Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
1.
Nat Commun ; 15(1): 5171, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886409

ABSTRACT

Efficient utilization of nutrients is crucial for microbial survival and virulence. The same nutrient may be utilized by multiple catabolic pathways, indicating that the physical and chemical environments for induction as well as their functional roles may differ. Here, we study the tagatose and Leloir pathways for galactose catabolism of the human pathogen Streptococcus pneumoniae. We show that galactose utilization potentiates pneumococcal virulence, the induction of galactose catabolic pathways is influenced differentially by the concentration of galactose and temperature, and sialic acid downregulates galactose catabolism. Furthermore, the genetic regulation and in vivo induction of each pathway differ, and both galactose catabolic pathways can be turned off with a galactose analogue in a substrate-specific manner, indicating that galactose catabolic pathways can be potential drug targets.


Subject(s)
Galactose , Gene Expression Regulation, Bacterial , Streptococcus pneumoniae , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Galactose/metabolism , Virulence/genetics , Animals , Hexoses/metabolism , Mice , Metabolic Networks and Pathways/genetics , Humans , Pneumococcal Infections/microbiology , Pneumococcal Infections/metabolism , N-Acetylneuraminic Acid/metabolism , Temperature , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Female
2.
Microbiology (Reading) ; 169(2)2023 02.
Article in English | MEDLINE | ID: mdl-36811449

ABSTRACT

The in vivo temperature can vary according to the host tissue and the response to infection. Streptococcus pneumoniae has evolved mechanisms to survive these temperature differences, but neither the consequences of different temperatures for pneumococcal phenotype nor the genetic basis of thermal adaptation are known in detail. In our previous study [16], we found that CiaR, which is a part of two-component regulatory system CiaRH, as well as 17 genes known to be controlled by CiaRH, were identified to be differentially expressed with temperature. One of the CiaRH-regulated genes shown to be differentially regulated by temperature is for the high-temperature requirement protein (HtrA), coded by SPD_2068 (htrA). In this study, we hypothesized that the CiaRH system plays an important role in pneumococcal thermal adaptation through its control over htrA. This hypothesis was evaluated by testing strains mutated or overexpressing ciaR and/or htrA, in in vitro and in vivo assays. The results showed that in the absence of ciaR, the growth, haemolytic activity, amount of capsule and biofilm formation were considerably diminished at 40 °C only, while the cell size and virulence were affected at both 34 and 40 °C. The overexpression of htrA in the ∆ciaR background reconstituted the growth at all temperatures, and the haemolytic activity, biofilm formation and virulence of ∆ciaR partially at 40 °C. We also showed that overexpression of htrA in the wild-type promoted pneumococcal virulence at 40 °C, while the increase of capsule was observed at 34 °C, suggesting that the role of htrA changes at different temperatures. Our data suggest that CiaR and HtrA play an important role in pneumococcal thermal adaptation.


Subject(s)
Serine Proteases , Streptococcus pneumoniae , Streptococcus pneumoniae/genetics , Bacterial Proteins/genetics , Protein Kinases/genetics , Serine Endopeptidases/genetics
3.
J Infect ; 85(4): 405-411, 2022 10.
Article in English | MEDLINE | ID: mdl-35948110

ABSTRACT

OBJECTIVES: Knowledge of Acute Respiratory virus Infection (ARI) is limited in relation to their substantial global burden. We completed a feasibility study of a novel method to study the natural transmission of respiratory viruses from young children to adults in hospital. METHODS: Between September 2012 and May 2015, we recruited healthy adults (contacts) and paediatric inpatients with ARIs (index) presenting to the University Hospitals Leicester NHS Trust, Leicester, UK. We took nose and throat swabs from all participants prior to controlled, 30 minute interactions between the children with ARIs and adult contacts. Contacts recorded symptoms and provided four nose and throat swabs over ten days post-interaction, which were tested for a panel of respiratory viruses to assess transmission. RESULTS: 111 interactions occurred between children with ARIs and adult contacts. Respiratory viruses were detected in 103 of 111 children (93%), most commonly rhinoviruses (RVs) (67 of 103, 65%). Transmission to an adult contact occurred in 15 (14·6%) of 103 interactions and was inversely associated with the contact being male (adjusted OR 0·12; 95% CI 0·02-0·72). CONCLUSION: Using a novel methodology, we found that natural transmission of ARIs occurred in 15% of an infected child's contacts following a 30 minute interaction, primarily by RVs and when the contact was female. Our model has key advantages in comparison with human challenge studies making it well-suited for further studies of respiratory virus transmission, disease pathogenesis, and clinical and public health interventions to interrupt transmission.


Subject(s)
Respiratory Tract Infections , Virus Diseases , Viruses , Adult , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Male , Rhinovirus
4.
Environ Microbiol ; 24(9): 4449-4465, 2022 09.
Article in English | MEDLINE | ID: mdl-35642645

ABSTRACT

Exposure to particulate matter (PM), a major component of air pollution, is associated with exacerbation of chronic respiratory disease, and infectious diseases such as community-acquired pneumonia. Although PM can cause adverse health effects through direct damage to host cells, our previous study showed that PM can also impact bacterial behaviour by promoting in vivo colonization. In this study we describe the genetic mechanisms involved in the bacterial response to exposure to black carbon (BC), a constituent of PM found in most sources of air pollution. We show that Staphylococcus aureus strain USA300 LAC grown in BC prior to inoculation showed increased murine respiratory tract colonization and pulmonary invasion in vivo, as well as adhesion and invasion of human epithelial cells in vitro. Global transcriptional analysis showed that BC has a widespread effect on S. aureus transcriptional responses, altering the regulation of the major virulence gene regulators Sae and Agr and causing increased expression of genes encoding toxins, proteases and immune evasion factors. Together these data describe a previously unrecognized causative mechanism of air pollution-associated infection, in that exposure to BC can increase bacterial colonization and virulence factor expression by acting directly on the bacterium rather than via the host.


Subject(s)
Air Pollution , Staphylococcal Infections , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carbon/metabolism , Gene Expression Regulation, Bacterial , Humans , Mice , Particulate Matter/metabolism , Peptide Hydrolases/genetics , Respiratory System/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Virulence/genetics , Virulence Factors/metabolism
5.
Front Immunol ; 13: 907461, 2022.
Article in English | MEDLINE | ID: mdl-35720383

ABSTRACT

Circadian rhythms affect the progression and severity of bacterial infections including those caused by Streptococcus pneumoniae, but the mechanisms responsible for this phenomenon remain largely elusive. Following advances in our understanding of the role of replication of S. pneumoniae within splenic macrophages, we sought to investigate whether events within the spleen correlate with differential outcomes of invasive pneumococcal infection. Utilising murine invasive pneumococcal disease (IPD) models, here we report that infection during the murine active phase (zeitgeber time 15; 15h after start of light cycle, 3h after start of dark cycle) resulted in significantly faster onset of septicaemia compared to rest phase (zeitgeber time 3; 3h after start of light cycle) infection. This correlated with significantly higher pneumococcal burden within the spleen of active phase-infected mice at early time points compared to rest phase-infected mice. Whole-section confocal microscopy analysis of these spleens revealed that the number of pneumococci is significantly higher exclusively within marginal zone metallophilic macrophages (MMMs) known to allow intracellular pneumococcal replication as a prerequisite step to the onset of septicaemia. Pneumococcal clusters within MMMs were more abundant and increased in size over time in active phase-infected mice compared to those in rest phase-infected mice which decreased in size and were present in a lower percentage of MMMs. This phenomenon preceded significantly higher levels of bacteraemia alongside serum IL-6 and TNF-α concentrations in active phase-infected mice following re-seeding of pneumococci into the blood. These data greatly advance our fundamental knowledge of pneumococcal infection by linking susceptibility to invasive pneumococcal infection to variation in the propensity of MMMs to allow persistence and replication of phagocytosed bacteria. These findings also outline a somewhat rare scenario whereby the active phase of an organism's circadian cycle plays a seemingly counterproductive role in the control of invasive infection.


Subject(s)
Pneumococcal Infections , Sepsis , Animals , Macrophages/microbiology , Mice , Phagocytosis , Pneumococcal Infections/microbiology , Sepsis/microbiology , Streptococcus pneumoniae
6.
Microbiology (Reading) ; 168(4)2022 04.
Article in English | MEDLINE | ID: mdl-35404222

ABSTRACT

Bacteria have evolved mechanisms which enable them to control intracellular concentrations of metals. In the case of transition metals, such as copper, iron and zinc, bacteria must ensure enough is available as a cofactor for enzymes whilst at the same time preventing the accumulation of excess concentrations, which can be toxic. Interestingly, metal homeostasis and resistance systems have been found to play important roles in virulence. This review will discuss the copper homeostasis and resistance systems in Staphylococcus aureus and Listeria monocytogenes and the implications that acquisition of additional copper resistance genes may have in these pathogens.


Subject(s)
Listeria monocytogenes , Staphylococcal Infections , Copper , Humans , Listeria monocytogenes/genetics , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Virulence/genetics
7.
Microbiology (Reading) ; 168(12)2022 12.
Article in English | MEDLINE | ID: mdl-36748691

ABSTRACT

Streptococcus pneumoniae may inhabit the upper respiratory tract of humans without causing harm but it also causes diseases with high morbidity and mortality. It has excellent adaptive capabilities thanks to its ability to shuffle its genetic content by acquiring and incorporating DNA from other bacteria and is highly competent for genetic transformation. Sugar sensing, cleavage and transport ensure its fitness and survival in the host, and intracellular survival in macrophages has been linked to virulence. The polysaccharide capsule and toxin pneumolysin are the most important virulence determinants. Polysaccharide-based vaccines provide protection against the serotypes represented in vaccine formulations.


Subject(s)
Pneumococcal Infections , Streptococcus pneumoniae , Humans , Streptococcus pneumoniae/genetics , Pneumococcal Infections/microbiology , Financial Stress , Virulence Factors , Virulence
8.
Lancet Microbe ; 2(12): e695-e703, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34901898

ABSTRACT

BACKGROUND: Hypervirulent Klebsiella pneumoniae (hvKp) strains of capsule type K1 and K2 cause invasive infections associated with hepatic abscesses, which can be difficult to treat and are frequently associated with relapsing infections. Other K pneumoniae strains (non-hvKp), including lineages that have acquired carbapenem resistance, do not manifest this pathology. In this work we aimed to test the hypothesis that within-macrophage replication is a key mechanism underpinning abscess formation in hvKp infections. METHODS: In this exploratory investigation, to study the pathophysiology of abscess formation, mice were intravenously infected with 106 colony forming units (CFU) of either hvKp isolates (six strains) or non-hvKp isolates (seven strains). Intracellular bacterial replication and neutrophil influx in liver and spleen was quantified by fluorescence microscopy of sliced cryopreserved organs of mice collected 30 min, 6 h, and 24 h after infection with the aim to provide data of bacterial association to Kupffer cells in the liver and to the different tissue macrophages in the spleen. Microbiological and microscopy analysis of an ex-vivo model of pig liver and spleen infection were used to confirm within-macrophage replication. Pig organs were perfused with heparinised, autologous pig's blood and injected with 6·5 × 107 CFU of hvKp K2 sequence type 25 strain GMR151. Blood and tissue biopsies collected before infection and 30 min, 1 h, 2 h, 3 h, 4 h, and 5 h after infection were used to measure bacterial counts and to identify the subcellular localisation of bacteria by immunohistochemistry analysis. FINDINGS: We show that hvKp resisted phagocyte-mediated clearance and replicated in mouse liver macrophages to form clusters 6 h after infection, with a mean of 7·0 bacteria per Kupffer cell (SD 6·2); however, non-hvKp were efficiently cleared (mean 1·5 bacteria per cell [SD 1·1]). HvKp infection promoted neutrophil recruitment to sites of infection, which in the liver resulted in histopathological signs of abscess formation as early as 24 h post-infection. Experiments in pig organs which share a high functional and anatomical resemblance to human organs, provided strong evidence for the propensity of hvKp to replicate within the hepatic macrophages. INTERPRETATION: These findings show subversion of innate immune processes in the liver by K pneumoniae and resistance to Kupffer cell mediated clearance as an explanation for the propensity of hvKp strains to cause hepatic abscesses. FUNDING: University of Oxford and a Royal Society Wolfson grant funded biosafety facility.


Subject(s)
Klebsiella Infections , Liver Abscess , Animals , Klebsiella Infections/diagnosis , Klebsiella pneumoniae , Liver Abscess/microbiology , Macrophages , Mice , Perfusion , Swine , Virulence
9.
EBioMedicine ; 72: 103601, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34619637

ABSTRACT

BACKGROUND: Severe community-acquired pneumococcal pneumonia is commonly associated with bacteraemia. Although it is assumed that the bacteraemia solely derives from pneumococci entering the blood from the lungs it is unknown if other organs are important in the pathogenesis of bacteraemia. Using three models, we tested the relevance of the spleen in pneumonia-associated bacteraemia. METHODS: We used human spleens perfused ex vivo to explore permissiveness to bacterial replication, a non-human primate model to check for splenic involvement during pneumonia and a mouse pneumonia-bacteraemia model to demonstrate that splenic involvement correlates with invasive disease. FINDINGS: Here we present evidence that the spleen is the reservoir of bacteraemia during pneumonia. We found that in the human spleen infected with pneumococci, clusters with increasing number of bacteria were detectable within macrophages. These clusters also were detected in non-human primates. When intranasally infected mice were treated with a non-therapeutic dose of azithromycin, which had no effect on pneumonia but concentrated inside splenic macrophages, bacteria were absent from the spleen and blood and importantly mice had no signs of disease. INTERPRETATION: We conclude that the bacterial load in the spleen, and not lung, correlates with the occurrence of bacteraemia. This supports the hypothesis that the spleen, and not the lungs, is the major source of bacteria during systemic infection associated with pneumococcal pneumonia; a finding that provides a mechanistic basis for using combination therapies including macrolides in the treatment of severe community-acquired pneumococcal pneumonia. FUNDING: Oxford University, Wolfson Foundation, MRC, NIH, NIHR, and MRC and BBSRC studentships supported the work.


Subject(s)
Bacteremia/microbiology , Macrophages/microbiology , Pneumonia, Pneumococcal/microbiology , Spleen/microbiology , Animals , Bacterial Load/physiology , Community-Acquired Infections/microbiology , Disease Models, Animal , Female , Humans , Mice , Papio/microbiology , Streptococcus pneumoniae/pathogenicity
10.
Infect Immun ; 89(12): e0040021, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34491792

ABSTRACT

During its progression from the nasopharynx to other sterile and nonsterile niches of its human host, Streptococcus pneumoniae must cope with changes in temperature. We hypothesized that the temperature adaptation is an important facet of pneumococcal survival in the host. Here, we evaluated the effect of temperature on pneumococcus and studied the role of glutamate dehydrogenase (GdhA) in thermal adaptation associated with virulence and survival. Microarray analysis revealed a significant transcriptional response to changes in temperature, affecting the expression of 252 genes in total at 34°C and 40°C relative to at 37°C. One of the differentially regulated genes was gdhA, which is upregulated at 40°C and downregulated at 34°C relative to 37°C. Deletion of gdhA attenuated the growth, cell size, biofilm formation, pH survival, and biosynthesis of proteins associated with virulence in a temperature-dependent manner. Moreover, deletion of gdhA stimulated formate production irrespective of temperature fluctuation. Finally, ΔgdhA grown at 40°C was less virulent than other temperatures or the wild type at the same temperature in a Galleria mellonella infection model, suggesting that GdhA is required for pneumococcal virulence at elevated temperature.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Glutamate Dehydrogenase/genetics , Host-Pathogen Interactions , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/physiology , Temperature , Adaptation, Biological , Bacterial Proteins/metabolism , Biofilms/growth & development , Glutamate Dehydrogenase/metabolism , Humans , Microbial Viability , Virulence/genetics , Virulence Factors
11.
Mol Microbiol ; 116(3): 996-1008, 2021 09.
Article in English | MEDLINE | ID: mdl-34328238

ABSTRACT

Rggs are a group of transcriptional regulators with diverse roles in metabolism and virulence. Here, we present work on the Rgg1518/SHP1518 quorum sensing system of Streptococcus pneumoniae. The activity of Rgg1518 is induced by its cognate peptide, SHP1518. In vitro analysis showed that the Rgg1518 system is active in conditions rich in galactose and mannose, key nutrients during nasopharyngeal colonization. Rgg1518 expression is highly induced in the presence of these sugars and its isogenic mutant is attenuated in growth on galactose and mannose. When compared with other Rgg systems, Rgg1518 has the largest regulon on galactose. On galactose it controls up- or downregulation of a functionally diverse set of genes involved in galactose metabolism, capsule biosynthesis, iron metabolism, protein translation, as well as other metabolic functions, acting mainly as a repressor of gene expression. Rgg1518 is a repressor of capsule biosynthesis, and binds directly to the capsule regulatory region. Comparison with other Rggs revealed inter-regulatory interactions among Rggs. Finally, the rgg1518 mutant is attenuated in colonization and virulence in a mouse model of colonization and pneumonia. We conclude that Rgg1518 is a virulence determinant that contributes to a regulatory network composed of multiple Rgg systems.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Galactose/metabolism , Mannose/metabolism , Quorum Sensing , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Animals , Carbohydrate Metabolism , Female , Gene Expression Regulation, Bacterial , Humans , Mice , Mutation , Pneumococcal Infections/microbiology , Promoter Regions, Genetic , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/pathogenicity , Virulence , Virulence Factors/metabolism
12.
PLoS Pathog ; 16(4): e1008464, 2020 04.
Article in English | MEDLINE | ID: mdl-32324805

ABSTRACT

Streptococcus pneumoniae is a major human pathogen, causing pneumonia and sepsis. Genetic components strongly influence host responses to pneumococcal infections, but the responsible loci are unknown. We have previously identified a locus on mouse chromosome 7 from a susceptible mouse strain, CBA/Ca, to be crucial for pneumococcal infection. Here we identify a responsible gene, Cd22, which carries a point mutation in the CBA/Ca strain, leading to loss of CD22 on B cells. CBA/Ca mice and gene-targeted CD22-deficient mice on a C57BL/6 background are both similarly susceptible to pneumococcal infection, as shown by bacterial replication in the lungs, high bacteremia and early death. After bacterial infections, CD22-deficient mice had strongly reduced B cell populations in the lung, including GM-CSF producing, IgM secreting innate response activator B cells, which are crucial for protection. This study provides striking evidence that CD22 is crucial for protection during invasive pneumococcal disease.


Subject(s)
B-Lymphocytes/immunology , Pneumococcal Infections/immunology , Sialic Acid Binding Ig-like Lectin 2/immunology , Animals , B-Lymphocytes/microbiology , Bacteremia/genetics , Bacteremia/immunology , Bacteremia/microbiology , Female , Host-Pathogen Interactions , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Pneumococcal Infections/genetics , Pneumococcal Infections/metabolism , Pneumonia, Pneumococcal/genetics , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/microbiology , Sialic Acid Binding Ig-like Lectin 2/deficiency , Sialic Acid Binding Ig-like Lectin 2/genetics , Streptococcus pneumoniae/pathogenicity
13.
Sci Rep ; 10(1): 4585, 2020 03 12.
Article in English | MEDLINE | ID: mdl-32165654

ABSTRACT

Oligomers of pneumolysin form transmembrane channels in cholesterol-containing lipid bilayers. The mechanism of pore formation involves a multistage process in which the protein, at first, assembles into a ring-shaped complex on the outer-bilayer leaflet. In a subsequent step, the complex inserts into the membrane. Contrary to most investigations of pore formation that have focussed on protein changes, we have deduced how the lipid-packing order is altered in different stages of the pore-forming mechanism. An optical tweezing apparatus was used, in combination with microfluidics, to isolate large-unilamellar vesicles and control exposure of the bilayer to pneumolysin. By monitoring Raman-scattered light from a single-trapped liposome, the effect of the protein on short-range order and rotational diffusion of lipids could be inferred from changes in the envelope of the C-H stretch. A significant change in the lipid-packing order takes place during assembly of pre-pore oligomers. We were not able to detect a change in the lipid-packing order during the initial stage of protein binding, or any further change during the insertion of oligomers. Pre-pore complexes induce a transformation in which a bilayer, resembling a liquid-ordered phase is changed into a bilayer resembling a fluid-liquid-disordered phase surrounding ordered microdomains enriched in cholesterol and protein complexes.


Subject(s)
Cholesterol/metabolism , Streptococcus pneumoniae/metabolism , Streptolysins/chemistry , Streptolysins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cholesterol/chemistry , Hemolysis , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Microfluidic Analytical Techniques , Models, Molecular , Mutation , Optical Tweezers , Protein Binding , Spectrum Analysis, Raman , Streptolysins/genetics , Unilamellar Liposomes/chemistry , Unilamellar Liposomes/metabolism
14.
Article in English | MEDLINE | ID: mdl-31572692

ABSTRACT

Streptococcus pneumoniae is able to cause deadly diseases by infecting different tissues, each with distinct environmental and nutritional compositions. We hypothesize that the adaptive capabilities of the microbe is an important facet of pneumococcal survival in fluctuating host environments. Quorum-sensing (QS) mechanisms are pivotal for microbial host adaptation. We previously demonstrated that the TprA/PhrA QS system is required for pneumococcal utilization of galactose and mannose, neuraminidase activity, and virulence. We also showed that the system can be modulated by using linear molecularly imprinted polymers. Due to being a drugable target, we further studied the operation of this QS system in S. pneumoniae. We found that TprA controls the expression of nine different operons on galactose and mannose. Our data revealed that TprA expression is modulated by a complex regulatory network, where the master regulators CcpA and GlnR are involved in a sugar dependent manner. Mutants in the TprA/PhrA system are highly attenuated in their survival in nasopharynx and lungs after intranasal infection, and growth in blood after intravenous infection.


Subject(s)
Blood/microbiology , DNA-Binding Proteins/metabolism , Microbial Viability , Quorum Sensing , Respiratory System/microbiology , Streptococcus pneumoniae/physiology , Transcription Factors/metabolism , Adaptation, Physiological , Animals , Bacterial Proteins , Carbohydrate Metabolism , DNA-Binding Proteins/genetics , Disease Models, Animal , Gene Expression Regulation, Bacterial , Gene Regulatory Networks , Mice , Pneumococcal Infections/microbiology , Transcription Factors/genetics
15.
Infect Immun ; 87(1)2019 01.
Article in English | MEDLINE | ID: mdl-30323030

ABSTRACT

Complement is a critical component of antimicrobial immunity. Various complement regulatory proteins prevent host cells from being attacked. Many pathogens have acquired the ability to sequester complement regulators from host plasma to evade complement attack. We describe here how Streptococcus pneumoniae adopts a strategy to prevent the formation of the C3 convertase C4bC2a by the rapid conversion of surface bound C4b and iC4b into C4dg, which remains bound to the bacterial surface but no longer forms a convertase complex. Noncapsular virulence factors on the pneumococcus are thought to facilitate this process by sequestering C4b-binding protein (C4BP) from host plasma. When S. pneumoniae D39 was opsonized with human serum, the larger C4 activation products C4b and iC4b were undetectable, but the bacteria were liberally decorated with C4dg and C4BP. With targeted deletions of either PspA or PspC, C4BP deposition was markedly reduced, and there was a corresponding reduction in C4dg and an increase in the deposition of C4b and iC4b. The effect was greatest when PspA and PspC were both knocked out. Infection experiments in mice indicated that the deletion of PspA and/or PspC resulted in the loss of bacterial pathogenicity. Recombinant PspA and PspC both bound serum C4BP, and both led to increased C4b and reduced C4dg deposition on S. pneumoniae D39. We conclude that PspA and PspC help the pneumococcus to evade complement attack by binding C4BP and so inactivating C4b.


Subject(s)
Bacterial Proteins/metabolism , Complement C4b-Binding Protein/metabolism , Complement C4b/antagonists & inhibitors , Immune Evasion , Streptococcus pneumoniae/immunology , Animals , Disease Models, Animal , Humans , Mice , Pneumococcal Infections/microbiology , Protein Binding , Streptococcus pneumoniae/pathogenicity
16.
Sci Rep ; 8(1): 6369, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29686372

ABSTRACT

Microbes communicate with each other by using quorum sensing (QS) systems and modulate their collective 'behavior' for in-host colonization and virulence, biofilm formation, and environmental adaptation. The recent increase in genome data availability reveals the presence of several putative QS sensing circuits in microbial pathogens, but many of these have not been functionally characterized yet, despite their possible utility as drug targets. To increase the repertoire of functionally characterized QS systems in bacteria, we studied Rgg144/Shp144 and Rgg939/Shp939, two putative QS systems in the important human pathogen Streptococcus pneumoniae. We find that both of these QS circuits are induced by short hydrophobic peptides (Shp) upon sensing sugars found in the respiratory tract, such as galactose and mannose. Microarray analyses using cultures grown on mannose and galactose revealed that the expression of a large number of genes is controlled by these QS systems, especially those encoding for essential physiological functions and virulence-related genes such as the capsular locus. Moreover, the array data revealed evidence for cross-talk between these systems. Finally, these Rgg systems play a key role in colonization and virulence, as deletion mutants of these QS systems are attenuated in the mouse models of colonization and pneumonia.


Subject(s)
Bacterial Capsules/physiology , Bacterial Proteins/metabolism , Mannose/metabolism , Peptide Fragments/pharmacology , Pneumococcal Infections/microbiology , Quorum Sensing , Streptococcus pneumoniae/physiology , Animals , Bacterial Proteins/genetics , Female , Galactose/metabolism , Gene Expression Regulation, Bacterial , Mice , Pneumococcal Infections/drug therapy , Pneumococcal Infections/metabolism , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/pathogenicity , Virulence
17.
Nat Microbiol ; 3(5): 600-610, 2018 05.
Article in English | MEDLINE | ID: mdl-29662129

ABSTRACT

Bacterial septicaemia is a major cause of mortality, but its pathogenesis remains poorly understood. In experimental pneumococcal murine intravenous infection, an initial reduction of bacteria in the blood is followed hours later by a fatal septicaemia. These events represent a population bottleneck driven by efficient clearance of pneumococci by splenic macrophages and neutrophils, but as we show in this study, accompanied by occasional intracellular replication of bacteria that are taken up by a subset of CD169+ splenic macrophages. In this model, proliferation of these sequestered bacteria provides a reservoir for dissemination of pneumococci into the bloodstream, as demonstrated by its prevention using an anti-CD169 monoclonal antibody treatment. Intracellular replication of pneumococci within CD169+ splenic macrophages was also observed in an ex vivo porcine spleen, where the microanatomy is comparable with humans. We also showed that macrolides, which effectively penetrate macrophages, prevented septicaemia, whereas beta-lactams, with inefficient intracellular penetration, failed to prevent dissemination to the blood. Our findings define a shift in our understanding of the pneumococcus from an exclusively extracellular pathogen to one with an intracellular phase. These findings open the door to the development of treatments that target this early, previously unrecognized intracellular phase of bacterial sepsis.


Subject(s)
DNA, Bacterial/genetics , Macrophages/microbiology , Pneumococcal Infections/complications , Sepsis/microbiology , Spleen/cytology , Streptococcus pneumoniae/physiology , Animals , DNA Replication , Disease Models, Animal , Humans , Macrolides/pharmacology , Macrolides/therapeutic use , Mice , Pneumococcal Infections/drug therapy , Sepsis/drug therapy , Sepsis/etiology , Sialic Acid Binding Ig-like Lectin 1/metabolism , Spleen/microbiology , Streptococcus pneumoniae/pathogenicity , Swine
18.
Environ Microbiol ; 20(4): 1576-1589, 2018 04.
Article in English | MEDLINE | ID: mdl-29521441

ABSTRACT

Excess copper is highly toxic and forms part of the host innate immune system's antibacterial arsenal, accumulating at sites of infection and acting within macrophages to kill engulfed pathogens. We show for the first time that a novel, horizontally gene transferred copper resistance locus (copXL), uniquely associated with the SCCmec elements of the highly virulent, epidemic, community acquired methicillin resistant Staphylococcus aureus (CA-MRSA) USA300, confers copper hyper-resistance. These genes are additional to existing core genome copper resistance mechanisms, and are not found in typical S. aureus lineages, but are increasingly identified in emerging pathogenic isolates. Our data show that CopX, a putative P1B-3 -ATPase efflux transporter, and CopL, a novel lipoprotein, confer copper hyper-resistance compared to typical S. aureus strains. The copXL genes form an operon that is tightly repressed in low copper environments by the copper regulator CsoR. Significantly, CopX and CopL are important for S. aureus USA300 intracellular survival within macrophages. Therefore, the emergence of new S. aureus clones with the copXL locus has significant implications for public health because these genes confer increased resistance to antibacterial copper toxicity, enhancing bacterial fitness by altering S. aureus interaction with innate immunity.


Subject(s)
Anti-Bacterial Agents/toxicity , Copper/toxicity , Drug Resistance, Bacterial/genetics , Macrophages/microbiology , Membrane Transport Proteins/genetics , Methicillin-Resistant Staphylococcus aureus , Gene Transfer, Horizontal/genetics , Humans , Immunity, Innate/immunology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/growth & development , Operon , Staphylococcal Infections/microbiology
19.
Angew Chem Int Ed Engl ; 56(52): 16555-16558, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29140595

ABSTRACT

We describe the development, characterization, and biological testing of a new type of linear molecularly imprinted polymer (LMIP) designed to act as an anti-infective by blocking the quorum sensing (QS) mechanism and so abrogating the virulence of the pathogen Streptococcus pneumoniae. The LMIP is prepared (polymerized) in presence of a template molecule, but unlike in traditional molecular imprinting approaches, no cross-linker is used. This results in soluble low-molecular-weight oligomers that can act as a therapeutic agent in vitro and in vivo. The LMIP was characterized by mass spectrometry to determine its monomer composition. Fragments identified were then aligned along the peptide template by computer modeling to predict the possible monomer sequence of the LMIP. These findings provide a proof of principle that LMIPs can be used to block QS, thus setting the stage for the development of LMIPs a novel drug-discovery platform and class of materials to target Gram-positive pathogens.


Subject(s)
Anti-Infective Agents/pharmacology , Polymers/chemistry , Quorum Sensing/drug effects , Streptococcus pneumoniae/physiology , ATP-Binding Cassette Transporters/chemistry , Anti-Infective Agents/chemistry , Bacterial Proteins/chemistry , Mass Spectrometry , Molecular Imprinting , Peptides/chemistry , Peptides/metabolism , Virulence/drug effects
20.
Sci Rep ; 7(1): 8589, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819244

ABSTRACT

A spectroscopic technique is presented that is able to identify rapid changes in the bending modulus and fluidity of vesicle lipid bilayers on the micrometer scale, and distinguish between the presence and absence of heterogeneities in lipid-packing order. Individual unilamellar vesicles have been isolated using laser tweezers and, by measuring the intensity modulation of elastic back-scattered light, changes in the biophysical properties of lipid bilayers were revealed. Our approach offers unprecedented temporal resolution and, uniquely, physical transformations of lipid bilayers can be monitored on a length scale of micrometers. As an example, the deformation of a membrane bilayer following the gel-to-fluid phase transition in a pure phospholipid vesicle was observed to take place across an interval of 54 ± 5 ms corresponding to an estimated full-width of only ~1 m°C. Dynamic heterogeneities in packing order were detected in mixed-lipid bilayers. Using a ternary mixture of lipids, the modulated-intensity profile of elastic back-scattered light from an optically-trapped vesicle revealed an abrupt change in the bending modulus of the bilayer which could be associated with the dissolution of ordered microdomains (i.e., lipid rafts). This occurred across an interval of 30 ± 5 ms (equivalent to ~1 m°C).

SELECTION OF CITATIONS
SEARCH DETAIL