to testing, treatment, and public health response. Rapid diagnostics combined with detailed knowledge of local epidemiology can enable high rates of appropriate empirical therapy even in multidrug-resistant infection. FUNDING: None.


Subject(s)
Anti-Infective Agents , Dysentery, Bacillary , Sexual and Gender Minorities , Shigella , Male , Humans , Female , Dysentery, Bacillary/drug therapy , Dysentery, Bacillary/epidemiology , Homosexuality, Male , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Retrospective Studies , Washington/epidemiology , Shigella/genetics , Disease Outbreaks , Anti-Infective Agents/therapeutic use , Genomics , Microbial Sensitivity Tests
4.
Methods Mol Biol ; 2427: 215-234, 2022.
Article in English | MEDLINE | ID: mdl-35619037

ABSTRACT

Efforts to understand molecular mechanisms of pathogenesis of the human-restricted pathogen Salmonella enterica serovar Typhi, the causative agent of typhoid fever, have been hampered by the lack of a tractable small animal model. This obstacle has been surmounted by a humanized mouse model in which genetically modified mice are engrafted with purified CD34+ stem cells from human umbilical cord blood, designated CD34+ Hu-NSG (formerly hu-SRC-SCID) mice. We have shown that these mice develop a lethal systemic infection with S. Typhi that is dependent on the presence of engrafted human hematopoietic cells. Immunological and pathological features of human typhoid are recapitulated in this model, which has been successfully employed for the identification of bacterial genetic determinants of S. Typhi virulence. Here we describe the methods used to infect CD34+ Hu-NSG mice with S. Typhi in humanized mice and to construct and analyze a transposon-directed insertion site sequencing S. Typhi library, and provide general considerations for the use of humanized mice for the study of a human-restricted pathogen.


Subject(s)
Salmonella typhi , Typhoid Fever , Animals , Disease Models, Animal , Mice , Mice, SCID , Salmonella typhi/genetics , Typhoid Fever/microbiology , Typhoid Fever/pathology , Virulence/genetics
5.
Infect Immun ; 90(1): e0047921, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34662213

ABSTRACT

A variety of eubacteria, plants, and protozoa can modify membrane lipids by cyclopropanation, which is reported to modulate membrane permeability and fluidity. The ability to cyclopropanate membrane lipids has been associated with resistance to oxidative stress in Mycobacterium tuberculosis, organic solvent stress in Escherichia coli, and acid stress in E. coli and Salmonella. In bacteria, the cfa gene encoding cyclopropane fatty acid (CFA) synthase is induced during the stationary phase of growth. In the present study, we constructed a cfa mutant of Salmonella enterica serovar Typhimurium 14028s (S. Typhimurium) and determined the contribution of CFA-modified lipids to stress resistance and virulence in mice. Cyclopropane fatty acid content was quantified in wild-type and cfa mutant S. Typhimurium. CFA levels in the cfa mutant were greatly reduced compared to CFA levels in the wild type, indicating that CFA synthase is the major enzyme responsible for cyclopropane modification of lipids in Salmonella. S. Typhimurium cfa mutants were more sensitive to extreme acid pH, the protonophore CCCP, and hydrogen peroxide compared to the wild type. In addition, cfa mutants exhibited reduced viability in murine macrophages and could be rescued by the addition of the NADPH phagocyte oxidase inhibitor diphenyleneiodonium (DPI) chloride. S. Typhimurium lacking cfa was also attenuated for virulence in mice. These observations indicate that CFA modification of lipids makes an important contribution to Salmonella virulence.


Subject(s)
Cyclopropanes/metabolism , Fatty Acids/metabolism , Salmonella Infections/microbiology , Salmonella typhimurium/physiology , Animals , Bacterial Physiological Phenomena , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biosynthetic Pathways , Cyclopropanes/chemistry , Cyclopropanes/pharmacology , Disease Models, Animal , Fatty Acids/chemistry , Fatty Acids/pharmacology , Hydrogen-Ion Concentration , Macrophages/drug effects , Macrophages/immunology , Macrophages/microbiology , Mice , Microbial Viability/drug effects , Microbial Viability/immunology , Mutation , Oxidative Stress , Salmonella Infections/immunology , Salmonella Infections/mortality , Salmonella typhimurium/drug effects , Virulence
6.
Metallomics ; 12(11): 1791-1801, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33078811

ABSTRACT

Nitric oxide (NO˙) is a radical molecule produced by mammalian phagocytic cells as part of the innate immune response to bacterial pathogens. It exerts its antimicrobial activity in part by impairing the function of metalloproteins, particularly those containing iron and zinc cofactors. The pathogenic Gram-negative bacterium Salmonella enterica serovar typhimurium undergoes dynamic changes in its cellular content of the four most common metal cofactors following exposure to NO˙ stress. Zinc, iron and magnesium all decrease in response to NO˙ while cellular manganese increases significantly. Manganese acquisition is driven primarily by increased expression of the mntH and sitABCD transporters following derepression of MntR and Fur. ZupT also contributes to manganese acquisition in response to nitrosative stress. S. Typhimurium mutants lacking manganese importers are more sensitive to NO˙, indicating that manganese is important for resistance to nitrosative stress.


Subject(s)
Manganese/metabolism , Nitrosative Stress , Salmonella typhimurium/metabolism , Bacterial Proteins/metabolism , Biological Transport , Cation Transport Proteins/metabolism , Colony Count, Microbial , Gene Expression Regulation, Bacterial , Iron/metabolism , Magnesium/metabolism , Microbial Viability , Mutation/genetics , Nitric Oxide/metabolism , Salmonella typhimurium/genetics , Up-Regulation/genetics
7.
Cell Host Microbe ; 26(3): 426-434.e6, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31447308

ABSTRACT

Salmonella enterica serovar Typhi causes typhoid fever only in humans. Murine infection with S. Typhimurium is used as a typhoid model, but its relevance to human typhoid is limited. Non-obese diabetic-scid IL2rγnull mice engrafted with human hematopoietic stem cells (hu-SRC-SCID) are susceptible to lethal S. Typhi infection. In this study, we use a high-density S. Typhi transposon library in hu-SRC-SCID mice to identify virulence loci using transposon-directed insertion site sequencing (TraDIS). Vi capsule, lipopolysaccharide (LPS), and aromatic amino acid biosynthesis were essential for virulence, along with the siderophore salmochelin. However, in contrast to the murine S. Typhimurium model, neither the PhoPQ two-component system nor the SPI-2 pathogenicity island was required for lethal S. Typhi infection, nor was the CdtB typhoid toxin. These observations highlight major differences in the pathogenesis of typhoid and non-typhoidal Salmonella infections and demonstrate the utility of humanized mice for understanding the pathogenesis of a human-specific pathogen.


Subject(s)
Genome-Wide Association Study/methods , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella typhi/genetics , Salmonella typhi/pathogenicity , Amino Acids, Aromatic/biosynthesis , Animals , Bacterial Proteins/genetics , Bacterial Toxins/genetics , DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Genomic Islands/genetics , Humans , Interleukin Receptor Common gamma Subunit/genetics , Iron/metabolism , Lipopolysaccharides/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Mice, Obese , Mice, SCID , Salmonella typhi/growth & development , Siderophores/metabolism , THP-1 Cells/microbiology , Typhoid Fever , Virulence/genetics
8.
mBio ; 10(2)2019 03 05.
Article in English | MEDLINE | ID: mdl-30837332

ABSTRACT

Gene duplication and subsequent evolutionary divergence have allowed conserved proteins to develop unique roles. The MarR family of transcription factors (TFs) has undergone extensive duplication and diversification in bacteria, where they act as environmentally responsive repressors of genes encoding efflux pumps that confer resistance to xenobiotics, including many antimicrobial agents. We have performed structural, functional, and genetic analyses of representative members of the SlyA/RovA lineage of MarR TFs, which retain some ancestral functions, including repression of their own expression and that of divergently transcribed multidrug efflux pumps, as well as allosteric inhibition by aromatic carboxylate compounds. However, SlyA and RovA have acquired the ability to countersilence horizontally acquired genes, which has greatly facilitated the evolution of Enterobacteriaceae by horizontal gene transfer. SlyA/RovA TFs in different species have independently evolved novel regulatory circuits to provide the enhanced levels of expression required for their new role. Moreover, in contrast to MarR, SlyA is not responsive to copper. These observations demonstrate the ability of TFs to acquire new functions as a result of evolutionary divergence of both cis-regulatory sequences and in trans interactions with modulatory ligands.IMPORTANCE Bacteria primarily evolve via horizontal gene transfer, acquiring new traits such as virulence and antibiotic resistance in single transfer events. However, newly acquired genes must be integrated into existing regulatory networks to allow appropriate expression in new hosts. This is accommodated in part by the opposing mechanisms of xenogeneic silencing and countersilencing. An understanding of these mechanisms is necessary to understand the relationship between gene regulation and bacterial evolution. Here we examine the functional evolution of an important lineage of countersilencers belonging to the ancient MarR family of classical transcriptional repressors. We show that although members of the SlyA lineage retain some ancestral features associated with the MarR family, their cis-regulatory sequences have evolved significantly to support their new function. Understanding the mechanistic requirements for countersilencing is critical to understanding the pathoadaptation of emerging pathogens and also has practical applications in synthetic biology.


Subject(s)
Enterobacteriaceae/genetics , Evolution, Molecular , Gene Expression Regulation, Bacterial , Gene Silencing , Transcription Factors/genetics , Gene Transfer, Horizontal
9.
mBio ; 9(4)2018 08 14.
Article in English | MEDLINE | ID: mdl-30108168

ABSTRACT

Nitric oxide (NO·) produced by mammalian cells exerts antimicrobial actions that result primarily from the modification of protein thiols (S-nitrosylation) and metal centers. A comprehensive approach was used to identify novel targets of NO· in Salmonella enterica serovar Typhimurium (S. Typhimurium). Newly identified targets include zinc metalloproteins required for DNA replication and repair (DnaG, PriA, and TopA), protein synthesis (AlaS and RpmE), and various metabolic activities (ClpX, GloB, MetE, PepA, and QueC). The cytotoxic actions of free zinc are mitigated by the ZntA and ZitB zinc efflux transporters, which are required for S. Typhimurium resistance to zinc overload and nitrosative stress in vitro Zinc efflux also ameliorates NO·-dependent zinc mobilization following internalization by activated macrophages and is required for virulence in NO·-producing mice, demonstrating that host-derived NO· causes zinc stress in intracellular bacteria.IMPORTANCE Nitric oxide (NO·) is produced by macrophages in response to inflammatory stimuli and restricts the growth of intracellular bacteria. Mechanisms of NO·-dependent antimicrobial actions are incompletely understood. Here, we show that zinc metalloproteins are important targets of NO· in Salmonella, including the DNA replication proteins DnaG and PriA, which were hypothesized to be NO· targets in earlier studies. Like iron, zinc is a cofactor for several essential proteins but is toxic at elevated concentrations. This study demonstrates that NO· mobilizes free zinc in Salmonella and that specific efflux transporters ameliorate the cytotoxic effects of free zinc during infection.


Subject(s)
Anti-Bacterial Agents/metabolism , Homeostasis/drug effects , Nitric Oxide/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/metabolism , Zinc/metabolism , Zinc/toxicity , Animals , Macrophages/immunology , Mice , Microbial Viability , RAW 264.7 Cells , Salmonella Infections, Animal/immunology
10.
Cell Host Microbe ; 23(5): 594-606.e7, 2018 05 09.
Article in English | MEDLINE | ID: mdl-29706505

ABSTRACT

Staphylococcus aureus is a commensal bacterium that can asymptomatically colonize its host but also causes invasive infections. Quorum sensing regulates S. aureus virulence and the transition from a commensal to a pathogenic organism. However, little is known about how host innate immunity affects interbacterial communication. We show that nitric oxide suppresses staphylococcal virulence by targeting the Agr quorum sensing system. Nitric oxide-mediated inhibition occurs through direct modification of cysteine residues C55, C123, and C199 of the AgrA transcription factor. Cysteine modification decreases AgrA promoter occupancy as well as transcription of the agr operon and quorum sensing-activated toxin genes. In a staphylococcal pneumonia model, mice lacking inducible nitric oxide synthase develop more severe disease with heightened mortality and proinflammatory cytokine responses. In addition, staphylococcal α-toxin production increases in the absence of nitric oxide or nitric oxide-sensitive AgrA cysteine residues. Our findings demonstrate an anti-virulence mechanism for nitric oxide in innate immunity.


Subject(s)
Cell Communication/immunology , Cell Communication/physiology , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Nitric Oxide/antagonists & inhibitors , Staphylococcus/drug effects , Staphylococcus/pathogenicity , Animals , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Cysteine , Cytokines/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Bacterial/drug effects , Hemolysin Proteins/metabolism , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Operon , Pneumonia, Staphylococcal/metabolism , Pneumonia, Staphylococcal/pathology , Promoter Regions, Genetic/drug effects , Quorum Sensing/drug effects , Quorum Sensing/genetics , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Trans-Activators/metabolism , Transcription Factors/drug effects , Virulence/drug effects
11.
mBio ; 8(3)2017 06 06.
Article in English | MEDLINE | ID: mdl-28588134

ABSTRACT

The Rcs phosphorelay and Psp (phage shock protein) systems are envelope stress responses that are highly conserved in gammaproteobacteria. The Rcs regulon was found to be strongly induced during metal deprivation of Salmonella enterica serovar Typhimurium lacking the Psp response. Nineteen genes activated by the RcsA-RcsB response regulator make up an operon responsible for the production of colanic acid capsular polysaccharide, which promotes biofilm development. Despite more than half a century of research, the physiological function of colanic acid has remained elusive. Here we show that Rcs-dependent colanic acid production maintains the transmembrane electrical potential and proton motive force in cooperation with the Psp response. Production of negatively charged exopolysaccharide covalently bound to the outer membrane may enhance the surface potential by increasing the local proton concentration. This provides a unifying mechanism to account for diverse Rcs/colanic acid-related phenotypes, including susceptibility to membrane-damaging agents and biofilm formation.IMPORTANCE Colanic acid is a negatively charged polysaccharide capsule produced by Escherichia coli, Salmonella, and other gammaproteobacteria. Research conducted over the 50 years since the discovery of colanic acid suggests that this exopolysaccharide plays an important role for bacteria living in biofilms. However, a precise physiological role for colanic acid has not been defined. In this study, we provide evidence that colanic acid maintains the transmembrane potential and proton motive force during envelope stress. This work provides a new and fundamental insight into bacterial physiology.


Subject(s)
Bacterial Capsules/physiology , Bacterial Proteins/genetics , Heat-Shock Proteins/genetics , Membrane Potentials , Polysaccharides, Bacterial/metabolism , Polysaccharides/metabolism , Salmonella typhimurium/physiology , Bacterial Proteins/metabolism , Biofilms , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Heat-Shock Proteins/metabolism , Polysaccharides/biosynthesis , Polysaccharides/genetics , Polysaccharides, Bacterial/chemistry , Proton-Motive Force , Salmonella typhimurium/genetics
12.
Nat Microbiol ; 2: 16224, 2016 Nov 28.
Article in English | MEDLINE | ID: mdl-27892921

ABSTRACT

Nitric oxide (NO•) is a ubiquitous molecular mediator in biology. Many signalling actions of NO• generated by mammalian NO• synthase (NOS) result from targeting of the haem moiety of soluble guanylate cyclase. Some pathogenic and environmental bacteria also produce a NOS that is evolutionary related to the mammalian enzymes, but a bacterial haem-containing receptor for endogenous enzymatically generated NO• has not been identified previously. Here, we show that NOS of the human pathogen Staphylococcus aureus, in concert with an NO•-metabolizing flavohaemoprotein, regulates electron transfer by targeting haem-containing cytochrome oxidases under microaerobic conditions to maintain membrane bioenergetics. This process is essential for staphylococcal nasal colonization and resistance to the membrane-targeting antibiotic daptomycin and demonstrates the conservation of NOS-derived NO•-haem receptor signalling between bacteria and mammals.


Subject(s)
Electron Transport , Nitric Oxide Synthase/metabolism , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism , Aerobiosis , Animals , Electron Transport Complex IV/metabolism , Mice, Inbred C57BL , Nasal Cavity/microbiology , Staphylococcus aureus/enzymology
13.
mBio ; 7(2): e02161-15, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27094335

ABSTRACT

UNLABELLED: The four-cysteine zinc finger motif of the bacterial RNA polymerase regulator DksA is essential for protein structure, canonical control of the stringent response to nutritional limitation, and thiol-based sensing of oxidative and nitrosative stress. This interdependent relationship has limited our understanding of DksA-mediated functions in bacterial pathogenesis. Here, we have addressed this challenge by complementing ΔdksA Salmonella with Pseudomonas aeruginosa dksA paralogues that encode proteins differing in cysteine and zinc content. We find that four-cysteine, zinc-bound (C4) and two-cysteine, zinc-free (C2) DksA proteins are able to mediate appropriate stringent control in Salmonella and that thiol-based sensing of reactive species is conserved among C2 and C4 orthologues. However, variations in cysteine and zinc content determine the threshold at which individual DksA proteins sense and respond to reactive species. In particular, zinc acts as an antioxidant, dampening cysteine reactivity and raising the threshold of posttranslational thiol modification with reactive species. Consequently, C2 DksA triggers transcriptional responses in Salmonella at levels of oxidative or nitrosative stress normally tolerated by Salmonella expressing C4 orthologues. Inappropriate transcriptional regulation by C2 DksA increases the susceptibility of Salmonella to the antimicrobial effects of hydrogen peroxide and nitric oxide, and attenuates virulence in macrophages and mice. Our findings suggest that the redox-active sensory function of DksA proteins is finely tuned to optimize bacterial fitness according to the levels of oxidative and nitrosative stress encountered by bacterial species in their natural and host environments. IMPORTANCE: In order to cause disease, pathogenic bacteria must rapidly sense and respond to antimicrobial pressures encountered within the host. Prominent among these stresses, and of particular relevance to intracellular pathogens such as Salmonella, are nutritional restriction and the enzymatic generation of reactive oxygen and nitrogen species. The conserved transcriptional regulator DksA controls adaptive responses to nutritional limitation, as well as to oxidative and nitrosative stress. Here, we demonstrate that each of these functions contributes to bacterial pathogenesis. Our observations highlight the importance of metabolic adaptation in bacterial pathogenesis and show the mechanism by which DksA orthologues are optimized to sense the levels of oxidative and nitrosative stress encountered in their natural habitats. An improved understanding of the conserved processes used by bacteria to sense, respond to, and limit host defense will inform the development of novel strategies to treat infections caused by pathogenic, potentially multidrug-resistant bacteria.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cysteine/metabolism , Oxidative Stress , Salmonella typhimurium/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Virulence Factors/chemistry , Virulence Factors/metabolism , Zinc/metabolism , Animals , Bacterial Proteins/genetics , Cysteine/chemistry , Cysteine/genetics , Gene Expression Regulation, Bacterial , Humans , Mice , Oxidation-Reduction , Salmonella Infections/microbiology , Salmonella typhimurium/chemistry , Salmonella typhimurium/genetics , Transcription Factors/genetics , Virulence Factors/genetics , Zinc/analysis
14.
mBio ; 7(2): e02265, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26933058

ABSTRACT

UNLABELLED: Nontyphoidal Salmonella enterica serovar Typhimurium is a frequent cause of bloodstream infections in children and HIV-infected adults in sub-Saharan Africa. Most isolates from African patients with bacteremia belong to a single sequence type, ST313, which is genetically distinct from gastroenteritis-associated ST19 strains, such as 14028s and SL1344. Some studies suggest that the rapid spread of ST313 across sub-Saharan Africa has been facilitated by anthroponotic (person-to-person) transmission, eliminating the need for Salmonella survival outside the host. While these studies have not ruled out zoonotic or other means of transmission, the anthroponotic hypothesis is supported by evidence of extensive genomic decay, a hallmark of host adaptation, in the sequenced ST313 strain D23580. We have identified and demonstrated 2 loss-of-function mutations in D23580, not present in the ST19 strain 14028s, that impair multicellular stress resistance associated with survival outside the host. These mutations result in inactivation of the KatE stationary-phase catalase that protects high-density bacterial communities from oxidative stress and the BcsG cellulose biosynthetic enzyme required for the RDAR (red, dry, and rough) colonial phenotype. However, we found that like 14028s, D23580 is able to elicit an acute inflammatory response and cause enteritis in mice and rhesus macaque monkeys. Collectively, these observations suggest that African S. Typhimurium ST313 strain D23580 is becoming adapted to an anthroponotic mode of transmission while retaining the ability to infect and cause enteritis in multiple host species. IMPORTANCE: The last 3 decades have witnessed an epidemic of invasive nontyphoidal Salmonella infections in sub-Saharan Africa. Genomic analysis and clinical observations suggest that the Salmonella strains responsible for these infections are evolving to become more typhoid-like with regard to patterns of transmission and virulence. This study shows that a prototypical African nontyphoidal Salmonella strain has lost traits required for environmental stress resistance, consistent with an adaptation to a human-to-human mode of transmission. However, in contrast to predictions, the strain remains capable of causing acute inflammation in the mammalian intestine. This suggests that the systemic clinical presentation of invasive nontyphoidal Salmonella infections in Africa reflects the immune status of infected hosts rather than intrinsic differences in the virulence of African Salmonella strains. Our study provides important new insights into the evolution of host adaptation in bacterial pathogens.


Subject(s)
Adaptation, Biological , Salmonella Infections/microbiology , Salmonella typhimurium/enzymology , Salmonella typhimurium/physiology , Stress, Physiological , Africa South of the Sahara/epidemiology , Animals , Catalase/genetics , Catalase/metabolism , Disease Models, Animal , Epidemics , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Humans , Macaca mulatta , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Salmonella Infections/epidemiology , Salmonella typhimurium/genetics , Salmonella typhimurium/isolation & purification
15.
Nat Commun ; 5: 5270, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25348042

ABSTRACT

Horizontal gene transfer plays a major role in bacterial evolution. Successful acquisition of new genes requires their incorporation into existing regulatory networks. This study compares the regulation of conserved genes in the PhoPQ regulon of Salmonella enterica serovar Typhimurium with that of PhoPQ-regulated horizontally acquired genes, which are silenced by the histone-like protein H-NS. We demonstrate that PhoP upregulates conserved and horizontally acquired genes by distinct mechanisms. Conserved genes are regulated by classical PhoP-mediated activation and are invariant in promoter architecture, whereas horizontally acquired genes exhibit variable promoter architecture and are regulated by PhoP-mediated counter-silencing. Biochemical analyses show that a horizontally acquired promoter adopts different structures in the silenced and counter-silenced states, implicating the remodelling of the H-NS nucleoprotein filament and the subsequent restoration of open-complex formation as the central mechanism of counter-silencing. Our results indicate that counter-silencing is favoured in the regulatory integration of newly acquired genes because it is able to accommodate multiple promoter architectures.


Subject(s)
Biological Evolution , Gene Regulatory Networks , Gene Silencing , Salmonella typhimurium/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA-Binding Proteins/metabolism , Deoxyribonuclease I/metabolism , Gene Expression Regulation, Bacterial , Promoter Regions, Genetic , Regulon/genetics , Ultraviolet Rays
16.
Cell Host Microbe ; 14(1): 26-37, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23870311

ABSTRACT

Host inflammation alters the availability of nutrients such as iron to limit microbial growth. However, Salmonella enterica serovar Typhimurium thrives in the inflamed gut by scavenging for iron with siderophores. By administering Escherichia coli strain Nissle 1917, which assimilates iron by similar mechanisms, we show that this nonpathogenic bacterium can outcompete and reduce S. Typhimurium colonization in mouse models of acute colitis and chronic persistent infection. This probiotic activity depends on E. coli Nissle iron acquisition, given that mutants deficient in iron uptake colonize the intestine but do not reduce S. Typhimurium colonization. Additionally, the ability of E. coli Nissle to overcome iron restriction by the host protein lipocalin 2, which counteracts some siderophores, is essential, given that S. Typhimurium is unaffected by E. coli Nissle in lipocalin 2-deficient mice. Thus, iron availability impacts S. Typhimurium growth, and E. coli Nissle reduces S. Typhimurium intestinal colonization by competing for this limiting nutrient.


Subject(s)
Colitis/drug therapy , Escherichia coli/metabolism , Intestines/microbiology , Iron/metabolism , Probiotics/therapeutic use , Salmonella Infections/microbiology , Salmonella typhimurium/growth & development , Salmonella typhimurium/metabolism , Animals , Colitis/metabolism , Colitis/microbiology , Female , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Probiotics/metabolism , Salmonella Infections/drug therapy , Salmonella Infections/metabolism
17.
Proc Natl Acad Sci U S A ; 110(4): 1470-5, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23302685

ABSTRACT

Gram-negative bacteria have an outer membrane containing LPS. LPS is constituted of an oligosaccharide portion and a lipid-A moiety that embeds this molecule within the outer membrane. LPS is a pathogen-associated molecular pattern, and several pathogens modify their lipid-A as a stealth strategy to avoid recognition by the innate immune system and gain resistance to host factors that disrupt the bacterial cell envelope. An essential feature of Salmonella enterica Typhimurium pathogenesis is its ability to replicate within vacuoles in professional macrophages. S. Typhimurium modifies its lipid-A by hydroxylation by the Fe2+/α-ketoglutarate-dependent dioxygenase enzyme (LpxO). Here, we show that a periplasmic protein of the bacterial oligonucleotide/oligosaccharide-binding fold family, herein named virulence and stress-related periplasmic protein (VisP), on binding to the sugar moiety of peptidoglycan interacts with LpxO. This interaction inhibits LpxO function, leading to decreased LpxO-dependent lipid-A modifications and increasing resistance to stressors within the vacuole environment during intramacrophage replication promoting systemic disease. Consequently, ΔvisP is avirulent in systemic murine infections, where VisP acts through LpxO. Several Gram-negative pathogens harbor both VisP and LpxO, suggesting that this VisP-LpxO mechanism of lipid-A modifications has broader implications in bacterial pathogenesis. Bacterial species devoid of LpxO (e.g., Escherichia coli) have no lipid-A phenotypes associated with the lack of VisP; however, VisP also controls LpxO-independent phenotypes. VisP and LpxO act independently in the S. Typhimurium murine colitis model, with both mutants being attenuated for diverging reasons; ΔvisP is less resistant to cationic antimicrobial peptides, whereas ΔlpxO is deficient for epithelial cell invasion. VisP converges bacterial cell wall homeostasis, stress responses, and pathogenicity.


Subject(s)
Bacterial Proteins/physiology , Host-Pathogen Interactions/physiology , Periplasmic Proteins/physiology , Salmonella typhimurium/pathogenicity , Virulence Factors/physiology , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cell Line , Female , Genes, Bacterial , HeLa Cells , Host-Pathogen Interactions/genetics , Humans , Lipid A/chemistry , Lipid A/metabolism , Macrophages/microbiology , Macrophages/physiology , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Periplasmic Proteins/chemistry , Periplasmic Proteins/genetics , Regulon , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/physiopathology , Salmonella typhimurium/genetics , Salmonella typhimurium/physiology , Sequence Homology, Amino Acid , Virulence/genetics , Virulence/physiology , Virulence Factors/chemistry , Virulence Factors/genetics
18.
PLoS Pathog ; 8(6): e1002733, 2012.
Article in English | MEDLINE | ID: mdl-22685400

ABSTRACT

Whereas the majority of pathogenic Salmonella serovars are capable of infecting many different animal species, typically producing a self-limited gastroenteritis, serovars with narrow host-specificity exhibit increased virulence and their infections frequently result in fatal systemic diseases. In our study, a genetic and functional analysis of the mannose-specific type 1 fimbrial adhesin FimH from a variety of serovars of Salmonella enterica revealed that specific mutant variants of FimH are common in host-adapted (systemically invasive) serovars. We have found that while the low-binding shear-dependent phenotype of the adhesin is preserved in broad host-range (usually systemically non-invasive) Salmonella, the majority of host-adapted serovars express FimH variants with one of two alternative phenotypes: a significantly increased binding to mannose (as in S. Typhi, S. Paratyphi C, S. Dublin and some isolates of S. Choleraesuis), or complete loss of the mannose-binding activity (as in S. Paratyphi B, S. Choleraesuis and S. Gallinarum). The functional diversification of FimH in host-adapted Salmonella results from recently acquired structural mutations. Many of the mutations are of a convergent nature indicative of strong positive selection. The high-binding phenotype of FimH that leads to increased bacterial adhesiveness to and invasiveness of epithelial cells and macrophages usually precedes acquisition of the non-binding phenotype. Collectively these observations suggest that activation or inactivation of mannose-specific adhesive properties in different systemically invasive serovars of Salmonella reflects their dynamic trajectories of adaptation to a life style in specific hosts. In conclusion, our study demonstrates that point mutations are the target of positive selection and, in addition to horizontal gene transfer and genome degradation events, can contribute to the differential pathoadaptive evolution of Salmonella.


Subject(s)
Adhesins, Bacterial/genetics , Phylogeny , Point Mutation , Salmonella Infections/genetics , Salmonella enterica/genetics , Salmonella enterica/pathogenicity , Amino Acid Sequence , Animals , Base Sequence , Biological Evolution , Gene Knockout Techniques , Humans , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutagenesis, Site-Directed , Virulence/genetics
19.
J Leukoc Biol ; 92(2): 353-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22706314

ABSTRACT

The expression of the cation transporter Nramp1 (Slc11a1) in late phagolysosomes confers resistance to infection with several intracellular pathogens, such as Salmonella enterica, in mice. The antimicrobial actions of Nramp1 are attributable, in part, to modulation of macrophage immune function and cellular iron metabolism--the latter affecting the availability of the essential nutrient iron for intraphagosomal bacteria. Here, we provide novel evidence that Nramp1 functionality increases the expression of the peptide Lcn2, which exerts its antimicrobial activity by scavenging iron-loaded bacterial siderophores and mediating iron efflux from macrophages. With the use of macrophage cell lines expressing functional or nonfunctional Nramp1, we found significantly elevated Lcn2 mRNA and protein levels in Nramp1-expressing cells. These resulted from Nramp1-mediated alterations in the production of ROS, which stimulated NF-κ B activity and subsequently, Lcn2 transcription. We observed that increased Lcn2 levels in primary Nramp1-positive macrophages resulted in a significant suppression of S. enterica serovar typhimurium growth. Stimulation of Lcn2 expression is a novel mechanism by which Nramp1 confers resistance against infection with the intracellular bacterium S. typhimurium.


Subject(s)
Acute-Phase Proteins/immunology , Cation Transport Proteins/immunology , Lipocalins/immunology , Macrophages/immunology , Macrophages/microbiology , Oncogene Proteins/immunology , Salmonella Infections, Animal/immunology , Salmonella typhimurium/immunology , Acute-Phase Proteins/genetics , Acute-Phase Proteins/metabolism , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line , Deferoxamine/pharmacology , Hydrogen Peroxide/metabolism , Immunity, Innate/immunology , Iron/metabolism , Lipocalin-2 , Lipocalins/genetics , Lipocalins/metabolism , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-kappa B/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Reactive Nitrogen Species/metabolism , Salmonella typhimurium/growth & development , Siderophores/pharmacology
20.
Cell Host Microbe ; 10(1): 33-43, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21767810

ABSTRACT

Host nitric oxide (NO⋅) production is important for controlling intracellular bacterial pathogens, including Salmonella enterica serovar Typhimurium, but the underlying mechanisms are incompletely understood. S. Typhmurium 14028s is prototrophic for all amino acids but cannot synthesize methionine (M) or lysine (K) during nitrosative stress. Here, we show that NO⋅-induced MK auxotrophy results from reduced succinyl-CoA availability as a consequence of NO⋅ targeting of lipoamide-dependent lipoamide dehydrogenase (LpdA) activity. LpdA is an essential component of the pyruvate and α-ketoglutarate dehydrogenase complexes. Additional effects of NO⋅ on gene regulation prevent compensatory pathways of succinyl-CoA production. Microarray analysis indicates that over 50% of the transcriptional response of S. Typhimurium to nitrosative stress is attributable to LpdA inhibition. Bacterial methionine transport is essential for virulence in NO⋅-producing mice, demonstrating that NO⋅-induced MK auxotrophy occurs in vivo. These observations underscore the importance of metabolic targets for antimicrobial actions of NO⋅.


Subject(s)
Citric Acid Cycle , Nitric Oxide/metabolism , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Acyl Coenzyme A/metabolism , Animals , Biological Transport , Culture Media , Dihydrolipoamide Dehydrogenase/metabolism , Female , Gene Expression Regulation, Bacterial , Host-Pathogen Interactions , Ketoglutarate Dehydrogenase Complex/metabolism , Lysine/metabolism , Lysine/pharmacology , Methionine/metabolism , Methionine/pharmacology , Mice , Mice, Inbred C3H , Nitric Oxide/pharmacology , Salmonella Infections/metabolism , Salmonella typhimurium/drug effects , Stress, Physiological , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...