Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Access Microbiol ; 6(2)2024.
Article in English | MEDLINE | ID: mdl-38482354

ABSTRACT

In supermarkets and chemists worldwide, consumers are faced with an array of antimicrobial domestic cleaning and personal hygiene products purporting to kill germs and keep people safe. Many of these proven active ingredients (biocides) encourage the development of antimicrobial resistance (AMR) in microbes and microbial populations, in turn increasing the likelihood of AMR infections. In order to understand and address the selective pressure towards AMR posed by the unrestricted use of biocides, it is necessary to understand which biocides are most frequently found in consumer products and the current regulatory framework that governs their use. In this research we survey the biocidal active ingredients in the major categories of cleaning and personal care products available from supermarkets and pharmacies in Australia, and comment on the regulations that dictate how these products are tested and marketed. Benzalkonium chloride and ethanol were the two most prevalent antimicrobial biocides in this study, while triclosan, which is banned in several jurisdictions, was found in a small number of products. In Australia, many antimicrobial consumer products are regulated for efficacy and safety under the Therapeutic Goods Act, but the potential to drive microbial adaptation and AMR is not considered. Overall this survey underscores the broad use and light regulation of antimicrobial biocides in products available to the general public in Australia, and provides an information resource to inform further research and stewardship efforts.

2.
Small ; 20(6): e2305052, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37798622

ABSTRACT

The rapid increase and spread of Gram-negative bacteria resistant to many or all existing treatments threaten a return to the preantibiotic era. The presence of bacterial polysaccharides that impede the penetration of many antimicrobials and protect them from the innate immune system contributes to resistance and pathogenicity. No currently approved antibiotics target the polysaccharide regions of microbes. Here, describe monolaurin-based niosomes, the first lipid nanoparticles that can eliminate bacterial polysaccharides from hypervirulent Klebsiella pneumoniae, are described. Their combination with polymyxin B shows no cytotoxicity in vitro and is highly effective in combating K. pneumoniae infection in vivo. Comprehensive mechanistic studies have revealed that antimicrobial activity proceeds via a multimodal mechanism. Initially, lipid nanoparticles disrupt polysaccharides, then outer and inner membranes are destabilized and destroyed by polymyxin B, resulting in synergistic cell lysis. This novel lipidic nanoparticle system shows tremendous promise as a highly effective antimicrobial treatment targeting multidrug-resistant Gram-negative pathogens.


Subject(s)
Nanoparticles , Polymyxin B , Polymyxin B/pharmacology , Liposomes/pharmacology , Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria , Klebsiella pneumoniae , Polysaccharides, Bacterial/pharmacology , Microbial Sensitivity Tests , Drug Resistance, Multiple, Bacterial
3.
Nat Microbiol ; 8(11): 1995-2005, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37814070

ABSTRACT

Concerns exist that widespread use of antiseptic or disinfectant biocides could contribute to the emergence and spread of multidrug-resistant bacteria. To investigate this, we performed transposon-directed insertion-site sequencing (TraDIS) on the multidrug-resistant pathogen, Acinetobacter baumannii, exposed to a panel of ten structurally diverse and clinically relevant biocides. Multiple gene targets encoding cell envelope or cytoplasmic proteins involved in processes including fatty acid biogenesis, multidrug efflux, the tricarboxylic acid cycle, cell respiration and cell division, were identified to have effects on bacterial fitness upon biocide exposure, suggesting that these compounds may have intracellular targets in addition to their known effects on the cell envelope. As cell respiration genes are required for A. baumannii fitness in biocides, we confirmed that sub-inhibitory concentrations of the biocides that dissipate membrane potential can promote A. baumannii tolerance to antibiotics that act intracellularly. Our results support the concern that residual biocides might promote antibiotic resistance in pathogenic bacteria.


Subject(s)
Acinetobacter baumannii , Disinfectants , Anti-Bacterial Agents/pharmacology , Disinfectants/pharmacology , Drug Resistance, Bacterial , Bacteria
4.
Cell Rep ; 42(6): 112551, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37224021

ABSTRACT

To kill bacteria, bacteriophages (phages) must first bind to a receptor, triggering the release of the phage DNA into the bacterial cell. Many bacteria secrete polysaccharides that had been thought to shield bacterial cells from phage attack. We use a comprehensive genetic screen to distinguish that the capsule is not a shield but is instead a primary receptor enabling phage predation. Screening of a transposon library to select phage-resistant Klebsiella shows that the first receptor-binding event docks to saccharide epitopes in the capsule. We discover a second step of receptor binding, dictated by specific epitopes in an outer membrane protein. This additional and necessary event precedes phage DNA release to establish a productive infection. That such discrete epitopes dictate two essential binding events for phages has profound implications for understanding the evolution of phage resistance and what dictates host range, two issues critically important to translating knowledge of phage biology into phage therapies.


Subject(s)
Bacteriophages , Klebsiella pneumoniae , Klebsiella pneumoniae/genetics , Bacteriophages/genetics , Porins/genetics , Porins/metabolism , Polysaccharides
5.
BMC Microbiol ; 23(1): 137, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37202723

ABSTRACT

BACKGROUND: Acinetobacter baumannii is an opportunistic human pathogen that causes a variety of infections in immunosuppressed individuals and patients in intensive care units. The success of this pathogen in nosocomial settings can be directly attributed to its persistent nature and its ability to rapidly acquire multidrug resistance. It is now considered to be one of the top priority pathogens for development of novel therapeutic approaches. Several high-throughput techniques have been utilised to identify the genetic determinants contributing to the success of A. baumannii as a global pathogen. However, targeted gene-function studies remain challenging due to the lack of appropriate genetic tools. RESULTS: Here, we have constructed a series of all-synthetic allelic exchange vectors - pALFI1, pALFI2 and pALFI3 - with suitable selection markers for targeted genetic studies in highly drug resistant A. baumannii isolates. The vectors follow the Standard European Vector Architecture (SEVA) framework for easy replacement of components. This method allows for rapid plasmid construction with the mutant allele, efficient conjugational transfer using a diaminopimelic acid-dependent Escherichia coli donor strain, efficient positive selection using the suitable selection markers and finally, sucrose-dependent counter-selection to obtain double-crossovers. CONCLUSIONS: We have used this method to create scar-less deletion mutants in three different strains of A. baumannii, which resulted in up to 75% deletion frequency of the targeted gene. We believe this method can be effectively used to perform genetic manipulation studies in multidrug resistant Gram-negative bacterial strains.


Subject(s)
Acinetobacter baumannii , Anti-Bacterial Agents , Humans , Anti-Bacterial Agents/pharmacology , Alleles , Plasmids/genetics , Drug Resistance, Multiple, Bacterial/genetics , Mutagenesis , Microbial Sensitivity Tests
6.
Nucleic Acids Res ; 51(12): 6101-6119, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37158230

ABSTRACT

Coordination of bacterial stress response mechanisms is critical for long-term survival in harsh environments for successful host infection. The general and specific stress responses of well-studied Gram-negative pathogens like Escherichia coli are controlled by alternative sigma factors, archetypically RpoS. The deadly hospital pathogen Acinetobacter baumannii is notoriously resistant to environmental stresses, yet it lacks RpoS, and the molecular mechanisms driving this incredible stress tolerance remain poorly defined. Here, using functional genomics, we identified the transcriptional regulator DksA as a master regulator for broad stress protection and virulence in A. baumannii. Transcriptomics, phenomics and in vivo animal studies revealed that DksA controls ribosomal protein expression, metabolism, mutation rates, desiccation, antibiotic resistance, and host colonization in a niche-specific manner. Phylogenetically, DksA was highly conserved and well-distributed across Gammaproteobacteria, with 96.6% containing DksA, spanning 88 families. This study lays the groundwork for understanding DksA as a major regulator of general stress response and virulence in this important pathogen.


Subject(s)
Acinetobacter baumannii , Escherichia coli Proteins , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Acinetobacter baumannii/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Sigma Factor/genetics , Sigma Factor/metabolism , Gene Expression Regulation, Bacterial
7.
Nat Commun ; 14(1): 702, 2023 02 09.
Article in English | MEDLINE | ID: mdl-36759602

ABSTRACT

Acinetobacter baumannii and Klebsiella pneumoniae are opportunistic pathogens frequently co-isolated from polymicrobial infections. The infections where these pathogens co-exist can be more severe and recalcitrant to therapy than infections caused by either species alone, however there is a lack of knowledge on their potential synergistic interactions. In this study we characterise the genomes of A. baumannii and K. pneumoniae strains co-isolated from a single human lung infection. We examine various aspects of their interactions through transcriptomic, phenomic and phenotypic assays that form a basis for understanding their effects on antimicrobial resistance and virulence during co-infection. Using co-culturing and analyses of secreted metabolites, we discover the ability of K. pneumoniae to cross-feed A. baumannii by-products of sugar fermentation. Minimum inhibitory concentration testing of mono- and co-cultures reveals the ability for A. baumannii to cross-protect K. pneumoniae against the cephalosporin, cefotaxime. Our study demonstrates distinct syntrophic interactions occur between A. baumannii and K. pneumoniae, helping to elucidate the basis for their co-existence in polymicrobial infections.


Subject(s)
Acinetobacter baumannii , Coinfection , Humans , Anti-Bacterial Agents/pharmacology , Acinetobacter baumannii/genetics , Klebsiella pneumoniae/genetics , Cephalosporins , Microbial Sensitivity Tests
8.
Nat Commun ; 13(1): 1065, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35246533

ABSTRACT

Naturally competent bacteria encode sophisticated protein machinery for the uptake and translocation of exogenous DNA into the cell. If this DNA is integrated into the bacterial genome, the bacterium is said to be naturally transformed. Most competent bacterial species utilise type IV pili for the initial DNA uptake step. These proteinaceous cell-surface structures are composed of thousands of pilus subunits (pilins), designated as major or minor according to their relative abundance in the pilus. Here, we show that the minor pilin FimT plays an important role in the natural transformation of Legionella pneumophila. We use NMR spectroscopy, in vitro DNA binding assays and in vivo transformation assays to understand the molecular basis of FimT's role in this process. FimT binds to DNA via an electropositive patch, rich in arginines, several of which are well-conserved and located in a conformationally flexible C-terminal tail. FimT orthologues from other Gammaproteobacteria share the ability to bind to DNA. Our results suggest that FimT plays an important role in DNA uptake in a wide range of competent species.


Subject(s)
Fimbriae Proteins , Legionella pneumophila , Bacterial Proteins/metabolism , DNA/metabolism , DNA, Bacterial/metabolism , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Legionella pneumophila/genetics , Legionella pneumophila/metabolism , Transformation, Bacterial
9.
Microb Genom ; 8(2)2022 02.
Article in English | MEDLINE | ID: mdl-35166651

ABSTRACT

Acinetobacter baumannii is a critically important pathogen known for its widespread antibiotic resistance and ability to persist in hospital-associated environments. Whilst the majority of A. baumannii infections are hospital-acquired, infections from outside the hospital have been reported with high mortality. Despite this, little is known about the natural environmental reservoir(s) of A. baumannii and the virulence potential underlying non-clinical strains. Here, we report the complete genome sequences of six diverse strains isolated from environments such as river, soil, and industrial sites around the world. Phylogenetic analyses showed that four of these strains were unrelated to representative nosocomial strains and do not share a monophyletic origin, whereas two had sequence types belonging to the global clone lineages GC1 and GC2. Further, the majority of these strains harboured genes linked to virulence and stress protection in nosocomial strains. These genotypic properties correlated well with in vitro virulence phenotypic assays testing resistance to abiotic stresses, serum survival, and capsule formation. Virulence potential was confirmed in vivo, with most environmental strains able to effectively kill Galleria mellonella greater wax moth larvae. Using phenomic arrays and antibiotic resistance profiling, environmental and nosocomial strains were shown to have similar substrate utilisation patterns although environmental strains were distinctly more sensitive to antibiotics. Taken together, these features of environmental A. baumannii strains suggest the existence of a strain-specific distinct gene pools for niche specific adaptation. Furthermore, environmental strains appear to be equally virulent as contemporary nosocomial strains but remain largely antibiotic sensitive.


Subject(s)
Acinetobacter baumannii/classification , Acinetobacter baumannii/genetics , Drug Resistance, Multiple, Bacterial/genetics , Genomics , Phylogeny , Virulence Factors/genetics , Acinetobacter Infections , Acinetobacter baumannii/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Biofilms , Cross Infection , Hospitals , Moths , Virulence/genetics , Whole Genome Sequencing
10.
EBioMedicine ; 73: 103653, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34717227

ABSTRACT

BACKGROUND: Biocide disinfectants are essential tools in infection control, but their use can inadvertently contribute to emergence of antibiotic-resistant bacteria. In this study we systematically examine the effect of the biocide benzalkonium chloride, which is primarily used for surface disinfection but is also present as a preservative in many consumer products, on the activity of aminoglycoside antibiotics in Acinetobacter baumannii. METHODS: The effect of subinhibitory BAC on aminoglycoside treatment of A. baumannii ATCC17978 was investigated using time-to-kill assays, MIC determination, directed evolution experiments, fluctuation tests and labelled gentamicin accumulation assays. Further MIC determinations and directed evolution experiments were performed with additional Gram-negative ESKAPE pathogens. FINDINGS: In A. baumannii ATCC17978, BAC prevents gentamicin killing and drastically increases the frequency at which resistant mutants emerge, through reducing intracellular antibiotic accumulation. BAC also increases the MIC of multiple aminoglycoside antibiotics (kanamycin, tobramycin, streptomycin, gentamicin and amikacin). BAC promotes the emergence of mutants with reduced gentamicin susceptibility in other Gram-negative ESKAPE pathogens but does not always alter the MIC. These effects occur at BAC concentrations which are similar to residual levels in high-use environments, and just below the concentration range for BAC when used as a preservative in eye drops and ear drops. INTERPRETATION: Our results suggest that subinhibitory BAC has the potential to antagonise aminoglycoside activity and promote the emergence of bacterial mutants with reduced susceptibility. We suggest that the extremely widespread use of BAC in clinical and home settings and its long half-life mean there is potential for these interactions to occur in the environment, or in patients who use BAC-containing products while taking aminoglycosides to treat skin, eye or ear infections, although such co-exposure is likely to be rare. We suggest that biocide stewardship is needed to prevent the types of exposure that can contribute to antibiotic resistance. FUNDING: This work was funded by the National Health and Medical Research Council of Australia. The funders had no role in study design, interpretation or decision to publish.


Subject(s)
Acinetobacter baumannii/drug effects , Aminoglycosides/pharmacology , Benzalkonium Compounds/adverse effects , Drug Resistance, Bacterial/drug effects , Acinetobacter baumannii/physiology , Anti-Bacterial Agents/pharmacology , Benzalkonium Compounds/pharmacology , Disinfectants/adverse effects , Disinfectants/pharmacology , Microbial Sensitivity Tests
11.
Commun Biol ; 4(1): 1114, 2021 09 22.
Article in English | MEDLINE | ID: mdl-34552198

ABSTRACT

Antimicrobial resistance genes, including multidrug efflux pumps, evolved long before the ubiquitous use of antimicrobials in medicine and infection control. Multidrug efflux pumps often transport metabolites, signals and host-derived molecules in addition to antibiotics or biocides. Understanding their ancestral physiological roles could inform the development of strategies to subvert their activity. In this study, we investigated the response of Acinetobacter baumannii to polyamines, a widespread, abundant class of amino acid-derived metabolites, which led us to identify long-chain polyamines as natural substrates of the disinfectant efflux pump AmvA. Loss of amvA dramatically reduced tolerance to long-chain polyamines, and these molecules induce expression of amvA through binding to its cognate regulator AmvR. A second clinically-important efflux pump, AdeABC, also contributed to polyamine tolerance. Our results suggest that the disinfectant resistance capability that allows A. baumannii to survive in hospitals may have evolutionary origins in the transport of polyamine metabolites.


Subject(s)
Acinetobacter baumannii/genetics , Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial/genetics , Spermidine/metabolism , Acinetobacter baumannii/metabolism , Bacterial Proteins/metabolism , Disinfectants/pharmacology , Spermine/metabolism
12.
Microbiol Spectr ; 9(1): e0102321, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34431721

ABSTRACT

The production of capsular polysaccharides by Klebsiella pneumoniae protects the bacterial cell from harmful environmental factors such as antimicrobial compounds and infection by bacteriophages (phages). To bypass this protective barrier, some phages encode polysaccharide-degrading enzymes referred to as depolymerases to provide access to cell surface receptors. Here, we characterized the phage RAD2, which infects K. pneumoniae strains that produce the widespread, hypervirulence-associated K2-type capsular polysaccharide. Using transposon-directed insertion sequencing, we have shown that the production of capsule is an absolute requirement for efficient RAD2 infection by serving as a first-stage receptor. We have identified the depolymerase responsible for recognition and degradation of the capsule, determined that the depolymerase forms globular appendages on the phage virion tail tip, and present the cryo-electron microscopy structure of the RAD2 capsule depolymerase at 2.7-Å resolution. A putative active site for the enzyme was identified, comprising clustered negatively charged residues that could facilitate the hydrolysis of target polysaccharides. Enzymatic assays coupled with mass spectrometric analyses of digested oligosaccharide products provided further mechanistic insight into the hydrolase activity of the enzyme, which, when incubated with K. pneumoniae, removes the capsule and sensitizes the cells to serum-induced killing. Overall, these findings expand our understanding of how phages target the Klebsiella capsule for infection, providing a framework for the use of depolymerases as antivirulence agents against this medically important pathogen. IMPORTANCE Klebsiella pneumoniae is a medically important pathogen that produces a thick protective capsule that is essential for pathogenicity. Phages are natural predators of bacteria, and many encode diverse "capsule depolymerases" which specifically degrade the capsule of their hosts, an exploitable trait for potential therapies. We have determined the first structure of a depolymerase that targets the clinically relevant K2 capsule and have identified its putative active site, providing hints to its mechanism of action. We also show that Klebsiella cells treated with a recombinant form of the depolymerase are stripped of capsule, inhibiting their ability to grow in the presence of serum, demonstrating the anti-infective potential of these robust and readily producible enzymes against encapsulated bacterial pathogens such as K. pneumoniae.


Subject(s)
Bacterial Capsules/virology , Bacteriophages/enzymology , Klebsiella pneumoniae/virology , Polysaccharide-Lyases/metabolism , Viral Proteins/metabolism , Bacterial Capsules/metabolism , Bacterial Capsules/ultrastructure , Bacteriophages/genetics , Bacteriophages/physiology , Cryoelectron Microscopy , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/metabolism , Klebsiella pneumoniae/ultrastructure , Polysaccharide-Lyases/genetics , Viral Proteins/genetics
13.
Pathog Dis ; 79(2)2021 02 19.
Article in English | MEDLINE | ID: mdl-33476383

ABSTRACT

Galleria mellonella has risen to fame as an invertebrate model organism given its ethical advantages, low maintenance costs, rapid reproduction time, short life cycle, high number of progeny, tolerance for human body temperatures, innate immune system and similarities to mammalian host models. It is increasingly being utilised to evaluate in vivo toxicity and efficacy of chemical compounds and antimicrobials, modelling microbial (bacterial, fungal and viral) pathogenicity and assessing host-pathogen interaction during infection. During this molecular age of genomic, transcriptomic, proteomic and genetic manipulation approaches, our understanding of microbial pathogenicity and host-pathogen interactions has deepened from high-throughput molecular studies performed in G. mellonella. In this review, we describe the use of G. mellonella in a broad range of studies involving omics, drug resistance, functional analysis and host-microbial community relationships. The future of G. mellonella in the molecular age is bright, with a multitude of new approaches and uses for this model from clinical to biotechnological on the horizon.


Subject(s)
Anti-Infective Agents/pharmacology , Genomics , Host-Pathogen Interactions , Larva/microbiology , Microbiota , Moths/microbiology , Proteomics , Animals , Disease Models, Animal , Humans
14.
Pathog Dis ; 79(3)2021 03 20.
Article in English | MEDLINE | ID: mdl-33512418

ABSTRACT

Infections caused by Klebsiella pneumoniae are a major public health threat. Extensively drug-resistant and even pan-resistant strains have been reported. Understanding K. pneumoniae pathogenesis is hampered by the fact that murine models of infection offer limited resolution for non-hypervirulent strains which cause the majority of infections. The insect Galleria mellonella larva is a widely used alternative model organism for bacterial pathogens. We have performed genome-scale fitness profiling of a multidrug-resistant K. pneumoniae ST258 strain during infection of G. mellonella, to determine if this model is suitable for large-scale virulence factor discovery in this pathogen. Our results demonstrated a dominant role for surface polysaccharides in infection, with contributions from siderophores, cell envelope proteins, purine biosynthesis genes and additional genes of unknown function. Comparison with a hypervirulent strain, ATCC 43816, revealed substantial overlap in important infection-related genes, as well as additional putative virulence factors specific to ST258, reflecting strain-dependent fitness effects. Our analysis also identified a role for the metalloregulatory protein NfeR (YqjI) in virulence. Overall, this study offers new insight into the infection fitness landscape of K. pneumoniae, and provides a framework for using the highly flexible and easily scalable G. mellonella infection model to dissect molecular virulence mechanisms of bacterial pathogens.


Subject(s)
Klebsiella Infections/microbiology , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/pathogenicity , Larva/microbiology , Moths/microbiology , Virulence Factors/genetics , Virulence , Animals , Bacterial Proteins/genetics , DNA, Bacterial , Disease Models, Animal , Drug Resistance, Multiple, Bacterial , Genetic Complementation Test , Genome, Bacterial , Humans , Klebsiella pneumoniae/drug effects , Membrane Proteins/genetics , Mutagenesis , Polysaccharides/genetics , Purines , Siderophores/genetics , Siderophores/metabolism
15.
Chemistry ; 27(6): 2021-2029, 2021 Jan 26.
Article in English | MEDLINE | ID: mdl-33231906

ABSTRACT

Resistance to currently available antifungal drugs has quietly been on the rise but overshadowed by the alarming spread of antibacterial resistance. There is a striking lack of attention to the threat of drug-resistant fungal infections, with only a handful of new drugs currently in development. Given that metal complexes have proven to be useful new chemotypes in the fight against diseases such as cancer, malaria, and bacterial infections, it is reasonable to explore their possible utility in treating fungal infections. Herein we report a series of cobalt(III) Schiff base complexes with broad-spectrum antifungal activity. Some of these complexes show minimum inhibitory concentrations (MIC) in the low micro- to nanomolar range against a series of Candida and Cryptococcus yeasts. Additionally, we demonstrate that these compounds show no cytotoxicity against both bacterial and human cells. Finally, we report the first in vivo toxicity data on these compounds in Galleria mellonella, showing that doses as high as 266 mg kg-1 are tolerated without adverse effects, paving the way for further in vivo studies of these complexes.


Subject(s)
Antifungal Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Candida , Cobalt , Coordination Complexes/toxicity , Humans , Microbial Sensitivity Tests , Schiff Bases
16.
Infect Immun ; 88(8)2020 07 21.
Article in English | MEDLINE | ID: mdl-32513855

ABSTRACT

The serum complement system is a first line of defense against bacterial invaders. Resistance to killing by serum enhances the capacity of Klebsiella pneumoniae to cause infection, but it is an incompletely understood virulence trait. Identifying and characterizing the factors responsible for preventing activation of, and killing by, serum complement could inform new approaches to treatment of K. pneumoniae infections. Here, we used functional genomic profiling to define the genetic basis of complement resistance in four diverse serum-resistant K. pneumoniae strains (NTUH-K2044, B5055, ATCC 43816, and RH201207), and explored their recognition by key complement components. More than 90 genes contributed to resistance in one or more strains, but only three, rfaH, lpp, and arnD, were common to all four strains. Deletion of the antiterminator rfaH, which controls the expression of capsule and O side chains, resulted in dramatic complement resistance reductions in all strains. The murein lipoprotein gene lpp promoted capsule retention through a mechanism dependent on its C-terminal lysine residue; its deletion led to modest reductions in complement resistance. Binding experiments with the complement components C3b and C5b-9 showed that the underlying mechanism of evasion varied in the four strains: B5055 and NTUH-K2044 appeared to bypass recognition by complement entirely, while ATCC 43816 and RH201207 were able to resist killing despite being associated with substantial levels of C5b-9. All rfaH and lpp mutants bound C3b and C5b-9 in large quantities. Our findings show that, even among this small selection of isolates, K. pneumoniae adopts differing mechanisms and utilizes distinct gene sets to avoid complement attack.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Carboxy-Lyases/immunology , Gene Expression Regulation, Bacterial/immunology , Genes, Bacterial , Immune Evasion , Klebsiella pneumoniae/immunology , Peptide Elongation Factors/immunology , Bacterial Outer Membrane Proteins/genetics , Blood Bactericidal Activity/immunology , Carboxy-Lyases/deficiency , Carboxy-Lyases/genetics , Complement C3b/genetics , Complement C3b/immunology , Complement Membrane Attack Complex/genetics , Complement Membrane Attack Complex/immunology , DNA Transposable Elements , Gene Expression Profiling , Gene Library , Humans , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/pathogenicity , Mutation , Peptide Elongation Factors/deficiency , Peptide Elongation Factors/genetics , Sequence Analysis, DNA
17.
J Vis Exp ; (143)2019 01 07.
Article in English | MEDLINE | ID: mdl-30663644

ABSTRACT

Capsule is a key virulence factor in many bacterial species, mediating immune evasion and resistance to various physical stresses. While many methods are available to quantify and compare capsule production between different strains or mutants, there is no widely used method for sorting bacteria based on how much capsule they produce. We have developed a method to separate bacteria by capsule amount, using a discontinuous density gradient. This method is used to compare capsule amounts semi-quantitatively between cultures, to isolate mutants with altered capsule production, and to purify capsulated bacteria from complex samples. This method can also be coupled with transposon-insertion sequencing to identify genes involved in capsule regulation. Here, the method is demonstrated in detail, including how to optimize the gradient conditions for a new bacterial species or strain, and how to construct and run the density gradient.


Subject(s)
Bacterial Capsules/immunology , Virulence Factors/genetics , Animals
18.
mBio ; 9(6)2018 11 20.
Article in English | MEDLINE | ID: mdl-30459193

ABSTRACT

Klebsiella pneumoniae infections affect infants and the immunocompromised, and the recent emergence of hypervirulent and multidrug-resistant K. pneumoniae lineages is a critical health care concern. Hypervirulence in K. pneumoniae is mediated by several factors, including the overproduction of extracellular capsule. However, the full details of how K. pneumoniae capsule biosynthesis is achieved or regulated are not known. We have developed a robust and sensitive procedure to identify genes influencing capsule production, density-TraDISort, which combines density gradient centrifugation with transposon insertion sequencing. We have used this method to explore capsule regulation in two clinically relevant Klebsiella strains, K. pneumoniae NTUH-K2044 (capsule type K1) and K. pneumoniae ATCC 43816 (capsule type K2). We identified multiple genes required for full capsule production in K. pneumoniae, as well as putative suppressors of capsule in NTUH-K2044, and have validated the results of our screen with targeted knockout mutants. Further investigation of several of the K. pneumoniae capsule regulators identified-ArgR, MprA/KvrB, SlyA/KvrA, and the Sap ABC transporter-revealed effects on capsule amount and architecture, serum resistance, and virulence. We show that capsule production in K. pneumoniae is at the center of a complex regulatory network involving multiple global regulators and environmental cues and that the majority of capsule regulatory genes are located in the core genome. Overall, our findings expand our understanding of how capsule is regulated in this medically important pathogen and provide a technology that can be easily implemented to study capsule regulation in other bacterial species.IMPORTANCE Capsule production is essential for K. pneumoniae to cause infections, but its regulation and mechanism of synthesis are not fully understood in this organism. We have developed and applied a new method for genome-wide identification of capsule regulators. Using this method, many genes that positively or negatively affect capsule production in K. pneumoniae were identified, and we use these data to propose an integrated model for capsule regulation in this species. Several of the genes and biological processes identified have not previously been linked to capsule synthesis. We also show that the methods presented here can be applied to other species of capsulated bacteria, providing the opportunity to explore and compare capsule regulatory networks in other bacterial strains and species.


Subject(s)
Bacterial Capsules/genetics , Centrifugation, Density Gradient/methods , Gene Expression Regulation, Bacterial , Genome, Bacterial , Klebsiella pneumoniae/genetics , Sequence Analysis, DNA/methods , Animals , DNA Transposable Elements , Gene Knockout Techniques , Klebsiella Infections/microbiology , Klebsiella pneumoniae/pathogenicity , Larva/microbiology , Moths/microbiology , Mutagenesis, Insertional , Mutation , Virulence Factors/genetics
19.
Sci Rep ; 8(1): 1013, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29343718

ABSTRACT

Bacteria have evolved numerous defense systems to protect themselves from viral (bacteriophage) infection. The ToxIN system of Pectobacterium atrosepticum is a Type III toxin-antitoxin complex and "altruistic suicide" anti-phage system, which kills phage-infected cells through the release of a ribonuclease toxin, ToxN. ToxN is counteracted by a co-transcribed antitoxic RNA pseudoknot, ToxI, which self-assembles with ToxN into an inactive 3 ToxI:3 ToxN complex in vitro. However it is not known whether this complex is predominant in vivo, or how the complex is disassembled following infection to trigger a lethal, "altruistic" response. In this study, we characterise ToxI turnover and folding, and explore the link between complex stability and anti-phage activity, with a view to understanding events that lead to ToxN-mediated suicide following phage infection. We present evidence that ToxN constantly cleaves fresh ToxI in vivo rather than staying associated with pre-processed antitoxin, and that the ToxI antitoxin can partially fold spontaneously using conserved nucleotides. We also show that reducing the stability of the ToxIN complex can increase the strength of the antiviral response in a phage-dependent manner. Based on this information, we propose a revised model for ToxN inhibition, complex assembly and activation by infecting bacteriophage.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Pectobacterium/genetics , RNA, Bacterial/genetics , Ribonucleases/genetics , Toxin-Antitoxin Systems/genetics , Antitoxins , Bacterial Proteins/metabolism , Bacteriophages/genetics , Bacteriophages/growth & development , Base Pairing , Nucleic Acid Conformation , Pectobacterium/metabolism , Pectobacterium/virology , Protein Binding , Protein Folding , Protein Stability , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , Ribonucleases/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...