Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 924
Filter
1.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Article in English | MEDLINE | ID: mdl-34732571

ABSTRACT

Many pathogenic bacteria are encased in a layer of capsular polysaccharide (CPS). This layer is important for virulence by masking surface antigens, preventing opsonophagocytosis, and avoiding mucus entrapment. The bacterial tyrosine kinase (BY-kinase) regulates capsule synthesis and helps bacterial pathogens to survive different host niches. BY-kinases autophosphorylate at the C-terminal tyrosine residues upon external stimuli, but the role of phosphorylation is still unclear. Here, we report that the BY-kinase CpsCD is required for growth in Streptococcus pneumoniae Cells lacking a functional cpsC or cpsD accumulated low molecular weight CPS and lysed because of the lethal sequestration of the lipid carrier undecaprenyl phosphate, resulting in inhibition of peptidoglycan (PG) synthesis. CpsC interacts with CpsD and the polymerase CpsH. CpsD phosphorylation reduces the length of CPS polymers presumably by controlling the activity of CpsC. Finally, pulse-chase experiments reveal the spatiotemporal coordination between CPS and PG synthesis. This coordination is dependent on CpsC and CpsD. Together, our study provides evidence that BY-kinases regulate capsule polymer length by fine-tuning CpsC activity through autophosphorylation.


Subject(s)
Bacterial Capsules/metabolism , Bacterial Proteins/metabolism , Galactosyltransferases/metabolism , Polysaccharides, Bacterial/metabolism , Protein-Tyrosine Kinases/metabolism , Streptococcus pneumoniae/enzymology , Bacterial Proteins/genetics , Galactosyltransferases/genetics , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/growth & development
2.
Proc Natl Acad Sci U S A ; 117(52): 33507-33518, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33318198

ABSTRACT

Streptococcus pneumoniae (Spn) colonizes the nasopharynx and can cause pneumonia. From the lungs it spreads to the bloodstream and causes organ damage. We characterized the in vivo Spn and mouse transcriptomes within the nasopharynx, lungs, blood, heart, and kidneys using three Spn strains. We identified Spn genes highly expressed at all anatomical sites and in an organ-specific manner; highly expressed genes were shown to have vital roles with knockout mutants. The in vivo bacterial transcriptome during colonization/disease was distinct from previously reported in vitro transcriptomes. Distinct Spn and host gene-expression profiles were observed during colonization and disease states, revealing specific genes/operons whereby Spn adapts to and influences host sites in vivo. We identified and experimentally verified host-defense pathways induced by Spn during invasive disease, including proinflammatory responses and the interferon response. These results shed light on the pathogenesis of Spn and identify therapeutic targets.


Subject(s)
Host-Pathogen Interactions/genetics , Pneumococcal Infections/genetics , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/physiology , Transcriptome/genetics , Animals , Colony Count, Microbial , Female , Gene Expression Regulation, Bacterial , Genes, Bacterial , Interferons/metabolism , Male , Mice, Inbred C57BL , Mutation/genetics , Phylogeny , Principal Component Analysis , Signal Transduction , Streptococcus pneumoniae/growth & development
3.
Proc Natl Acad Sci U S A ; 117(49): 31386-31397, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229573

ABSTRACT

Influenza A virus (IAV)-related mortality is often due to secondary bacterial infections, primarily by pneumococci. Here, we study how IAV-modulated changes in the lungs affect bacterial replication in the lower respiratory tract (LRT). Bronchoalveolar lavages (BALs) from coinfected mice showed rapid bacterial proliferation 4 to 6 h after pneumococcal challenge. Metabolomic and quantitative proteomic analyses demonstrated capillary leakage with efflux of nutrients and antioxidants into the alveolar space. Pneumococcal adaptation to IAV-induced inflammation and redox imbalance increased the expression of the pneumococcal chaperone/protease HtrA. Presence of HtrA resulted in bacterial growth advantage in the IAV-infected LRT and protection from complement-mediated opsonophagocytosis due to capsular production. Absence of HtrA led to growth arrest in vitro that was partially restored by antioxidants. Pneumococcal ability to grow in the IAV-infected LRT depends on the nutrient-rich milieu with increased levels of antioxidants such as ascorbic acid and its ability to adapt to and cope with oxidative damage and immune clearance.


Subject(s)
Antioxidants/metabolism , Capillaries/pathology , Influenza, Human/microbiology , Pneumococcal Infections/microbiology , Respiratory System/microbiology , Respiratory System/virology , Streptococcus pneumoniae/growth & development , Animals , Bacterial Proteins/metabolism , Glucose/metabolism , Humans , Inflammation/complications , Inflammation/pathology , Mice, Inbred C57BL , Models, Biological , Molecular Chaperones/metabolism , Orthomyxoviridae Infections/microbiology , Oxidation-Reduction , Oxidative Stress , Phagocytosis , Respiratory System/pathology
4.
Mol Microbiol ; 115(6): 1152-1169, 2021 06.
Article in English | MEDLINE | ID: mdl-33269494

ABSTRACT

Bacterial peptidoglycan (PG) synthesis requires strict spatiotemporal organization to reproduce specific cell shapes. In ovoid-shaped Streptococcus pneumoniae (Spn), septal and peripheral (elongation) PG synthesis occur simultaneously at midcell. To uncover the organization of proteins and activities that carry out these two modes of PG synthesis, we examined Spn cells vertically oriented onto their poles to image the division plane at the high lateral resolution of 3D-SIM (structured-illumination microscopy). Labeling with fluorescent D-amino acids (FDAA) showed that areas of new transpeptidase (TP) activity catalyzed by penicillin-binding proteins (PBPs) separate into a pair of concentric rings early in division, representing peripheral PG (pPG) synthesis (outer ring) and the leading-edge (inner ring) of septal PG (sPG) synthesis. Fluorescently tagged PBP2x or FtsZ locate primarily to the inner FDAA-marked ring, whereas PBP2b and FtsX remain in the outer ring, suggesting roles in sPG or pPG synthesis, respectively. Pulses of FDAA labeling revealed an arrangement of separate regularly spaced "nodes" of TP activity around the division site of predivisional cells. Tagged PBP2x, PBP2b, and FtsX proteins also exhibited nodal patterns with spacing comparable to that of FDAA labeling. Together, these results reveal new aspects of spatially ordered PG synthesis in ovococcal bacteria during cell division.


Subject(s)
Cell Division/physiology , Peptidoglycan/biosynthesis , Streptococcus pneumoniae/metabolism , Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cell Cycle Proteins/metabolism , Fluorescent Dyes , Penicillin-Binding Proteins/metabolism , Peptidyl Transferases/metabolism , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/growth & development
5.
Mol Microbiol ; 116(2): 438-458, 2021 08.
Article in English | MEDLINE | ID: mdl-33811693

ABSTRACT

Streptococcus pneumoniae resides in the human upper airway as a commensal but also causes pneumonia, bacteremia, meningitis, and otitis media. It remains unclear how pneumococci adapt to nutritional conditions of various host niches. We here show that MetR, a LysR family transcriptional regulator, serves as a molecular adaptor for pneumococcal fitness, particularly in the upper airway. The metR mutant of strain D39 rapidly disappeared from the nasopharynx but was marginally attenuated in the lungs and bloodstream of mice. RNA-seq and ChIP-seq analyses showed that MetR broadly regulates transcription of the genes involved in methionine synthesis and other functions under methionine starvation. Genetic and biochemical analyses confirmed that MetR is essential for the activation of methionine synthesis but not uptake. Co-infection of influenza virus partially restored the colonization defect of the metR mutant. These results strongly suggest that MetR is particularly evolved for pneumococcal carriage in the upper airway of healthy individuals where free methionine is severely limited, but it becomes dispensable where environmental methionine is relatively more abundant (e.g., inflamed upper airway and sterile sites). To the best of our knowledge, MetR represents the first known regulator particularly for pneumococcal carriage in healthy individuals.


Subject(s)
Bacterial Proteins/genetics , Methionine/biosynthesis , Nasopharynx/microbiology , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/genetics , Trans-Activators/genetics , Animals , Bacterial Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Methionine/metabolism , Mice , Pneumococcal Infections/pathology , Trans-Activators/metabolism , Transcription, Genetic/genetics
6.
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
7.
PLoS Pathog ; 16(3): e1008332, 2020 03.
Article in English | MEDLINE | ID: mdl-32130269

ABSTRACT

Pathogens of the Streptococcus genus inhabit many different environmental niches during the course of an infection in a human host and the bacteria must adjust their metabolism according to available nutrients. Despite their lack of the citric-acid cycle, some streptococci proliferate in niches devoid of a readily available carbohydrate source. Instead they rely on carbohydrate scavenging for energy acquisition, which are obtained from the host. Here we discover a two-component system (TCS07) of Streptococcus pneumoniae that responds to glycoconjugated structures on proteins present on the host cells. Using next-generation RNA sequencing we find that the uncharacterized TCS07 regulon encodes proteins important for host-glycan processing and transporters of the released glycans, as well as intracellular carbohydrate catabolizing enzymes. We find that a functional TCS07 allele is required for growth on the glycoconjugated model protein fetuin. Consistently, we see a TCS07-dependent activation of the glycan degradation pathway. Thus, we pinpoint the molecular constituents responsible for sensing host derived glycans and link this to the induction of the proteins necessary for glycan degradation. Furthermore, we connect the TCS07 regulon to virulence in a mouse model, thereby establishing that host-derived glycan-metabolism is important for infection in vivo. Finally, a comparative phylogenomic analysis of strains from the Streptococcus genus reveal that TCS07 and most of its regulon is specifically conserved in species that utilize host-glycans for growth.


Subject(s)
Bacterial Proteins/metabolism , Pneumococcal Infections/metabolism , Polysaccharides/metabolism , Streptococcus pneumoniae/metabolism , Animals , Bacterial Proteins/genetics , Genome, Bacterial , Host-Pathogen Interactions , Humans , Mice , Pneumococcal Infections/microbiology , Regulon , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/pathogenicity , Virulence
8.
BMC Microbiol ; 22(1): 31, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35057744

ABSTRACT

BACKGROUND: Nasopharyngeal colonization is considered a necessary step in the initiation of pneumococcal diseases. Real time PCR (RT-PCR) is an alternative approach for the identification and quantification of pneumococci directly from samples. OBJECTIVES: To compare pneumococcal detection rates using culture-based method versus RT-PCR direct detection and to quantify pneumococcal colonization in two study cohorts (healthy children and hospitalized children with respiratory symptoms) using quantitation through RT-PCR. METHODOLOGY: A total of 101 nasopharyngeal swabs (NPS) from healthy children and 183 NPSs from hospitalized children with respiratory symptoms were included in the study. None of the children were vaccinated. All children were between 2 months to 2 years. In parallel to routine culture and identification, a RT-PCR assay targeting the lytA gene was done. RESULTS: Considering all 284 samples tested, colonization rate by conventional culture was 41.2% (n = 117) while positive colonization using RT-PCR was 43.7% (n = 124). The colonization rate detected by RT-PCR in the healthy cohort was 33.7% (n = 34) and it was 49.2% (n = 90) in the hospitalized cohort. It was 37.6% (n = 38) and 43.2% (n = 79) for the two cohorts by culture. The mean Cq value for the healthy cohort is 29.61 (SD 2.85) and 28.93 (SD 3.62) for the hospitalized cohort. With the standard curve obtained from amplifying a dilution series of control DNA, the mean amount of genomic DNA copy numbers detected in children with respiratory symptoms was log10 7.49 (SD 1.07) while it was log10 7.30 (SD 0.23) in healthy children and the difference was not statistically significant. CONCLUSIONS: The overall colonization rate was higher when detected using RT-PCR compared to culture. However, it was lower in the healthy group when detected with RT-PCR compared to culture. Even though there was a higher detection of pneumococcal colonization density in children with respiratory symptoms, this was not significantly higher unlike many previous studies. Therefore, the use of RT-PCR to detect pneumococcal colonization needs further evaluation with careful analysis of interpretation and confounders.


Subject(s)
Bacteriological Techniques/standards , Hospitalization/statistics & numerical data , Pneumococcal Infections/microbiology , Real-Time Polymerase Chain Reaction/standards , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/genetics , Child, Preschool , Cohort Studies , Healthy Volunteers/statistics & numerical data , Humans , Infant , Nasopharynx/microbiology , Real-Time Polymerase Chain Reaction/methods , Respiratory Tract Infections/microbiology , Streptococcus pneumoniae/isolation & purification
9.
Eur J Clin Microbiol Infect Dis ; 41(1): 37-44, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34432166

ABSTRACT

The otopathogens colonizing the nasopharynx (NP) and causing acute otitis media (AOM) have shown dynamic changes following introduction of pneumococcal conjugate vaccines. Five hundred eighty-nine children were prospectively enrolled, 2015-2019. Two thousand fifty-nine visits (1528 healthy, 393 AOM, and 138 AOM follow-up) were studied. Two thousand forty-two NP and 495 middle ear fluid (MEF) samples by tympanocentesis from 319 AOM cases were cultured for bacterial identification and antibiotic susceptibility. Streptococcus pneumoniae (Spn) isolates were serotyped by Quellung, and multi-locus sequence type (ST) determined by genomic analysis. Haemophilus influenzae (Hi) was the most common otopathogen cultured from MEF during AOM (34% in MEF) followed by Spn (24% in MEF), then Moraxella catarrhalis (Mcat) (15% in MEF). NP isolates during healthy visit were Mcat (39%), Spn (32%), Hi (12%). 48.6% of Hi isolates from MEF were beta-lactamase-producing. Spn non-susceptibility to penicillin and other antibiotics was high. The most common Spn serotypes associated with AOM (and colonizing the NP during healthy visits) were 35B, 23B, and 15B/C. ST558 and ST199 were the most common sequence types. During 2015-2019, Hi was the most common otopathogen cultured from MEF during AOM among young children. Pneumococcal AOM was most commonly caused by non-PCV13 serotypes of Spn, predominantly 35B, 23B, and 15B/C. Resistance to common antibiotics among Spn strains showed an increasing trend.


Subject(s)
Nasopharynx/microbiology , Otitis Media/microbiology , Pneumococcal Infections/microbiology , Pneumococcal Vaccines/administration & dosage , Streptococcus pneumoniae/growth & development , Anti-Bacterial Agents/pharmacology , Child, Preschool , Female , Humans , Infant , Longitudinal Studies , Male , Otitis Media/prevention & control , Phylogeny , Pneumococcal Infections/prevention & control , Prospective Studies , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/immunology
10.
Infect Immun ; 89(12): e0046321, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34543118

ABSTRACT

Streptococcus pneumoniae colonizes the nasopharynx of children and the elderly but also kills millions worldwide yearly. The secondary bile acid metabolite deoxycholic acid (DoC) affects the viability of human pathogens but also plays multiple roles in host physiology. We assessed in vitro the antimicrobial activity of DoC and investigated its potential to eradicate S. pneumoniae colonization using a model of human nasopharyngeal colonization and an in vivo mouse model of colonization. At a physiological concentration, DoC (0.5 mg/ml; 1.27 mM) killed all tested S. pneumoniae strains (n = 48) 2 h postinoculation. The model of nasopharyngeal colonization showed that DoC eradicated colonization by S. pneumoniae strains as soon as 10 min postexposure. The mechanism of action did not involve activation of autolysis, since the autolysis-defective double mutants ΔlytAΔlytC and ΔspxBΔlctO were as susceptible to DoC as was the wild type (WT). Oral streptococcal species (n = 20), however, were not susceptible to DoC (0.5 mg/ml). Unlike trimethoprim, whose spontaneous resistance frequency (srF) for TIGR4 or EF3030 was ≥1 × 10-9, no spontaneous resistance was observed with DoC (srF, ≥1 × 10-12). Finally, the efficacy of DoC to eradicate S. pneumoniae colonization was assessed in vivo using a topical route via intranasal (i.n.) administration and as a prophylactic treatment. Mice challenged with S. pneumoniae EF3030 carried a median of 4.05 × 105 CFU/ml 4 days postinoculation compared to 6.67 × 104 CFU/ml for mice treated with DoC. Mice in the prophylactic group had an ∼99% reduction of the pneumococcal density (median, 2.61 × 103 CFU/ml). Thus, DoC, an endogenous human bile salt, has therapeutic potential against S. pneumoniae.


Subject(s)
Deoxycholic Acid/pharmacology , Host-Pathogen Interactions , Pneumococcal Infections/microbiology , Pneumococcal Infections/prevention & control , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/growth & development , Animals , Bile Acids and Salts/metabolism , Deoxycholic Acid/metabolism , Disease Models, Animal , Disease Susceptibility , Drug Resistance, Bacterial , Humans , Mice , Mutation , N-Acetylmuramoyl-L-alanine Amidase/genetics , Nasopharynx/microbiology , Pneumococcal Infections/metabolism , Streptococcus pneumoniae/genetics
11.
Microbiology (Reading) ; 167(5)2021 05.
Article in English | MEDLINE | ID: mdl-33983874

ABSTRACT

Streptococcus pneumoniae, S. pyogenes (Group A Streptococcus; GAS) and S. agalactiae (Group B Streptococcus; GBS) are major aetiological agents of diseases in humans. The cellular membrane, a crucial site in host-pathogen interactions, is poorly characterized in streptococci. Moreover, little is known about whether or how environmental conditions influence their lipid compositions. Using normal phase liquid chromatography coupled with electrospray ionization MS, we characterized the phospholipids and glycolipids of S. pneumoniae, GAS and GBS in routine undefined laboratory medium, streptococcal defined medium and, in order to mimic the host environment, defined medium supplemented with human serum. In human serum-supplemented medium, all three streptococcal species synthesize phosphatidylcholine (PC), a zwitterionic phospholipid commonly found in eukaryotes but relatively rare in bacteria. We previously reported that S. pneumoniae utilizes the glycerophosphocholine (GPC) biosynthetic pathway to synthesize PC. Through substrate tracing experiments, we confirm that GAS and GBS scavenge lysoPC, a major metabolite in human serum, thereby using an abbreviated GPC pathway for PC biosynthesis. Furthermore, we found that plasmanyl-PC is uniquely present in the GBS membrane during growth with human serum, suggesting GBS possesses unusual membrane biochemical or biophysical properties. In summary, we report cellular lipid remodelling by the major pathogenic streptococci in response to metabolites present in human serum.


Subject(s)
Phospholipids/metabolism , Serum/metabolism , Streptococcal Infections/microbiology , Streptococcus agalactiae/metabolism , Streptococcus pneumoniae/metabolism , Streptococcus pyogenes/metabolism , Cell Membrane/chemistry , Cell Membrane/genetics , Culture Media/metabolism , Humans , Phospholipids/chemistry , Streptococcal Infections/blood , Streptococcus agalactiae/chemistry , Streptococcus agalactiae/growth & development , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/growth & development , Streptococcus pyogenes/chemistry , Streptococcus pyogenes/growth & development
12.
PLoS Pathog ; 15(8): e1007957, 2019 08.
Article in English | MEDLINE | ID: mdl-31437249

ABSTRACT

Human zinc deficiency increases susceptibility to bacterial infection. Although zinc supplementation therapies can reduce the impact of disease, the molecular basis for protection remains unclear. Streptococcus pneumoniae is a major cause of bacterial pneumonia, which is prevalent in regions of zinc deficiency. We report that dietary zinc levels dictate the outcome of S. pneumoniae infection in a murine model. Dietary zinc restriction impacts murine tissue zinc levels with distribution post-infection altered, and S. pneumoniae virulence and infection enhanced. Although the activation and infiltration of murine phagocytic cells was not affected by zinc restriction, their efficacy of bacterial control was compromised. S. pneumoniae was shown to be highly sensitive to zinc intoxication, with this process impaired in zinc restricted mice and isolated phagocytic cells. Collectively, these data show how dietary zinc deficiency increases sensitivity to S. pneumoniae infection while revealing a role for zinc as a component of host antimicrobial defences.


Subject(s)
Dietary Supplements , Disease Models, Animal , Lung Diseases/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Virulence/drug effects , Zinc/administration & dosage , Animals , Female , Lung Diseases/drug therapy , Lung Diseases/microbiology , Mice , Pneumococcal Infections/drug therapy , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/growth & development
13.
Eur J Clin Microbiol Infect Dis ; 40(11): 2397-2401, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33797644

ABSTRACT

Streptococcus pneumoniae (pneumococcus) is a human pathogen that colonizes the nasopharynx. We investigated serotype distribution in paired invasive and nasopharyngeal samples obtained from 57 children during invasive pneumococcal disease. Of 39 nasopharyngeal samples positive for pneumococci, 46.2% contained a serotype different from the one causing disease. This study reports a high frequency of pneumococcal multiple serotype carriage in children with invasive pneumococcal disease. Whether multiple serotype carriage is important for the onset and progress to pneumococcal infection warrants further investigation.


Subject(s)
Carrier State/microbiology , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/isolation & purification , Child, Preschool , Female , Humans , Infant , Male , Nasopharynx/microbiology , Prospective Studies , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/growth & development
14.
World J Microbiol Biotechnol ; 37(11): 187, 2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34586515

ABSTRACT

Bacterial meningitis remains a very important disease worldwide, and the major causative pathogens were Neisseria meningitidis (N. meningitidis), Streptococcus pneumoniae (S. pneumoniae) and Haemophilus influenzae (H. influenzae). In our context, the technical difficulties encountered in the routine practice were associated with the fragility of these bacteria, the high rates of negative culture and the demanding transport conditions. That's why the need to look for a solution to its technical problems and to propose a new proper solution with the local situation. The aim of this study was to develop, perform and evaluate a novel biphasic medium used for the transport, culture and conservation at an ambient temperature of N. meningitidis, S. pneumoniae and H. influenzae. The results showed that this biphasic medium provided more, novels and easy nutriments through the addition of liquid phase and solid phase medium and it was found to be conducive to the growth and conservation of N. meningitidis, S. pneumoniae and H. influenzae at an ambient temperature of a minimum of 40 days. And the ingredients used in the medium are readily available at a low cost as well as the components prepared in large quantities, they could be stored at + 4 ± 1 °C for 2 years without significantly altering their growth and conservation supporting their potential. The survival and recovery for the fastidious bacteria on the biphasic medium and the other media used for comparison in this study were significantly different (P < 0.05). In addition, the Sensitivity, Specificity, Positive and Negative Predictive Value of biphasic medium showed highest among the three bacteria at least 40 days of storage at room temperature in this study. In conclusion, we found the biphasic medium to be low cost and suitable for previously mentioned bacteria from suspected meningitis patients, offering an optimal condition and an increase in the viability of the isolates at ambient temperature. And it was concluded that this biphasic medium could be used as a technical solution in laboratories for the management of meningitis.


Subject(s)
Culture Media/chemistry , Haemophilus influenzae/isolation & purification , Neisseria meningitidis/isolation & purification , Streptococcus pneumoniae/isolation & purification , Temperature , Bacteria , DNA, Bacterial , Haemophilus influenzae/genetics , Haemophilus influenzae/growth & development , Humans , Meningitis, Bacterial/microbiology , Neisseria meningitidis/genetics , Neisseria meningitidis/growth & development , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/growth & development
15.
J Bacteriol ; 203(1)2020 12 07.
Article in English | MEDLINE | ID: mdl-33046563

ABSTRACT

Streptococcus pneumoniae (the pneumococcus) is a formidable human pathogen that is capable of asymptomatically colonizing the nasopharynx. Progression from colonization to invasive disease involves adaptation to distinct host niches, which vary markedly in the availability of key nutrients such as sugars. We previously reported that cell-cell signaling via the autoinducer 2 (AI-2)/LuxS quorum-sensing system boosts the capacity of S. pneumoniae to utilize galactose as a carbon source by upregulation of the Leloir pathway. This resulted in increased capsular polysaccharide production and a hypervirulent phenotype. We hypothesized that this effect was mediated by phosphorylation of GalR, the transcriptional activator of the Leloir pathway. GalR is known to possess three putative phosphorylation sites, S317, T319, and T323. In the present study, derivatives of S. pneumoniae D39 with putative phosphorylation-blocking alanine substitution mutations at each of these GalR sites (singly or in combination) were constructed. Growth assays and transcriptional analyses revealed complex phenotypes for these GalR mutants, with impacts on the regulation of both the Leloir and tagatose 6-phosphate pathways. The alanine substitution mutations significantly reduced the capacity of pneumococci to colonize the nasopharynx, middle ear, and lungs in a murine intranasal challenge model.IMPORTANCE Pneumococcal survival in the host and capacity to transition from a commensal to a pathogenic lifestyle are closely linked to the organism's ability to utilize specific nutrients in distinct niches. Galactose is a major carbon source for pneumococci in the upper respiratory tract. We have shown that both the Leloir and tagatose 6-phosphate pathways are necessary for pneumococcal growth in galactose and demonstrated GalR-mediated interplay between the two pathways. Moreover, the three putative phosphorylation sites in the transcriptional regulator GalR play a critical role in galactose metabolism and are important for pneumococcal colonization of the nasopharynx, middle ear, and lungs.


Subject(s)
Galactose/metabolism , Mutation/genetics , Repressor Proteins/genetics , Streptococcus pneumoniae/genetics , Animals , Ear, Middle/microbiology , Female , Galactose/genetics , Gene Expression , Humans , Lung/microbiology , Mice , Mutagenesis, Site-Directed , Nasopharynx/microbiology , Phosphorylation , Repressor Proteins/chemistry , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/metabolism
16.
Infect Immun ; 88(4)2020 03 23.
Article in English | MEDLINE | ID: mdl-31988172

ABSTRACT

The competence regulon of pneumococcus regulates both genetic transformation and virulence. However, competence induction during host infection has not been examined. By using the serotype 2 strain D39, we transcriptionally fused the firefly luciferase (luc) to competence-specific genes and spatiotemporally monitored the competence development in a mouse model of pneumonia-derived sepsis. In contrast to the universally reported short transient burst of competent state in vitro, the naturally developed competent state was prolonged and persistent during pneumonia-derived sepsis. The competent state began at approximately 20 h postinfection (hpi) and facilitated systemic invasion and sepsis development and progressed in different manners. In some mice, acute pneumonia quickly led to sepsis and death, accompanied by increasing intensity of the competence signal. In the remaining mice, pneumonia lasted longer, with the competence signal decreasing at first but increasing as the infection became septic. The concentration of pneumococcal inoculum (1 × 106 to 1 × 108 CFU/mouse) and postinfection lung bacterial burden did not appreciably impact the kinetics of competence induction. Exogenously provided competence stimulating peptide 1 (CSP1) failed to modulate the onset kinetics of competence development in vivo The competence shutoff regulator DprA was highly expressed during pneumonia-derived sepsis but failed to turn off the competent state in mice. Competent D39 bacteria propagated the competence signal through cell-to-cell contact rather than the classically described quorum-sensing mechanism. Finally, clinical pneumococcal strains of different serotypes were also able to develop natural competence during pneumonia-derived sepsis.


Subject(s)
DNA Transformation Competence , Pneumonia, Pneumococcal/complications , Pneumonia, Pneumococcal/microbiology , Sepsis/microbiology , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/genetics , Animals , Disease Models, Animal , Gene Expression Profiling , Mice , Virulence
17.
PLoS Pathog ; 14(10): e1007396, 2018 10.
Article in English | MEDLINE | ID: mdl-30379943

ABSTRACT

Young age is a risk factor for prolonged colonization by common pathogens residing in their upper respiratory tract (URT). Why children present with more persistent colonization is unknown and there is relatively little insight into the host-pathogen interactions that contribute to persistent colonization. To identify factors permissive for persistent colonization during infancy, we utilized an infant mouse model of Streptococcus pneumoniae colonization in which clearance from the mucosal surface of the URT requires many weeks to months. Loss of a single bacterial factor, the pore-forming toxin pneumolysin (Ply), and loss of a single host factor, IL-1α, led to more persistent colonization. Exogenous administration of Ply promoted IL-1 responses and clearance, and intranasal treatment with IL-1α was sufficient to reduce colonization density. Major factors known to affect the duration of natural colonization include host age and pneumococcal capsular serotype. qRT-PCR analysis of the uninfected URT mucosa showed reduced baseline expression of genes involved in IL-1 signaling in infant compared to adult mice. In line with this observation, IL-1 signaling was important in initiating clearance in adult mice but had no effect on early colonization of infant mice. In contrast to the effect of age, isogenic constructs of different capsular serotype showed differences in colonization persistence but induced similar IL-1 responses. Altogether, this work underscores the importance of toxin-induced IL-1α responses in determining the outcome of colonization, clearance versus persistence. Our findings about IL-1 signaling as a function of host age may provide an explanation for the increased susceptibility and more prolonged colonization during early childhood.


Subject(s)
Aging , Bacterial Capsules/physiology , Interleukin-1/metabolism , Pneumococcal Infections/transmission , Serogroup , Streptococcus pneumoniae/growth & development , Animals , Bacterial Proteins/metabolism , Disease Models, Animal , Host-Pathogen Interactions , Interleukin-1/genetics , Mice , Mice, Inbred C57BL , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/immunology , Streptolysins/metabolism
18.
PLoS Pathog ; 14(10): e1007328, 2018 10.
Article in English | MEDLINE | ID: mdl-30308062

ABSTRACT

Streptococcus pneumoniae (pneumococcus) is an opportunistic pathogen that causes otitis media, sinusitis, pneumonia, meningitis and sepsis. The progression to this pathogenic lifestyle is preceded by asymptomatic colonization of the nasopharynx. This colonization is associated with biofilm formation; the competence pathway influences the structure and stability of biofilms. However, the molecules that link the competence pathway to biofilm formation are unknown. Here, we describe a new competence-induced gene, called briC, and demonstrate that its product promotes biofilm development and stimulates colonization in a murine model. We show that expression of briC is induced by the master regulator of competence, ComE. Whereas briC does not substantially influence early biofilm development on abiotic surfaces, it significantly impacts later stages of biofilm development. Specifically, briC expression leads to increases in biofilm biomass and thickness at 72h. Consistent with the role of biofilms in colonization, briC promotes nasopharyngeal colonization in the murine model. The function of BriC appears to be conserved across pneumococci, as comparative genomics reveal that briC is widespread across isolates. Surprisingly, many isolates, including strains from clinically important PMEN1 and PMEN14 lineages, which are widely associated with colonization, encode a long briC promoter. This long form captures an instance of genomic plasticity and functions as a competence-independent expression enhancer that may serve as a precocious point of entry into this otherwise competence-regulated pathway. Moreover, overexpression of briC by the long promoter fully rescues the comE-deletion induced biofilm defect in vitro, and partially in vivo. These findings indicate that BriC may bypass the influence of competence in biofilm development and that such a pathway may be active in a subset of pneumococcal lineages. In conclusion, BriC is a part of the complex molecular network that connects signaling of the competence pathway to biofilm development and colonization.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Peptide Fragments/metabolism , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/growth & development , Virulence , Amino Acid Sequence , Animals , Chinchilla , Female , Mice , Pneumococcal Infections/genetics , Pneumococcal Infections/metabolism , Promoter Regions, Genetic , Sequence Homology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism
19.
PLoS Pathog ; 14(6): e1007118, 2018 06.
Article in English | MEDLINE | ID: mdl-29883472

ABSTRACT

Streptococcus pneumoniae is an opportunistic human bacterial pathogen that usually colonizes the upper respiratory tract, but the invasion and survival mechanism in respiratory epithelial cells remains elusive. Previously, we described that acidic stress-induced lysis (ASIL) and intracellular survival are controlled by ComE through a yet unknown activation mechanism under acidic conditions, which is independent of the ComD histidine kinase that activates this response regulator for competence development at pH 7.8. Here, we demonstrate that the serine/threonine kinase StkP is essential for ASIL, and show that StkP phosphorylates ComE at Thr128. Molecular dynamic simulations predicted that Thr128-phosphorylation induces conformational changes on ComE's DNA-binding domain. Using nonphosphorylatable (ComET128A) and phosphomimetic (ComET128E) proteins, we confirmed that Thr128-phosphorylation increased the DNA-binding affinity of ComE. The non-phosphorylated form of ComE interacted more strongly with StkP than the phosphomimetic form at acidic pH, suggesting that pH facilitated crosstalk. To identify the ComE-regulated genes under acidic conditions, a comparative transcriptomic analysis was performed between the comET128A and wt strains, and differential expression of 104 genes involved in different cellular processes was detected, suggesting that the StkP/ComE pathway induced global changes in response to acidic stress. In the comET128A mutant, the repression of spxB and sodA correlated with decreased H2O2 production, whereas the reduced expression of murN correlated with an increased resistance to cell wall antibiotic-induced lysis, compatible with cell wall alterations. In the comET128A mutant, ASIL was blocked and acid tolerance response was higher compared to the wt strain. These phenotypes, accompanied with low H2O2 production, are likely responsible for the increased survival in pneumocytes of the comET128A mutant. We propose that the StkP/ComE pathway controls the stress response, thus affecting the intracellular survival of S. pneumoniae in pneumocytes, one of the first barriers that this pathogen must cross to establish an infection.


Subject(s)
Acids/pharmacology , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Protein Serine-Threonine Kinases/metabolism , Streptococcus pneumoniae/growth & development , Stress, Physiological , A549 Cells , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Humans , Oxidative Stress/drug effects , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Streptococcus pneumoniae/drug effects
20.
BMC Microbiol ; 20(1): 16, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959125

ABSTRACT

BACKGROUND: The polysaccharide capsule is a major virulence factor of S. pneumoniae in diseases such as meningitis. While some capsular serotypes are more often found in invasive disease, high case fatality rates are associated with those serotypes more commonly found in asymptomatic colonization. We tested whether growth patterns and capsule size in human cerebrospinal fluid depends on serotype using a clinical isolate of S. pneumoniae and its capsule switch mutants. RESULTS: We found that the growth pattern differed markedly from that in culture medium by lacking the exponential and lysis phases. Growth in human cerebrospinal fluid was reduced when strains lost their capsules. When a capsule was present, growth was serotype-specific: high carriage serotypes (6B, 9 V, 19F and 23F) grew better than low carriage serotypes (7F, 14, 15B/C and 18C). Growth correlated with the case-fatality rates of serotypes reported in the literature. Capsule size in human cerebrospinal fluid also depended on serotype. CONCLUSIONS: We propose that serotype-specific differences in disease severity observed in meningitis patients may, at least in part, be explained by differences in growth and capsule size in human cerebrospinal fluid. This information could be useful to guide future vaccine design.


Subject(s)
Bacterial Capsules/genetics , Cerebrospinal Fluid/microbiology , Meningitis, Pneumococcal/cerebrospinal fluid , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/growth & development , Adult , Child , Culture Media/chemistry , Humans , Meningitis, Pneumococcal/microbiology , Microbial Viability , Mutation , Serotyping , Severity of Illness Index , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL