Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters

Country/Region as subject
Publication year range
1.
EMBO J ; 40(7): e106103, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33522633

ABSTRACT

Streptococcus agalactiae, also known as group B Streptococcus (GBS), is the major cause of neonatal sepsis in humans. A critical step to infection is adhesion of bacteria to epithelial surfaces. GBS adhesins have been identified to bind extracellular matrix components and cellular receptors. However, several putative adhesins have no host binding partner characterised. We report here that surface-expressed ß protein of GBS binds to human CEACAM1 and CEACAM5 receptors. A crystal structure of the complex showed that an IgSF domain in ß represents a novel Ig-fold subtype called IgI3, in which unique features allow binding to CEACAM1. Bioinformatic assessment revealed that this newly identified IgI3 fold is not exclusively present in GBS but is predicted to be present in adhesins from other clinically important human pathogens. In agreement with this prediction, we found that CEACAM1 binds to an IgI3 domain found in an adhesin from a different streptococcal species. Overall, our results indicate that the IgI3 fold could provide a broadly applied mechanism for bacteria to target CEACAMs.


Subject(s)
Adhesins, Bacterial/chemistry , Antigens, CD/chemistry , Carcinoembryonic Antigen/chemistry , Cell Adhesion Molecules/chemistry , Adhesins, Bacterial/metabolism , Animals , Antigens, CD/metabolism , Binding Sites , CHO Cells , Carcinoembryonic Antigen/metabolism , Cell Adhesion Molecules/metabolism , Cricetinae , Cricetulus , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/metabolism , HeLa Cells , Humans , Protein Binding , Streptococcus agalactiae/metabolism
2.
Eur J Clin Microbiol Infect Dis ; 43(3): 511-516, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38206519

ABSTRACT

PURPOSE: Rapid diagnosis and treatment of infectious meningitis and encephalitis (ME) is critical to minimize morbidity and mortality. Recently, Qiagen introduced the CE-IVD QIAstat-Dx ME panel (QS-ME) for syndromic diagnostic testing of meningitis and encephalitis. Some data on the performance of the QS-ME in comparison to the BioFire FilmArray ME panel are available. In this study, the performance of the QS-ME is compared to the current diagnostic workflow in two academic medical centers in the Netherlands. METHODS: A total of 110 cerebrospinal fluid samples were retrospectively tested with the QS-ME. The results obtained were compared to the results of laboratory-developed real-time PCR assays (LDTs), IS-pro, bacterial culture, and cryptococcal antigen (CrAg) testing. In addition, the accuracy of the QS-ME was also investigated using an external quality assessment (EQA) panel consisting of ten samples. RESULTS: Four of the 110 samples tested failed to produce a valid QS-ME result. In the remaining 106 samples, the QS-ME detected 53/53 viral targets, 38/40 bacterial targets, and 7/13 Cryptococcus neoformans targets. The discrepant bacterial results consisted of two samples that were previously tested positive for Listeria monocytogenes (CT 35.8) and Streptococcus pneumoniae (CT 40), respectively. The QS-ME detected one additional result, consisting of a varicella-zoster virus signal (CT 35.9), in a sample in which both techniques detected Streptococcus pyogenes. Finally, 100% concordance was achieved in testing a blinded bacterial ME EQA panel. CONCLUSION: The QS-ME is a relevant addition to the syndromic testing landscape to assist in diagnosing infectious ME.


Subject(s)
Cryptococcus neoformans , Encephalitis , Infectious Encephalitis , Meningitis, Bacterial , Meningitis , Humans , Retrospective Studies , Workflow , Multiplex Polymerase Chain Reaction/methods , Meningitis/diagnosis , Encephalitis/cerebrospinal fluid , Meningitis, Bacterial/diagnosis , Bacteria
3.
J Bacteriol ; 205(3): e0046222, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36856428

ABSTRACT

Neisseria meningitidis (meningococcus) colonizes the human nasopharynx, primarily as a commensal, but sporadically causing septicemia and meningitis. During colonization and invasion, it encounters different niches with specific nutrient compositions. Small noncoding RNAs (sRNAs) are used to fine-tune expression of genes, allowing adaptation to their physiological differences. We have previously characterized sRNAs (Neisseria metabolic switch regulators [NmsRs]) controlling switches between cataplerotic and anaplerotic metabolism. Here, we extend the NmsR regulon by studying methylcitrate lyase (PrpF) and propionate kinase (AckA-1) involved in the methylcitrate cycle and serine hydroxymethyltransferase (GlyA) and 3-hydroxyacid dehydrogenase (MmsB) involved in protein degradation. These proteins were previously shown to be dysregulated in a ΔnmsRs strain. Levels of transcription of target genes and NmsRs were assessed by reverse transcriptase quantitative PCR (RT-qPCR). We also used a novel gene reporter system in which the 5' untranslated region (5' UTR) of the target gene is fused to mcherry to study NmsRs-target gene interaction in the meningococcus. Under nutrient-rich conditions, NmsRs downregulate expression of PrpF and AckA-1 by direct interaction with the 5' UTR of their mRNA. Overexpression of NmsRs impaired growth under nutrient-limiting growth conditions with pyruvate and propionic acid as the only carbon sources. Our data strongly suggest that NmsRs downregulate propionate metabolism by lowering methylcitrate enzyme activity under nutrient-rich conditions. Under nutrient-poor conditions, NmsRs are downregulated, increasing propionate metabolism, resulting in higher tricarboxylic acid (TCA) activities. IMPORTANCE Neisseria meningitidis colonizes the human nasopharynx, forming a reservoir for the sporadic occurrence of epidemic invasive meningococcal disease like septicemia and meningitis. Propionic acid generated by other bacteria that coinhabit the human nasopharynx can be utilized by meningococci for replication in this environment. Here, we showed that sibling small RNAs, designated NmsRs, riboregulate propionic acid utilization by meningococci and, thus, colonization. Under conditions mimicking the nasopharyngeal environment, NmsRs are downregulated. This leads to the conversion of propionic acid to pyruvate and succinate, resulting in higher tricarboxylic acid cycle activity, allowing colonization of the nasopharynx. NmsRs link metabolic state with colonization, which is a crucial step on the trajectory to invasive meningococcal disease.


Subject(s)
Meningococcal Infections , Neisseria meningitidis , RNA, Small Untranslated , Humans , Propionates/metabolism , 5' Untranslated Regions , Siblings , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Pyruvates/metabolism
4.
PLoS Pathog ; 17(1): e1009227, 2021 01.
Article in English | MEDLINE | ID: mdl-33481964

ABSTRACT

Infections with Gram-negative bacteria form an increasing risk for human health due to antibiotic resistance. Our immune system contains various antimicrobial proteins that can degrade the bacterial cell envelope. However, many of these proteins do not function on Gram-negative bacteria, because the impermeable outer membrane of these bacteria prevents such components from reaching their targets. Here we show that complement-dependent formation of Membrane Attack Complex (MAC) pores permeabilizes this barrier, allowing antimicrobial proteins to cross the outer membrane and exert their antimicrobial function. Specifically, we demonstrate that MAC-dependent outer membrane damage enables human lysozyme to degrade the cell wall of E. coli. Using flow cytometry and confocal microscopy, we show that the combination of MAC pores and lysozyme triggers effective E. coli cell wall degradation in human serum, thereby altering the bacterial cell morphology from rod-shaped to spherical. Completely assembled MAC pores are required to sensitize E. coli to the antimicrobial actions of lysozyme and other immune factors, such as Human Group IIA-secreted Phospholipase A2. Next to these effects in a serum environment, we observed that the MAC also sensitizes E. coli to more efficient degradation and killing inside human neutrophils. Altogether, this study serves as a proof of principle on how different players of the human immune system can work together to degrade the complex cell envelope of Gram-negative bacteria. This knowledge may facilitate the development of new antimicrobials that could stimulate or work synergistically with the immune system.


Subject(s)
Anti-Infective Agents/pharmacology , Bacterial Outer Membrane/drug effects , Complement Activation , Complement Membrane Attack Complex/metabolism , Gram-Negative Bacteria/drug effects , Anti-Bacterial Agents/pharmacology , Cell Wall/drug effects , Escherichia coli/drug effects , Escherichia coli/immunology , Flow Cytometry , Gram-Negative Bacteria/immunology , Group II Phospholipases A2/metabolism , Humans , Microscopy, Confocal , Muramidase/metabolism , Neutrophils/microbiology , Phagocytes/microbiology
5.
Trends Immunol ; 41(4): 313-326, 2020 04.
Article in English | MEDLINE | ID: mdl-32151494

ABSTRACT

The enzyme type IIA secreted phospholipase A2 (sPLA2-IIA) is crucial for mammalian innate host defense against bacterial pathogens. Most studies have investigated the role of sPLA2-IIA in systemic bacterial infections, identifying molecular pathways of bacterial resistance against sPLA2-IIA-mediated killing, and providing insight into sPLA2-IIA mechanisms of action. Sensitization of (antibiotic-resistant) bacteria to sPLA2-IIA action by blocking bacterial resistance or by applying sPLA2-IIA to treat bacterial infections might represent a therapeutic option in the future. Because sPLA2-IIA is highly expressed at mucosal barriers, we also discuss how sPLA2-IIA is likely to be an important driver of microbiome composition; we anticipate that future research in this area may bring new insights into the role of sPLA2-IIA in health and disease.


Subject(s)
Bacterial Infections , Host Microbial Interactions , Phospholipases A2, Secretory , Animals , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/enzymology , Bacterial Infections/immunology , Bacterial Infections/therapy , Host Microbial Interactions/immunology , Humans , Phospholipases A2, Secretory/immunology , Sepsis/enzymology , Sepsis/immunology , Sepsis/therapy
6.
Nat Chem Biol ; 17(8): 878-887, 2021 08.
Article in English | MEDLINE | ID: mdl-34045745

ABSTRACT

In ovoid-shaped, Gram-positive bacteria, MapZ guides FtsZ-ring positioning at cell equators. The cell wall of the ovococcus Streptococcus mutans contains peptidoglycan decorated with serotype c carbohydrates (SCCs). In the present study, we identify the major cell separation autolysin AtlA as an SCC-binding protein. AtlA binding to SCC is attenuated by the glycerol phosphate (GroP) modification. Using fluorescently labeled AtlA constructs, we mapped SCC distribution on the streptococcal surface, revealing enrichment of GroP-deficient immature SCCs at the cell poles and equators. The immature SCCs co-localize with MapZ at the equatorial rings throughout the cell cycle. In GroP-deficient mutants, AtlA is mislocalized, resulting in dysregulated cellular autolysis. These mutants display morphological abnormalities associated with MapZ mislocalization, leading to FtsZ-ring misplacement. Altogether, our data support a model in which maturation of a cell wall polysaccharide provides the molecular cues for the recruitment of cell division machinery, ensuring proper daughter cell separation and FtsZ-ring positioning.


Subject(s)
Cell Wall/metabolism , Polysaccharides/metabolism , Streptococcus mutans/metabolism , Cell Division , Cell Wall/chemistry , Polysaccharides/chemistry , Streptococcus mutans/cytology
7.
Mol Cell Proteomics ; 20: 100034, 2021.
Article in English | MEDLINE | ID: mdl-33444734

ABSTRACT

Staphylococcus aureus is a major cause of infections worldwide, and infection results in a variety of diseases. As of no surprise, protein phosphorylation is an important game player in signaling cascades and has been shown to be involved in S. aureus virulence. Albeit long neglected, eukaryotic-type serine/threonine kinases in S. aureus have been implicated in this complex signaling cascades. Due to the substoichiometric nature of protein phosphorylation and a lack of suitable analysis tools, the knowledge of these cascades is, however, to date, still limited. Here, were apply an optimized protocol for efficient phosphopeptide enrichment via Fe3+-IMAC followed by LC-MS/MS to get a better understanding of the impact of protein phosphorylation on the complex signaling networks involved in pathogenicity. By profiling a serine/threonine kinase and phosphatase mutant from a methicillin-resistant S. aureus mutant library, we generated the most comprehensive phosphoproteome data set of S. aureus to date, aiding a better understanding of signaling in bacteria. With the identification of 3800 class I p-sites, we were able to increase the number of identifications by more than 21 times compared with recent literature. In addition, we were able to identify 74 downstream targets of the only reported eukaryotic-type Ser/Thr kinase of the S. aureus strain USA300, Stk1. This work allowed an extensive analysis of the bacterial phosphoproteome and indicates that Ser/Thr kinase signaling is far more abundant than previously anticipated in S. aureus.


Subject(s)
Bacterial Proteins/metabolism , Phosphopeptides/metabolism , Phosphoproteins/metabolism , Staphylococcus aureus/metabolism , Bacillus subtilis/metabolism , Bacterial Proteins/genetics , Phosphopeptides/genetics , Phosphoproteins/genetics , Phosphorylation , Protein Kinases/metabolism , Proteome , Staphylococcus aureus/genetics
8.
Euro Surveill ; 28(1)2023 01.
Article in English | MEDLINE | ID: mdl-36695447

ABSTRACT

In 2022, a sevenfold increase in the number of notifiable invasive Streptococcus pyogenes (iGAS) infections among children aged 0-5 years was observed in the Netherlands compared with pre-COVID-19 pandemic years. Of 42 cases in this age group, seven had preceding or coinciding varicella zoster infections, nine were fatal. This increase is not attributable to a specific emm type. Vigilance for clinical deterioration as iGAS sign is warranted in young children, especially those with varicella zoster infection.


Subject(s)
COVID-19 , Chickenpox , Herpes Zoster , Streptococcal Infections , Child , Humans , Child, Preschool , Adult , Streptococcus pyogenes , Streptococcal Infections/diagnosis , Streptococcal Infections/epidemiology , Netherlands/epidemiology , Pandemics
9.
Clin Infect Dis ; 74(12): 2173-2180, 2022 07 06.
Article in English | MEDLINE | ID: mdl-34525199

ABSTRACT

BACKGROUND: In response to the recent serogroup W invasive meningococcal disease (IMD-W) epidemic in the Netherlands, meningococcal serogroup C (MenC) conjugate vaccination for children aged 14 months was replaced with a MenACWY conjugate vaccination, and a mass campaign targeting individuals aged 14-18 years was executed. We investigated the impact of MenACWY vaccination implementation in 2018-2020 on incidence rates and estimated vaccine effectiveness (VE). METHODS: We extracted IMD cases diagnosed between July 2014 and December 2020 from the national surveillance system. We calculated age group-specific incidence rate ratios by comparing incidence rates before (July 2017-March 2018) and after (July 2019-March 2020) MenACWY vaccination implementation. We estimated VE in vaccine-eligible cases using the screening method. RESULTS: Overall, the IMD-W incidence rate declined by 61% (95% confidence interval [CI], 40 to 74). It declined by 82% (95% CI, 18 to 96) in the vaccine-eligible age group (individuals aged 15-36 months and 14-18 years) and by 57% (95% CI, 34 to 72) in vaccine-noneligible age groups. VE was 92% (95% CI, -20 to 99.5) in vaccine-eligible toddlers (aged 15-36 months). No IMD-W cases were reported in vaccine-eligible teenagers after the campaign. CONCLUSIONS: The MenACWY vaccination program was effective in preventing IMD-W in the target population. The IMD-W incidence reduction in vaccine-noneligible age groups may be caused by indirect effects of the vaccination program. However, disentangling natural fluctuation from vaccine effect was not possible. Our findings encourage the use of toddler and teenager MenACWY vaccination in national immunization programs.


Subject(s)
Meningococcal Infections , Meningococcal Vaccines , Neisseria meningitidis, Serogroup C , Adolescent , Humans , Meningococcal Infections/epidemiology , Meningococcal Infections/prevention & control , Netherlands/epidemiology , Serogroup , Vaccination/methods , Vaccines, Conjugate
10.
Clin Infect Dis ; 75(4): 719-722, 2022 09 10.
Article in English | MEDLINE | ID: mdl-35134152

ABSTRACT

The Streptococcus pneumoniae capsule is regarded as indispensable in bacteremia. We report an infant with a ventricular septal defect and infective endocarditis caused by nontypeable S. pneumoniae. In-depth investigation confirmed a deficient capsule yet favored pneumococcal fitness for causing infective endocarditis, rather than a host immune disorder, as the cause of infective endocarditis in this case.


Subject(s)
Endocarditis, Bacterial , Endocarditis , Pneumococcal Infections , Pneumonia , Endocarditis/diagnosis , Endocarditis, Bacterial/diagnosis , Humans , Infant , Pneumococcal Infections/diagnosis , Streptococcus pneumoniae
12.
FASEB J ; 35(10): e21875, 2021 10.
Article in English | MEDLINE | ID: mdl-34533845

ABSTRACT

Signal inhibitory receptor on leukocytes-1 (SIRL-1) is a negative regulator of myeloid cell function and dampens antimicrobial responses. We here show that different species of the genus Staphylococcus secrete SIRL-1-engaging factors. By screening a library of single-gene transposon mutants in Staphylococcus aureus, we identified these factors as phenol-soluble modulins (PSMs). PSMs are amphipathic α-helical peptides involved in multiple aspects of staphylococcal virulence and physiology. They are cytotoxic and activate the chemotactic formyl peptide receptor 2 (FPR2) on immune cells. Human cathelicidin LL-37 is also an amphipathic α-helical peptide with antimicrobial and chemotactic activities, structurally and functionally similar to α-type PSMs. We demonstrate that α-type PSMs from multiple staphylococcal species as well as human cathelicidin LL-37 activate SIRL-1, suggesting that SIRL-1 recognizes α-helical peptides with an amphipathic arrangement of hydrophobicity, although we were not able to show direct binding to SIRL-1. Upon rational peptide design, we identified artificial peptides in which the capacity to ligate SIRL-1 is segregated from cytotoxic and FPR2-activating properties, allowing specific engagement of SIRL-1. In conclusion, we propose staphylococcal PSMs and human LL-37 as a potential new class of natural ligands for SIRL-1.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Bacterial Toxins/metabolism , Peptide Fragments/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Sirtuin 1/metabolism , Staphylococcus aureus/metabolism , Humans , Quorum Sensing , Cathelicidins
13.
Chemistry ; 27(40): 10461-10469, 2021 Jul 16.
Article in English | MEDLINE | ID: mdl-33991006

ABSTRACT

Wall teichoic acids (WTAs) are important components of the cell wall of the opportunistic Gram-positive bacterium Staphylococcus aureus. WTAs are composed of repeating ribitol phosphate (RboP) residues that are decorated with d-alanine and N-acetyl-d-glucosamine (GlcNAc) modifications, in a seemingly random manner. These WTA-modifications play an important role in shaping the interactions of WTA with the host immune system. Due to the structural heterogeneity of WTAs, it is impossible to isolate pure and well-defined WTA molecules from bacterial sources. Therefore, here synthetic chemistry to assemble a broad library of WTA-fragments, incorporating all possible glycosylation modifications (α-GlcNAc at the RboP C4; ß-GlcNAc at the RboP C4; ß-GlcNAc at the RboP C3) described for S. aureus WTAs, is reported. DNA-type chemistry, employing ribitol phosphoramidite building blocks, protected with a dimethoxy trityl group, was used to efficiently generate a library of WTA-hexamers. Automated solid phase syntheses were used to assemble a WTA-dodecamer and glycosylated WTA-hexamer. The synthetic fragments have been fully characterized and diagnostic signals were identified to discriminate the different glycosylation patterns. The different glycosylated WTA-fragments were used to probe binding of monoclonal antibodies using WTA-functionalized magnetic beads, revealing the binding specificity of these WTA-specific antibodies and the importance of the specific location of the GlcNAc modifications on the WTA-chains.


Subject(s)
Staphylococcal Infections , Teichoic Acids , Cell Wall/metabolism , Glycosylation , Humans , Staphylococcus aureus/metabolism
14.
Nat Chem Biol ; 15(5): 463-471, 2019 05.
Article in English | MEDLINE | ID: mdl-30936502

ABSTRACT

Cell wall glycopolymers on the surface of Gram-positive bacteria are fundamental to bacterial physiology and infection biology. Here we identify gacH, a gene in the Streptococcus pyogenes group A carbohydrate (GAC) biosynthetic cluster, in two independent transposon library screens for its ability to confer resistance to zinc and susceptibility to the bactericidal enzyme human group IIA-secreted phospholipase A2. Subsequent structural and phylogenetic analysis of the GacH extracellular domain revealed that GacH represents an alternative class of glycerol phosphate transferase. We detected the presence of glycerol phosphate in the GAC, as well as the serotype c carbohydrate from Streptococcus mutans, which depended on the presence of the respective gacH homologs. Finally, nuclear magnetic resonance analysis of GAC confirmed that glycerol phosphate is attached to approximately 25% of the GAC N-acetylglucosamine side-chains at the C6 hydroxyl group. This previously unrecognized structural modification impacts host-pathogen interaction and has implications for vaccine design.


Subject(s)
Glycerol/metabolism , Phosphates/metabolism , Polysaccharides, Bacterial/metabolism , Streptococcus/metabolism , Glycerol/chemistry , Phosphates/chemistry , Polysaccharides, Bacterial/chemistry , Streptococcus/chemistry
15.
Proc Natl Acad Sci U S A ; 115(45): E10662-E10671, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30352847

ABSTRACT

Group A Streptococcus (GAS) is a Gram-positive bacterial pathogen that causes a range of diseases, including fatal invasive infections. However, the mechanisms by which the innate immune system recognizes GAS are not well understood. We herein report that the C-type lectin receptor macrophage inducible C-type lectin (Mincle) recognizes GAS and initiates antibacterial immunity. Gene expression analysis of myeloid cells upon GAS stimulation revealed the contribution of the caspase recruitment domain-containing protein 9 (CARD9) pathway to the antibacterial responses. Among receptors signaling through CARD9, Mincle induced the production of inflammatory cytokines, inducible nitric oxide synthase, and reactive oxygen species upon recognition of the anchor of lipoteichoic acid, monoglucosyldiacylglycerol (MGDG), produced by GAS. Upon GAS infection, Mincle-deficient mice exhibited impaired production of proinflammatory cytokines, severe bacteremia, and rapid lethality. GAS also possesses another Mincle ligand, diglucosyldiacylglycerol; however, this glycolipid interfered with MGDG-induced activation. These results indicate that Mincle plays a central role in protective immunity against acute GAS infection.


Subject(s)
Lectins, C-Type/metabolism , Lipopolysaccharides/metabolism , Membrane Proteins/metabolism , Streptococcal Infections/immunology , Streptococcus pyogenes/pathogenicity , Teichoic Acids/metabolism , Animals , CARD Signaling Adaptor Proteins/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Streptococcal Infections/microbiology
16.
Euro Surveill ; 26(42)2021 10.
Article in English | MEDLINE | ID: mdl-34676819

ABSTRACT

The incidence of most respiratory-transmitted diseases decreased during the COVID-19 pandemic as a result of containment measures. In contrast, in the Netherlands we noted an increase in invasive disease caused by Haemophilus influenzae b (Hib) (from < 0.3/100,000 before 2019 to 0.39 and 0.33/100,000 in 2020 and 2021) in vaccinated and unvaccinated age groups. We did not find a change in vaccine effectiveness against Hib invasive disease (effectiveness > 90%). We discuss factors that may have contributed to this rise.


Subject(s)
COVID-19 , Haemophilus Infections , Haemophilus Vaccines , Haemophilus influenzae type b , Haemophilus Infections/epidemiology , Haemophilus Infections/prevention & control , Haemophilus influenzae , Humans , Infant , Netherlands/epidemiology , Pandemics , SARS-CoV-2
17.
Mol Microbiol ; 111(4): 951-964, 2019 04.
Article in English | MEDLINE | ID: mdl-30600561

ABSTRACT

Biosynthesis of the nucleotide sugar precursor dTDP-L-rhamnose is critical for the viability and virulence of many human pathogenic bacteria, including Streptococcus pyogenes (Group A Streptococcus; GAS), Streptococcus mutans and Mycobacterium tuberculosis. Streptococcal pathogens require dTDP-L-rhamnose for the production of structurally similar rhamnose polysaccharides in their cell wall. Via heterologous expression in S. mutans, we confirmed that GAS RmlB and RmlC are critical for dTDP-L-rhamnose biosynthesis through their action as dTDP-glucose-4,6-dehydratase and dTDP-4-keto-6-deoxyglucose-3,5-epimerase enzymes respectively. Complementation with GAS RmlB and RmlC containing specific point mutations corroborated the conservation of previous identified catalytic residues. Bio-layer interferometry was used to identify and confirm inhibitory lead compounds that bind to GAS dTDP-rhamnose biosynthesis enzymes RmlB, RmlC and GacA. One of the identified compounds, Ri03, inhibited growth of GAS, other rhamnose-dependent streptococcal pathogens as well as M. tuberculosis with an IC50 of 120-410 µM. Importantly, we confirmed that Ri03 inhibited dTDP-L-rhamnose formation in a concentration-dependent manner through a biochemical assay with recombinant rhamnose biosynthesis enzymes. We therefore conclude that inhibitors of dTDP-L-rhamnose biosynthesis, such as Ri03, affect streptococcal and mycobacterial viability and can serve as lead compounds for the development of a new class of antibiotics that targets dTDP-rhamnose biosynthesis in pathogenic bacteria.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Hydro-Lyases/metabolism , Nucleoside Diphosphate Sugars/biosynthesis , Racemases and Epimerases/metabolism , Streptococcus/enzymology , Thymine Nucleotides/biosynthesis , Anti-Bacterial Agents/pharmacology , Biosynthetic Pathways , Hydro-Lyases/genetics , Inhibitory Concentration 50 , Racemases and Epimerases/genetics , Streptococcus/drug effects
18.
PLoS Pathog ; 14(10): e1007348, 2018 10.
Article in English | MEDLINE | ID: mdl-30321240

ABSTRACT

Human Group IIA secreted phospholipase A2 (hGIIA) is an acute phase protein with bactericidal activity against Gram-positive bacteria. Infection models in hGIIA transgenic mice have suggested the importance of hGIIA as an innate defense mechanism against the human pathogens Group A Streptococcus (GAS) and Group B Streptococcus (GBS). Compared to other Gram-positive bacteria, GAS is remarkably resistant to hGIIA activity. To identify GAS resistance mechanisms, we exposed a highly saturated GAS M1 transposon library to recombinant hGIIA and compared relative mutant abundance with library input through transposon-sequencing (Tn-seq). Based on transposon prevalence in the output library, we identified nine genes, including dltA and lytR, conferring increased hGIIA susceptibility. In addition, seven genes conferred increased hGIIA resistance, which included two genes, gacH and gacI that are located within the Group A Carbohydrate (GAC) gene cluster. Using GAS 5448 wild-type and the isogenic gacI mutant and gacI-complemented strains, we demonstrate that loss of the GAC N-acetylglucosamine (GlcNAc) side chain in the ΔgacI mutant increases hGIIA resistance approximately 10-fold, a phenotype that is conserved across different GAS serotypes. Increased resistance is associated with delayed penetration of hGIIA through the cell wall. Correspondingly, loss of the Lancefield Group B Carbohydrate (GBC) rendered GBS significantly more resistant to hGIIA-mediated killing. This suggests that the streptococcal Lancefield antigens, which are critical determinants for streptococcal physiology and virulence, are required for the bactericidal enzyme hGIIA to exert its bactericidal function.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Wall/metabolism , Group II Phospholipases A2/immunology , Immunity, Innate/drug effects , Polysaccharides, Bacterial/pharmacology , Streptococcal Infections/microbiology , Streptococcus/immunology , Blood Bactericidal Activity , Group II Phospholipases A2/blood , Group II Phospholipases A2/genetics , Host-Pathogen Interactions , Humans , Streptococcal Infections/blood , Streptococcal Infections/enzymology , Streptococcus/pathogenicity
19.
Cell Microbiol ; 21(10): e13072, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31219660

ABSTRACT

Staphylococcus aureus is a common skin commensal but is also associated with various skin and soft tissue pathologies. Upon invasion, S. aureus is detected by resident innate immune cells through pattern-recognition receptors (PRRs), although a comprehensive understanding of the specific molecular interactions is lacking. Recently, we demonstrated that the PRR langerin (CD207) on epidermal Langerhans cells senses the conserved ß-1,4-linked N-acetylglucosamine (GlcNAc) modification on S. aureus wall teichoic acid (WTA), thereby increasing skin inflammation. Interestingly, the S. aureus ST395 lineage as well as certain species of coagulase-negative staphylococci (CoNS) produce a structurally different WTA molecule, consisting of poly-glycerolphosphate with α-O-N-acetylgalactosamine (GalNAc) residues, which are attached by the glycosyltransferase TagN. Here, we demonstrate that S. aureus ST395 strains interact with the human Macrophage galactose-type lectin (MGL; CD301) receptor, which is expressed by dendritic cells and macrophages in the dermis. MGL bound S. aureus ST395 in a tagN- and GalNAc-dependent manner but did not interact with different tagN-positive CoNS species. However, heterologous expression of Staphylococcus lugdunensis tagN in S. aureus conferred phage infection and MGL binding, confirming the role of this CoNS enzyme as GalNAc-transferase. Functionally, the detection of GalNAc on S. aureus ST395 WTA by human monocyte-derived dendritic cells significantly enhanced cytokine production. Together, our findings highlight differential recognition of S. aureus glycoprofiles by specific human innate receptors, which may affect downstream adaptive immune responses and pathogen clearance.


Subject(s)
Cell Wall/metabolism , Dendritic Cells/immunology , Glycosyltransferases/metabolism , Lectins, C-Type/immunology , Staphylococcus aureus/enzymology , Teichoic Acids/chemistry , Acetylgalactosamine/analogs & derivatives , Acetylgalactosamine/chemistry , Cytokines/metabolism , Dermis/immunology , Dermis/microbiology , Glycerophosphates/chemistry , Glycosyltransferases/genetics , Host-Pathogen Interactions , Humans , Macrophages/immunology , Mutation , Staphylococcus aureus/chemistry , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Staphylococcus lugdunensis/chemistry , Staphylococcus lugdunensis/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL