Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
mBio ; 15(8): e0110724, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39041817

ABSTRACT

Neisserial adhesin A (NadA) is a meningococcal surface protein included as recombinant antigen in 4CMenB, a protein-based vaccine able to induce protective immune responses against Neisseria meningitidis serogroup B (MenB). Although NadA is involved in the adhesion/invasion of epithelial cells and human myeloid cells, its function in meningococcal physiology is still poorly understood. To clarify the role played by NadA in the host-pathogen interaction, we sought to identify its cellular receptors. We screened a protein microarray encompassing 2,846 human and 297 mouse surface/secreted recombinant proteins using recombinant NadA as probe. Efficient NadA binding was revealed on the paired sialic acid-binding immunoglobulin-type lectins receptors 5 and 14 (Siglec-5 and Siglec-14), but not on Siglec-9 therein used as control. The interaction was confirmed by biochemical tools with the determination of the KD value in the order of nanomolar and the identification of the NadA binding site by hydrogen-deuterium exchange coupled to mass spectrometry. The N-terminal domain of the Siglec-5 that recognizes the sialic acid was identified as the NadA binding domain. Intriguingly, exogenously added recombinant soluble Siglecs, including Siglec-9, were found to decorate N. meningitidis surface in a NadA-dependent manner. However, Siglec-5 and Siglec-14 transiently expressed in CHO-K1 cells endorsed NadA binding and increased N. meningitidis adhesion/invasion while Siglec-9 did not. Taken together, Siglec-5 and Siglec-14 satisfy all features of NadA receptors suggesting a possible role of NadA in the acute meningococcal infection.IMPORTANCEBacteria have developed several strategies for cell colonization and immune evasion. Knowledge of the host and pathogen factors involved in these mechanisms is crucial to build efficacious countermoves. Neisserial adhesin A (NadA) is a meningococcal surface protein included in the anti-meningococcus B vaccine 4CMenB, which mediates adhesion to and invasion of epithelial cells. Although NadA has been shown to bind to other cell types, like myeloid and endothelial cells, it still remains orphan of a defined host receptor. We have identified two strong NadA interactors, Siglec-5 and Siglec-14, which are mainly expressed on myeloid cells. This showcases that NadA is an additional and key player among the Neisseria meningitidis factors targeting immune cells. We thus provide novel insights on the strategies exploited by N. meningitidis during the infection process, which can progress to a severe illness and death.


Subject(s)
Adhesins, Bacterial , Antigens, CD , Antigens, Differentiation, Myelomonocytic , Bacterial Adhesion , Host-Pathogen Interactions , Lectins , Humans , Adhesins, Bacterial/metabolism , Adhesins, Bacterial/genetics , Antigens, CD/metabolism , Antigens, CD/genetics , Lectins/metabolism , Lectins/genetics , Lectins/immunology , Animals , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Protein Binding , Mice , CHO Cells , Cricetulus , Neisseria meningitidis/genetics , Neisseria meningitidis/metabolism , Neisseria meningitidis/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Epithelial Cells/immunology , Meningococcal Infections/microbiology , Meningococcal Infections/immunology , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , Neisseria meningitidis, Serogroup B/genetics , Neisseria meningitidis, Serogroup B/immunology , Neisseria meningitidis, Serogroup B/metabolism
2.
PLoS One ; 15(11): e0241793, 2020.
Article in English | MEDLINE | ID: mdl-33176334

ABSTRACT

BACKGROUND: Typing of Neisseria meningitidis isolates is crucial for the surveillance of invasive meningococcal disease (IMD). We performed a molecular epidemiology study of N. meningitidis serogroup B (MenB) causing IMD in Italy between 2014 and 2017 to describe circulating strains belonging to this serogroup, with particular regards to the two factor H-binding protein (FHbp) subfamilies present in the bivalent MenB vaccine. MATERIALS AND METHODS: A total of 109 culture positive and 46 culture negative MenB samples were collected within the National Surveillance System (NSS) of IMD in Italy and molecularly analyzed by conventional methods. RESULTS: Overall, 71 MenB samples showed the FHbp subfamily A and 83 the subfamily B. The subfamily variants were differently distributed by age. The most frequent variants, A05 and B231, were associated with cc213 and cc162, respectively. All MenB with the FHbp A05 variant displayed the PorA P1.22,14 and 85.7% of them the FetA F5-5. The majority of MenB with the FHbp B231 variant showed the PorA P1.22,14 (65.4%) and 84.6%, the FetA F3-6. CONCLUSION: MenB circulating in Italy were characterized by a remarkable association between clonal complex and FHbp variants, although a high degree of genetic diversity observed over time. A dynamic trend in clonal complexes distribution within MenB was detected. Our results stress the importance of continued meningococcal molecular surveillance to evaluate the potential vaccine coverage of the available MenB vaccines.


Subject(s)
Neisseria meningitidis, Serogroup B/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Vaccines/immunology , Genetic Variation , Meningococcal Vaccines/immunology , Meningococcal Vaccines/metabolism , Multilocus Sequence Typing , Neisseria meningitidis, Serogroup B/metabolism , Porins/immunology , Porins/metabolism , Software , Whole Genome Sequencing
3.
Biosci Rep ; 40(3)2020 03 27.
Article in English | MEDLINE | ID: mdl-32159209

ABSTRACT

BACKGROUND: The C5 complement inhibitor eculizumab is first-line treatment in atypical hemolytic uremic syndrome (aHUS) going along with a highly increased risk of meningococcal infections. Serogroup B meningococci (MenB) are the most frequently encountered cause for meningococcal infections in Europe. Efficacy of the protein-based MenB-vaccine Bexsero in aHUS has not been determined and testing is only possible in patients off-treatment with eculizumab as a human complement source is required. METHODS: Patients with aHUS were vaccinated with two doses of the protein-based MenB-vaccine Bexsero. Serum bactericidal antibody (SBA) titers against factor H binding protein (fHbp) of MenB were determined in 14 patients with aHUS off-treatment with eculizumab. RESULTS: Only 50% of patients showed protective human serum bactericidal antibody (hSBA) titers (≥1:4) against MenB following two vaccinations. Bactericidal antibody titers were relatively low (≤1:8) in three of seven patients with protective titers. While 71% of patients were on immunosuppressive treatment for either thrombotic microangiopathy or renal transplantation at either first or second vaccination, all four patients not receiving any immunosuppressive treatment showed protective bactericidal antibody response. Time between second vaccination and titer measurement was not significantly different between patients with protective titers compared with those with non-protective titers, while time between first and second vaccination was significantly longer in patients with protective titers going along with a tendency for reduction in immunosuppressive treatment. CONCLUSIONS: Efficacy of vaccination against MenB is insufficient in patients with aHUS. Response to vaccination seems to be hampered by immunosuppression. Therefore, implementation of adequate antibiotic prophylaxis seems pivotal.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Meningococcal Vaccines/pharmacology , Neisseria meningitidis, Serogroup B/immunology , Adult , Antibodies, Monoclonal, Humanized/pharmacology , Atypical Hemolytic Uremic Syndrome/complications , Atypical Hemolytic Uremic Syndrome/microbiology , Bacterial Proteins/immunology , Carrier Proteins , Complement Factor H/immunology , Female , Germany , Humans , Male , Meningococcal Infections/prevention & control , Middle Aged , Neisseria meningitidis, Serogroup B/metabolism , Serogroup , Treatment Outcome , Vaccination/methods
4.
J Biol Chem ; 293(16): 6000-6010, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29483188

ABSTRACT

Lytic transglycosylases (LTs) are a class of enzymes important for the recycling and metabolism of peptidoglycan (PG). LTs cleave the ß-1,4-glycosidic bond between N-acetylmuramic acid (MurNAc) and GlcNAc in the PG glycan strand, resulting in the concomitant formation of 1,6-anhydro-N-acetylmuramic acid and GlcNAc. No LTs reported to date have utilized chitins as substrates, despite the fact that chitins are GlcNAc polymers linked via ß-1,4-glycosidic bonds, which are the known site of chemical activity for LTs. Here, we demonstrate enzymatically that LtgA, a non-canonical, substrate-permissive LT from Neisseria meningitidis utilizes chitopentaose ((GlcNAc)5) as a substrate to produce three newly identified sugars: 1,6-anhydro-chitobiose, 1,6-anhydro-chitotriose, and 1,6-anhydro-chitotetraose. Although LTs have been widely studied, their complex reactions have not previously been visualized in the crystalline state because macromolecular PG is insoluble. Here, we visualized the cleavage of the glycosidic bond and the liberation of GlcNAc-derived residues by LtgA, followed by the synthesis of atypical 1,6-anhydro-GlcNAc derivatives. In addition to the newly identified anhydro-chitin products, we identified trapped intermediates, unpredicted substrate rearrangements, sugar distortions, and a conserved crystallographic water molecule bound to the catalytic glutamate of a high-resolution native LT. This study enabled us to propose a revised alternative mechanism for LtgA that could also be applicable to other LTs. Our work contributes to the understanding of the mechanisms of LTs in bacterial cell wall biology.


Subject(s)
Glycosyltransferases/metabolism , Neisseria meningitidis, Serogroup B/enzymology , Peptidoglycan/metabolism , Chitinases/chemistry , Chitinases/metabolism , Crystallography, X-Ray , Glycosides/chemistry , Glycosides/metabolism , Glycosyltransferases/chemistry , Meningitis, Meningococcal/microbiology , Models, Molecular , Muramic Acids/chemistry , Muramic Acids/metabolism , Neisseria meningitidis, Serogroup B/chemistry , Neisseria meningitidis, Serogroup B/metabolism , Peptidoglycan/chemistry , Protein Conformation
5.
Sci Rep ; 7(1): 5693, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28720847

ABSTRACT

Neisseria meningitidis express numerous virulence factors that enable it to interact with diverse microenvironments within the host, during both asymptomatic nasopharyngeal colonization and invasive disease. Many of these interactions involve bacterial or host glycans. In order to characterise the meningococcal glycointeractome, glycan arrays representative of structures found on human cells, were used as a screening tool to investigate host glycans bound by N. meningitidis. Arrays probed with fluorescently labelled wild-type MC58 revealed binding to 223 glycans, including blood group antigens, mucins, gangliosides and glycosaminoglycans. Mutant strains lacking surface components, including capsule, lipooligosaccharide (LOS), Opc and pili, were investigated to identify the factors responsible for glycan binding. Surface plasmon resonance and isothermal calorimetry were used to confirm binding and determine affinities between surface components and host glycans. We observed that the L3 LOS immunotype (whole cells and purified LOS) bound 26 structures, while L8 only bound 5 structures. We further demonstrated a direct glycan-glycan interaction between purified L3 LOS and Thomsen-Friedenreich (TF) antigen, with a KD of 13 nM. This is the highest affinity glycan-glycan interaction reported to date. These findings highlight the diverse glycointeractions that may occur during different stages of meningococcal disease, which could be exploited for development of novel preventative and therapeutic strategies.


Subject(s)
Neisseria meningitidis, Serogroup B/metabolism , Polysaccharides/chemistry , Polysaccharides/metabolism , Calorimetry/methods , Gene Knockout Techniques , Glycomics , Host-Pathogen Interactions , Humans , Meningococcal Infections/metabolism , Mutation , Neisseria meningitidis, Serogroup B/chemistry , Neisseria meningitidis, Serogroup B/genetics , Neisseria meningitidis, Serogroup B/pathogenicity , Surface Plasmon Resonance , Virulence Factors
6.
Mol Microbiol ; 105(6): 934-953, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28708335

ABSTRACT

PorB is a well-characterized outer membrane protein that is common among Neisseria species and is required for survival. A vaccine candidate, PorB induces antibody responses that are directed against six variable surface-exposed loops that differ in sequence depending on serotype. Although Neisseria meningitidis is naturally competent and porB genetic mosaicism provides evidence for strong positive selection, the sequences of PorB serotypes commonly associated with invasive disease are often conserved, calling into question the interaction of specific PorB loop sequences in immune engagement. In this report, we provide evidence that antibody binding to a PorB epitope can be altered by sequence mutations in non-epitope loops. Through the construction of hybrid PorB types and PorB molecular dynamics simulations, we demonstrate that loops both adjacent and non-adjacent to the epitope loop can enhance or diminish antibody binding, a phenotype that correlates with serum bactericidal activity. We further examine the interaction of PorB with outer membrane-associated proteins, including PorA and RmpM. Deletion of these proteins alters the composition of PorB-containing native complexes and reduces antibody binding and serum killing relative to the parental strain, suggesting that both intramolecular and intermolecular PorB interactions contribute to host adaptive immune evasion.


Subject(s)
Neisseria meningitidis, Serogroup B/metabolism , Neisseria meningitidis/metabolism , Porins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/metabolism , Epitopes/metabolism , Genetic Heterogeneity , Neisseria meningitidis/genetics , Neisseria meningitidis, Serogroup B/genetics , Porins/genetics , Protein Binding , Serogroup , Signal Transduction
7.
Proc Natl Acad Sci U S A ; 113(10): 2714-9, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26888286

ABSTRACT

Factor H binding protein (fHbp) is a lipoprotein of Neisseria meningitidis important for the survival of the bacterium in human blood and a component of two recently licensed vaccines against serogroup B meningococcus (MenB). Based on 866 different amino acid sequences this protein is divided into three variants or two families. Quantification of the protein is done by immunoassays such as ELISA or FACS that are susceptible to the sequence variation and expression level of the protein. Here, selected reaction monitoring mass spectrometry was used for the absolute quantification of fHbp in a large panel of strains representative of the population diversity of MenB. The analysis revealed that the level of fHbp expression can vary at least 15-fold and that variant 1 strains express significantly more protein than variant 2 or variant 3 strains. The susceptibility to complement-mediated killing correlated with the amount of protein expressed by the different meningococcal strains and this could be predicted from the nucleotide sequence of the promoter region. Finally, the absolute quantification allowed the calculation of the number of fHbp molecules per cell and to propose a mechanistic model of the engagement of C1q, the recognition component of the complement cascade.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Neisseria meningitidis, Serogroup B/metabolism , Amino Acid Sequence , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Genetic Variation , Humans , Mass Spectrometry/methods , Meningitis, Meningococcal/immunology , Meningitis, Meningococcal/microbiology , Meningococcal Vaccines/immunology , Neisseria meningitidis, Serogroup B/classification , Neisseria meningitidis, Serogroup B/genetics , Phylogeny , Species Specificity
8.
J Biol Chem ; 291(7): 3224-38, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26655715

ABSTRACT

The degree of phosphorylation and phosphoethanolaminylation of lipid A on neisserial lipooligosaccharide (LOS), a major cell-surface antigen, can be correlated with inflammatory potential and the ability to induce immune tolerance in vitro. On the oligosaccharide of the LOS, the presence of phosphoethanolamine and sialic acid substituents can be correlated with in vitro serum resistance. In this study, we analyzed the structure of the LOS from 40 invasive isolates and 25 isolates from carriers of Neisseria meningitidis without disease. Invasive strains were classified as groups 1-3 that caused meningitis, septicemia without meningitis, and septicemia with meningitis, respectively. Intact LOS was analyzed by high resolution matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Prominent peaks for lipid A fragment ions with three phosphates and one phosphoethanolamine were detected in all LOS analyzed. LOS from groups 2 and 3 had less abundant ions for highly phosphorylated lipid A forms and induced less TNF-α in THP-1 monocytic cells compared with LOS from group 1. Lipid A from all invasive strains was hexaacylated, whereas lipid A of 6/25 carrier strains was pentaacylated. There were fewer O-acetyl groups and more phosphoethanolamine and sialic acid substitutions on the oligosaccharide from invasive compared with carrier isolates. Bioinformatic and genomic analysis of LOS biosynthetic genes indicated significant skewing to specific alleles, dependent on the disease outcome. Our results suggest that variable LOS structures have multifaceted effects on homeostatic innate immune responses that have critical impact on the pathophysiology of meningococcal infections.


Subject(s)
Antigens, Bacterial/toxicity , Carrier State/microbiology , Lipopolysaccharides/toxicity , Meningitis, Meningococcal/microbiology , Meningococcal Infections/microbiology , Neisseria meningitidis, Serogroup B/pathogenicity , Neisseria meningitidis, Serogroup C/pathogenicity , Acylation , Adolescent , Antigens, Bacterial/biosynthesis , Antigens, Bacterial/chemistry , Carrier State/blood , Carrier State/cerebrospinal fluid , Carrier State/immunology , Cell Line, Tumor , Computational Biology , Gene Expression Profiling , Humans , Immunity, Innate/drug effects , Lipopolysaccharides/biosynthesis , Lipopolysaccharides/chemistry , Meningitis, Meningococcal/blood , Meningitis, Meningococcal/cerebrospinal fluid , Meningitis, Meningococcal/immunology , Meningococcal Infections/blood , Meningococcal Infections/cerebrospinal fluid , Meningococcal Infections/immunology , Molecular Structure , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Neisseria meningitidis, Serogroup B/classification , Neisseria meningitidis, Serogroup B/immunology , Neisseria meningitidis, Serogroup B/metabolism , Neisseria meningitidis, Serogroup C/classification , Neisseria meningitidis, Serogroup C/immunology , Neisseria meningitidis, Serogroup C/metabolism , Norway , Phosphorylation , Sepsis/blood , Sepsis/cerebrospinal fluid , Sepsis/immunology , Sepsis/microbiology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Necrosis Factor-alpha/metabolism , Virulence
11.
J Biol Chem ; 289(22): 15602-10, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24755216

ABSTRACT

GNA2091 of Neisseria meningitidis is a lipoprotein of unknown function that is included in the novel 4CMenB vaccine. Here, we investigated the biological function and the subcellular localization of the protein. We demonstrate that GNA2091 functions in the assembly of outer membrane proteins (OMPs) because its absence resulted in the accumulation of misassembled OMPs. Cell fractionation and protease accessibility experiments showed that the protein is localized at the periplasmic side of the outer membrane. Pulldown experiments revealed that it is not stably associated with the ß-barrel assembly machinery, the previously identified complex for OMP assembly. Thus, GNA2091 constitutes a novel outer membrane-based lipoprotein required for OMP assembly. Furthermore, its location at the inner side of the outer membrane indicates that protective immunity elicited by this antigen cannot be due to bactericidal or opsonic activity of antibodies.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Vaccines/metabolism , Neisseria meningitidis, Serogroup B/metabolism , Amino Acid Sequence , Bacterial Outer Membrane Proteins/chemistry , Conserved Sequence , Gene Deletion , Lipoproteins/genetics , Lipoproteins/metabolism , Molecular Sequence Data , Mutagenesis , Neisseria meningitidis, Serogroup B/genetics , Phenotype , Porins/metabolism , Protein Structure, Tertiary
12.
J Proteomics ; 101: 63-76, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24561796

ABSTRACT

Meningococcal surface proteins capable of evoking a protective immune response are candidates for inclusion in protein-based vaccines against serogroup B Neisseria meningitidis (NmB). In this study, a 2-dimensional (2-D) gel-based platform integrating surface and immune-proteomics was developed to characterize NmB surface protein antigens. The surface proteome was analyzed by differential 2-D gel electrophoresis following treatment of live bacteria with proteinase K. Alongside, proteins recognized by immune sera from mice challenged with live meningococci were detected using 2-D immunoblots. In combination, seventeen proteins were identified including the well documented antigens PorA, OpcA and factor H-binding protein, previously reported potential antigens and novel potential immunogens. Results were validated for the macrophage infectivity potentiator (MIP), a recently proposed NmB vaccine candidate. MIP-specific antisera bound to meningococci in whole-cell ELISA and facilitated opsonophagocytosis and deposition of complement factors on the surface of meningococcal isolates of different serosubtypes. Cleavage by proteinase K was confirmed in western blots and shown to occur in a fraction of the MIP expressed by meningococci suggesting transient or limited surface exposure. These observations add knowledge for the development of a protein NmB vaccine. The proteomic workflow presented here may be used for the discovery of vaccine candidates against other pathogens. BIOLOGICAL SIGNIFICANCE: This study presents an integrated proteomic strategy to identify proteins from N. meningitidis with desirable properties (i.e. surface exposure and immunogenicity) for inclusion in subunit vaccines against bacterial meningitis. The effectiveness of the method was demonstrated by the identification of some of the major meningococcal vaccine antigens. Information was also obtained about novel potential immunogens as well as the recently described potential antigen macrophage infectivity potentiator which can be useful for its consideration as a vaccine candidate. Additionally, the proteomic strategy presented in this study provides a generic 2-D gel-based platform for the discovery of vaccine candidates against other bacterial infections.


Subject(s)
Antigens, Bacterial/metabolism , Antigens, Surface/metabolism , Meningitis, Meningococcal/immunology , Meningococcal Vaccines/metabolism , Neisseria meningitidis, Serogroup B/chemistry , Neisseria meningitidis, Serogroup B/immunology , Proteomics/methods , Animals , Antigens, Bacterial/isolation & purification , Antigens, Surface/analysis , Antigens, Surface/isolation & purification , Bacterial Proteins/immunology , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Endopeptidase K/pharmacology , Female , Meningococcal Vaccines/isolation & purification , Mice , Mice, Inbred BALB C , Neisseria meningitidis, Serogroup B/metabolism
13.
PLoS One ; 8(1): e54314, 2013.
Article in English | MEDLINE | ID: mdl-23372704

ABSTRACT

Outer membrane vesicles (OMV) contain immunogenic proteins and contribute to in vivo survival and virulence of bacterial pathogens. The first OMV vaccines successfully stopped Neisseria meningitidis serogroup B outbreaks but required detergent-extraction for endotoxin removal. Current vaccines use attenuated endotoxin, to preserve immunological properties and allow a detergent-free process. The preferred process is based on spontaneously released OMV (sOMV), which are most similar to in vivo vesicles and easier to purify. The release mechanism however is poorly understood resulting in low yield. This study with N. meningitidis demonstrates that an external stimulus, cysteine depletion, can trigger growth arrest and sOMV release in sufficient quantities for vaccine production (±1500 human doses per liter cultivation). Transcriptome analysis suggests that cysteine depletion impairs iron-sulfur protein assembly and causes oxidative stress. Involvement of oxidative stress is confirmed by showing that addition of reactive oxygen species during cysteine-rich growth also triggers vesiculation. The sOMV in this study are similar to vesicles from natural infection, therefore cysteine-dependent vesiculation is likely to be relevant for the in vivo pathogenesis of N. meningitidis.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Cell Membrane/immunology , Cysteine/deficiency , Meningococcal Infections/prevention & control , Meningococcal Vaccines/isolation & purification , Neisseria meningitidis, Serogroup B/immunology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bioreactors , Cell Membrane/chemistry , Culture Media , Humans , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/immunology , Meningococcal Infections/immunology , Meningococcal Vaccines/chemistry , Meningococcal Vaccines/immunology , Neisseria meningitidis, Serogroup B/chemistry , Neisseria meningitidis, Serogroup B/metabolism , Oxidative Stress , Proteome/genetics , Proteome/immunology
14.
Vaccine ; 30(42): 6064-9, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22867717

ABSTRACT

Serogroup B outer membrane vesicles (OMV) with iron regulated proteins (IRP) from Neisseria meningitidis constitute the antigen for the vaccine against the disease caused by this bacterium. Aiming to enhance final OMV concentration, seven batch experiments were carried out under four different conditions: (i) with original Catlin medium; (ii) with original Catlin medium and lactate and amino acids pulse at the 6th cultivation hour; (iii) with Catlin medium with double initial concentrations of lactate and amino acids and (iv) Catlin medium without glycerol and with double initial concentrations of lactate and amino acids. The cultivation experiments were carried out in a 7-L bioreactor under the following conditions: 36°C, 0.5atm, overlay air 1L/min, agitation: 250-850 rpm, and O(2) control at 10%, 20 h. After lactate and amino acids exhaustion, cell growth reached stationary phase and a significant release increase of OMV was observed. According to the Luedeking & Piret model, OMV liberation is non-growth associated. Glycerol was not consumed during cultivation. The maximum OMV concentration value attained was 162 mg/L with correspondent productivity of 8.1mg/(Lh) employing Catlin medium with double initial concentrations of lactate and amino acids. The obtained OMV satisfied constitution and protein pattern criteria and were suitable for vaccine production.


Subject(s)
Bacterial Outer Membrane Proteins/biosynthesis , Bioreactors , Neisseria meningitidis, Serogroup B/metabolism , Secretory Vesicles/chemistry , Amino Acids/chemistry , Batch Cell Culture Techniques , Culture Media/chemistry , Glycerol/metabolism , Iron-Regulatory Proteins/chemistry , Lactic Acid/chemistry , Meningococcal Vaccines/biosynthesis
15.
Microbes Infect ; 14(11): 979-88, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22565133

ABSTRACT

Neisseria meningitidis may be classified according to the lipooligosaccharide immunotype. We show that this classification can be achieved by PCR genotyping of the genes involved in the lipooligosaccharide inner-core biosynthesis, lpt3, lpt6, lgtG and lot3. Genotyping data correlated well (90-100%) with mass spectrometry data and was, therefore, applied to screen a random subset of recent N. meningitidis serogroup B isolates from Europe. Analysis of the proportion of the different lipooligosaccharide types highlighted the predominance of L3 strains. Surprisingly, high rates of L2 type strains were found in Spain (17%, versus 2.5% in Germany and 1.9% in the United Kingdom). Therefore, we also investigated further these Spanish L2 strains in an attempt to explain such prevalence despite the known sensitivity of L2 immunotype to complement. We explored the hypothesis that these strains express high amounts of factor H-binding protein (fHbp), but we found, on the contrary, that L2 strains express low or undetectable amounts of fHbp. Our findings suggest that, in addition to a genetic analysis, a multivalent approach may be necessary to estimate the effectiveness of a N. meningitidis serogroup B vaccine.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Lipopolysaccharides/chemistry , Meningitis, Meningococcal/microbiology , Neisseria meningitidis, Serogroup B/genetics , Antigens, Bacterial/analysis , Bacterial Proteins/analysis , Bacterial Proteins/genetics , Genotyping Techniques , Humans , Lipopolysaccharides/metabolism , Mass Spectrometry , Neisseria meningitidis, Serogroup B/chemistry , Neisseria meningitidis, Serogroup B/metabolism , Polymerase Chain Reaction , Prevalence , Reproducibility of Results , Spain
16.
Innate Immun ; 18(4): 580-91, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22180561

ABSTRACT

Neisseria meningitidis causes sepsis with coagulopathy. The present study evaluated the tissue factor (TF)-inducing capacity of bacterial LPS in different presentation forms, i.e. membrane-bound LPS versus purified LPS, and of non-LPS components of N. meningitidis. By using a wild-type N. meningitidis, a mutant N. meningitidis lacking LPS (LPS-deficient N. meningitidis), purified LPS from N. meningitidis and Escherichia coli, we measured TF-expression and TF-activity on human monocytes and microparticles (MPs). The effect of TF-modulators, such as phosphatidylserine (PS), tissue factor pathway inhibitor (TFPI) and recombinant IL-10 (rhIL-10) was investigated. In plasmas from meningococcal patients, fibrinopeptide A (FPA), LPS and IL-10 were quantified. Monocytes and MPs exposed to purified LPS or wild-type N. meningitidis had much higher TF-activity than monocytes and MPs exposed to LPS-deficient N. meningitidis (clot formation assay). Incubation with wild-type N. meningitidis, but also LPS-deficient N. meningitidis, resulted in TF-expression on monocytes (flow cytometry, qRT-PCR). Increased cellular TF-activity is associated with coincident surface-exposure of PS and the number of monocytes positive for both PS and TF was significantly higher for monocytes exposed to wild-type N. meningitidis (7.6%) compared with monocytes exposed to LPS-deficient N. meningitidis (1.8%). Treatment with rhIL-10 reduced monocyte- and MP-associated TF-activity, the number of monocytes positive for both TF and PS, and microvesiculation. Patients with meningococcal septicemia had significantly higher levels of LPS, FPA and IL-10 than patients with distinct meningitis. Our results indicate that LPS from N. meningitidis is crucial for inducing TF-activity, but not for monocyte- and MP-associated TF-expression. TF-activity seems to require coincident expression of TF and PS on monocytes, and LPS induces such double-positive monocytes.


Subject(s)
Cell-Derived Microparticles/immunology , Lipopolysaccharides/immunology , Meningitis, Meningococcal/immunology , Meningococcal Infections/immunology , Monocytes/immunology , Neisseria meningitidis, Serogroup B/immunology , Thromboplastin/metabolism , Blood Coagulation/drug effects , Blood Coagulation/immunology , Cell-Derived Microparticles/drug effects , Cells, Cultured , Escherichia coli/immunology , Escherichia coli Infections/immunology , Fibrinopeptide A/metabolism , Gene Expression Regulation, Bacterial/drug effects , Humans , Interleukin-10/pharmacology , Lipoproteins/pharmacology , Meningitis, Meningococcal/blood , Meningitis, Meningococcal/microbiology , Meningococcal Infections/blood , Meningococcal Infections/microbiology , Monocytes/drug effects , Neisseria meningitidis, Serogroup B/metabolism , Phosphatidylserines/pharmacology , Thromboplastin/genetics
17.
J Bacteriol ; 192(20): 5363-77, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20709895

ABSTRACT

Neisseria meningitidis serogroup B strains are responsible for most meningococcal cases in the industrialized countries, and strains belonging to the clonal complex ST-41/44 are among the most prevalent serogroup B strains in carriage and disease. Here, we report the first genome and transcriptome comparison of a serogroup B carriage strain from the clonal complex ST-41/44 to the serogroup B disease strain MC58 from the clonal complex ST-32. Both genomes are highly colinear, with only three major genome rearrangements that are associated with the integration of mobile genetic elements. They further differ in about 10% of their gene content, with the highest variability in gene presence as well as gene sequence found for proteins involved in host cell interactions, including Opc, NadA, TonB-dependent receptors, RTX toxin, and two-partner secretion system proteins. Whereas housekeeping genes coding for metabolic functions were highly conserved, there were considerable differences in their expression pattern upon adhesion to human nasopharyngeal cells between both strains, including differences in energy metabolism and stress response. In line with these genomic and transcriptomic differences, both strains also showed marked differences in their in vitro infectivity and in serum resistance. Taken together, these data support the concept of a polygenic nature of meningococcal virulence comprising differences in the repertoire of adhesins as well as in the regulation of metabolic genes and suggest a prominent role for immune selection and genetic drift in shaping the meningococcal genome.


Subject(s)
Meningococcal Infections/microbiology , Neisseria meningitidis, Serogroup B/genetics , Bacterial Adhesion/physiology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Cell Line , Gene Expression Profiling , Gene Expression Regulation, Bacterial/physiology , Genome, Bacterial , Genotype , Humans , Interspersed Repetitive Sequences/genetics , Neisseria meningitidis, Serogroup B/classification , Neisseria meningitidis, Serogroup B/metabolism , Neisseria meningitidis, Serogroup B/pathogenicity , Phylogeny , Virulence
18.
J Immunol ; 185(1): 507-16, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20530262

ABSTRACT

Properdin, a positive regulator of the alternative pathway (AP) of complement is important in innate immune defenses against invasive neisserial infections. Recently, commercially available unfractionated properdin was shown to bind to certain biological surfaces, including Neisseria gonorrhoeae, which facilitated C3 deposition. Unfractionated properdin contains aggregates or high-order oligomers, in addition to its physiological "native" (dimeric, trimeric, and tetrameric) forms. We examined the role of properdin in AP activation on diverse strains of Neisseria meningitidis and N. gonorrhoeae specifically using native versus unfractionated properdin. C3 deposition on Neisseria decreased markedly when properdin function was blocked using an anti-properdin mAb or when properdin was depleted from serum. Maximal AP-mediated C3 deposition on Neisseriae even at high (80%) serum concentrations required properdin. Consistent with prior observations, preincubation of bacteria with unfractionated properdin, followed by the addition of properdin-depleted serum resulted in higher C3 deposition than when bacteria were incubated with properdin-depleted serum alone. Unexpectedly, none of 10 Neisserial strains tested bound native properdin. Consistent with its inability to bind to Neisseriae, preincubating bacteria with native properdin followed by the addition of properdin-depleted serum did not cause detectable increases in C3 deposition. However, reconstituting properdin-depleted serum with native properdin a priori enhanced C3 deposition on all strains of Neisseria tested. In conclusion, the physiological forms of properdin do not bind directly to either N. meningitidis or N. gonorrhoeae but play a crucial role in augmenting AP-dependent C3 deposition on the bacteria through the "conventional" mechanism of stabilizing AP C3 convertases.


Subject(s)
Complement Pathway, Alternative/immunology , Neisseria gonorrhoeae/immunology , Neisseria meningitidis, Serogroup A/immunology , Neisseria meningitidis, Serogroup B/immunology , Neisseria meningitidis, Serogroup C/immunology , Neisseria meningitidis, Serogroup W-135/immunology , Neisseria meningitidis, Serogroup Y/immunology , Properdin/physiology , Bacterial Adhesion/immunology , Complement C3/metabolism , Complement C3 Convertase, Alternative Pathway/metabolism , Complement Pathway, Alternative/genetics , Enzyme Stability/immunology , Humans , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/metabolism , Neisseria meningitidis, Serogroup A/genetics , Neisseria meningitidis, Serogroup A/metabolism , Neisseria meningitidis, Serogroup B/genetics , Neisseria meningitidis, Serogroup B/metabolism , Neisseria meningitidis, Serogroup C/genetics , Neisseria meningitidis, Serogroup C/metabolism , Neisseria meningitidis, Serogroup W-135/genetics , Neisseria meningitidis, Serogroup W-135/metabolism , Neisseria meningitidis, Serogroup Y/genetics , Neisseria meningitidis, Serogroup Y/metabolism , Properdin/isolation & purification , Properdin/metabolism , Protein Binding/immunology
19.
PLoS Pathog ; 6(5): e1000911, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20502634

ABSTRACT

The host vasculature is believed to constitute the principal route of dissemination of Neisseria meningitidis (Nm) throughout the body, resulting in septicaemia and meningitis in susceptible humans. In vitro, the Nm outer membrane protein Opc can enhance cellular entry and exit, utilising serum factors to anchor to endothelial integrins; but the mechanisms of binding to serum factors are poorly characterised. This study demonstrates that Nm Opc expressed in acapsulate as well as capsulate bacteria can increase human brain endothelial cell line (HBMEC) adhesion and entry by first binding to serum vitronectin and, to a lesser extent, fibronectin. This study also demonstrates that Opc binds preferentially to the activated form of human vitronectin, but not to native vitronectin unless the latter is treated to relax its closed conformation. The direct binding of vitronectin occurs at its Connecting Region (CR) requiring sulphated tyrosines Y(56) and Y(59). Accordingly, Opc/vitronectin interaction could be inhibited with a conformation-dependent monoclonal antibody 8E6 that targets the sulphotyrosines, and with synthetic sulphated (but not phosphorylated or unmodified) peptides spanning the vitronectin residues 43-68. Most importantly, the 26-mer sulphated peptide bearing the cell-binding domain (45)RGD(47) was sufficient for efficient meningococcal invasion of HBMECs. To our knowledge, this is the first study describing the binding of a bacterial adhesin to sulphated tyrosines of the host receptor. Our data also show that a single region of Opc is likely to interact with the sulphated regions of both vitronectin and of heparin. As such, in the absence of heparin, Opc-expressing Nm interact directly at the CR but when precoated with heparin, they bind via heparin to the heparin-binding domain of the activated vitronectin, although with a lower affinity than at the CR. Such redundancy suggests the importance of Opc/vitronectin interaction in meningococcal pathogenesis and may enable the bacterium to harness the benefits of the physiological processes in which the host effector molecule participates.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Brain/cytology , Endothelial Cells/microbiology , Neisseria meningitidis, Serogroup A/metabolism , Neisseria meningitidis, Serogroup B/metabolism , Vitronectin/metabolism , Animals , Bacterial Adhesion/physiology , Bacterial Outer Membrane Proteins/genetics , Blood-Brain Barrier/cytology , Blood-Brain Barrier/microbiology , Cattle , Cell Line , Endothelial Cells/cytology , Fibronectins/metabolism , Heparin/chemistry , Heparin/metabolism , Humans , Mice , Neisseria meningitidis, Serogroup A/genetics , Neisseria meningitidis, Serogroup B/genetics , Phosphorylation/physiology , Protein Denaturation , Protein Structure, Tertiary , Species Specificity , Sulfates/metabolism , Tyrosine/metabolism , Vitronectin/chemistry
20.
FASEB J ; 24(1): 286-95, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19720623

ABSTRACT

S-nitrosylation is an important mediator of multiple nitric oxide-dependent biological processes, including eukaryotic cellular events such as macrophage apoptosis and proinflammatory signaling. Many pathogenic bacteria possess NO detoxification mechanisms, such as the nitric oxide reductase (NorB) of Neisseria meningitidis and the flavohemoglobins (Hmp) of Salmonella enterica and Escherichia coli, which serve to protect the microorganism from nitrosative stress within the intracellular environment. In this study, we demonstrate that expression of meningococcal NorB increases the rate at which low-molecular-weight S-nitrosothiol (SNO) decomposes in vitro. To determine whether this effect occurs in cells during infection by bacteria, we induced SNO formation in murine macrophages by activation with lipopolysaccharide and gamma-interferon and observed a reduced abundance of SNO during coincubation with N. meningitidis, S. enterica, or E. coli. In each case, this effect was shown to be dependent on bacterial NO detoxification genes, which act to prevent SNO formation through the removal of NO. This may represent a novel mechanism of host cell injury by bacteria.


Subject(s)
Bacteria/metabolism , Bacteria/pathogenicity , Nitric Oxide/metabolism , S-Nitrosothiols/metabolism , Animals , Cell Line , Escherichia coli K12/metabolism , Host-Pathogen Interactions/physiology , Inactivation, Metabolic , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/microbiology , Mice , Neisseria meningitidis, Serogroup B/genetics , Neisseria meningitidis, Serogroup B/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Recombinant Proteins , Salmonella typhimurium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL