Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Cell Microbiol ; 16(6): 925-37, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24397470

ABSTRACT

Neisseria meningitidis is a human pathogen that can cause fatal sepsis and meningitis once it reaches the blood stream and the nervous system. Here we demonstrate that a fragment, released upon proteolysis of the surface-exposed protein Neisserial Heparin Binding Antigen (NHBA), by the bacterial protease NalP, alters the endothelial permeability by inducing the internalization of the adherens junction protein VE-cadherin. We found that C2 rapidly accumulates in mitochondria where it induces the production of reactive oxygen species: the latter are required for the phosphorylation of the junctional protein and for its internalization that, in turn, is responsible for the endothelial leakage. Our data support the notion that the NHBA-derived fragment C2 might contribute to the extensive vascular leakage typically associated with meningococcal sepsis.


Subject(s)
Adherens Junctions/drug effects , Antigens, Bacterial/metabolism , Endothelial Cells/drug effects , Neisseria meningitidis/physiology , Permeability/drug effects , Antigens, Bacterial/genetics , Cell Line , Humans , Virulence Factors/genetics , Virulence Factors/metabolism
2.
Mol Microbiol ; 83(5): 1035-47, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22320452

ABSTRACT

Gram-positive pili are known to play a role in bacterial adhesion to epithelial cells and in the formation of biofilm microbial communities. In the present study we undertook the functional characterization of the pilus ancillary protein 1 (AP1_M6) from Streptococcus pyogenes isolates expressing the FCT-1 pilus variant, known to be strong biofilm formers. Cell binding and biofilm formation assays using S. pyogenes in-frame deletion mutants, Lactococcus expressing heterologous FCT-1 pili and purified recombinant AP1_M6, indicated that this pilin is a strong cell adhesin that is also involved in bacterial biofilm formation. Moreover, we show that AP1_M6 establishes homophilic interactions that mediate inter-bacterial contact, possibly promoting bacterial colonization of target epithelial cells in the form of three-dimensional microcolonies. Finally, AP1_M6 knockout mutants were less virulent in mice, indicating that this protein is also implicated in GAS systemic infection.


Subject(s)
Bacterial Adhesion , Biofilms , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Streptococcus pyogenes/pathogenicity , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Animals , Epithelial Cells/microbiology , Fimbriae Proteins/genetics , Mice , Sequence Deletion , Streptococcal Infections/microbiology , Streptococcus pyogenes/genetics , Streptococcus pyogenes/metabolism
3.
Cell Microbiol ; 14(3): 368-85, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22066472

ABSTRACT

NadA (N eisseria meningitidisadhesin A), a meningococcal surface protein, mediates adhesion to and invasion of human cells, an activity in which host membrane proteins have been implicated. While investigating these host factors in human epithelial cells by affinity chromatography, we discovered an unanticipated interaction of NadA with heat shock protein (Hsp) 90, a molecular chaperone. The specific in vitro interaction of recombinant soluble NadA and Hsp90 was confirmed by co-immunoprecipitations, dot and far-Western blot. Intriguingly, ADP, but not ATP, was required for this association, and the Hsp90 inhibitor 17-AAG promoted complex formation. Hsp90 binding to an Escherichia coli strain used as carrier to express surface exposed NadA confirmed these results in live bacteria. We also examined RNA interference, plasmid-driven overexpression, addition of exogenous rHsp90 and 17-AAG inhibition in human epithelial cells to further elucidate the involvement of Hsp90 in NadA-mediated adhesion and invasion. Together, these data suggest an inverse correlation between the amount of host Hsp90 and the NadA adhesive/invasive phenotype. Confocal microscopy also demonstrated that meningococci interact with cellular Hsp90, a completely novel finding. Altogether our results show that variation of host Hsp90 expression or activity interferes with adhesive and invasive events driven by NadA.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion , HSP90 Heat-Shock Proteins/metabolism , Neisseria meningitidis/physiology , Amino Acid Sequence , Benzoquinones/pharmacology , Cells, Cultured , Escherichia coli/genetics , Escherichia coli/physiology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Host-Pathogen Interactions , Humans , Lactams, Macrocyclic/pharmacology , Meningococcal Infections/metabolism , Meningococcal Infections/microbiology , Molecular Sequence Data , Protein Binding , Recombinant Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 107(8): 3770-5, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20133713

ABSTRACT

GNA2132 is a Neisseria meningitidis antigen of unknown function, discovered by reverse vaccinology, which has been shown to induce bactericidal antibodies in animal models. Here we show that this antigen induces protective immunity in humans and it is recognized by sera of patients after meningococcal disease. The protein binds heparin in vitro through an Arg-rich region and this property correlates with increased survival of the unencapsulated bacterium in human serum. Furthermore, two proteases, the meningococcal NalP and human lactoferrin, cleave the protein upstream and downstream from the Arg-rich region, respectively. We conclude that GNA2132 is an important protective antigen of N. meningitidis and we propose to rename it, Neisserial Heparin Binding Antigen (NHBA).


Subject(s)
Antigens, Bacterial/immunology , Antimicrobial Cationic Peptides/immunology , Blood Proteins/immunology , Carrier Proteins/immunology , Meningococcal Vaccines/immunology , Neisseria meningitidis/immunology , Virulence Factors/immunology , Amino Acid Sequence , Antibodies, Bacterial/blood , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/genetics , Blood Proteins/chemistry , Blood Proteins/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Humans , Lactoferrin/chemistry , Meningococcal Infections/immunology , Meningococcal Infections/prevention & control , Meningococcal Vaccines/chemistry , Meningococcal Vaccines/genetics , Neisseria meningitidis/pathogenicity , Virulence Factors/chemistry , Virulence Factors/genetics
5.
Sci Transl Med ; 3(91): 91ra62, 2011 Jul 13.
Article in English | MEDLINE | ID: mdl-21753121

ABSTRACT

The sequence variability of protective antigens is a major challenge to the development of vaccines. For Neisseria meningitidis, the bacterial pathogen that causes meningitis, the amino acid sequence of the protective antigen factor H binding protein (fHBP) has more than 300 variations. These sequence differences can be classified into three distinct groups of antigenic variants that do not induce cross-protective immunity. Our goal was to generate a single antigen that would induce immunity against all known sequence variants of N. meningitidis. To achieve this, we rationally designed, expressed, and purified 54 different mutants of fHBP and tested them in mice for the induction of protective immunity. We identified and determined the crystal structure of a lead chimeric antigen that was able to induce high levels of cross-protective antibodies in mice against all variant strains tested. The new fHBP antigen had a conserved backbone that carried an engineered surface containing specificities for all three variant groups. We demonstrate that the structure-based design of multiple immunodominant antigenic surfaces on a single protein scaffold is possible and represents an effective way to create broadly protective vaccines.


Subject(s)
Antigens, Bacterial/immunology , Drug Design , Immunity/immunology , Neisseria meningitidis/immunology , Animals , Anti-Bacterial Agents/pharmacology , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Crystallography, X-Ray , Humans , Immunity/drug effects , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/immunology , Mutation/genetics , Neisseria meningitidis/drug effects , Protein Engineering , Protein Structure, Secondary
6.
Proc Natl Acad Sci U S A ; 103(29): 10834-9, 2006 Jul 18.
Article in English | MEDLINE | ID: mdl-16825336

ABSTRACT

Meningitis and sepsis caused by serogroup B meningococcus are two severe diseases that still cause significant mortality. To date there is no universal vaccine that prevents these diseases. In this work, five antigens discovered by reverse vaccinology were expressed in a form suitable for large-scale manufacturing and formulated with adjuvants suitable for human use. The vaccine adjuvanted by aluminum hydroxide induced bactericidal antibodies in mice against 78% of a panel of 85 meningococcal strains representative of the global population diversity. The strain coverage could be increased to 90% and above by the addition of CpG oligonucleotides or by using MF59 as adjuvant. The vaccine has the potential to conquer one of the most devastating diseases of childhood.


Subject(s)
Meningococcal Vaccines/immunology , Neisseria meningitidis, Serogroup B/immunology , Animals , Antibodies/immunology , Antigens, Bacterial/immunology , Disease Models, Animal , Humans , Meningitis, Meningococcal/immunology , Meningitis, Meningococcal/microbiology , Meningitis, Meningococcal/prevention & control , Mice , Microscopy, Electron, Transmission , Neisseria meningitidis, Serogroup B/classification , Neisseria meningitidis, Serogroup B/ultrastructure , Rats
7.
Mol Microbiol ; 55(3): 687-98, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15660996

ABSTRACT

Neisseria meningitidis is a human pathogen, which is a major cause of sepsis and meningitis. The bacterium colonizes the upper respiratory tract of approximately 10% of humans where it lives as a commensal. On rare occasions, it crosses the epithelium and reaches the bloodstream causing sepsis. From the bloodstream it translocates the blood-brain barrier, causing meningitis. Although all strains have the potential to cause disease, a subset of them, which belongs to hypervirulent lineages, causes disease more frequently than others. Recently, we described NadA, a novel antigen of N. meningitidis, present in three of the four known hypervirulent lineages. Here we show that NadA is a novel bacterial invasin which, when expressed on the surface of Escherichia coli, promotes adhesion to and invasion into Chang epithelial cells. Deletion of the N-terminal globular domain of recombinant NadA or pronase treatment of human cells abrogated the adhesive phenotype. A hypervirulent strain of N. meningitidis where the nad A gene was inactivated had a reduced ability to adhere to and invade into epithelial cells in vitro. NadA is likely to improve the fitness of N. meningitidis contributing to the increased virulence of strains that belong to the hypervirulent lineages.


Subject(s)
Bacterial Adhesion , Epithelial Cells/microbiology , Neisseria meningitidis/pathogenicity , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Cell Line, Tumor , Escherichia coli/genetics , Escherichia coli/metabolism , Flow Cytometry , Humans , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Neisseria meningitidis/genetics , Neisseria meningitidis/physiology
8.
Infect Immun ; 72(7): 4217-23, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15213166

ABSTRACT

NadA is a novel vaccine candidate recently identified in Neisseria meningitidis and involved in adhesion to host tissues. The nadA gene has been found in approximately 50% of the strains isolated from patients and in three of the four hypervirulent lineages of non-serogroup A strains. Here we investigated the presence of the nadA gene in 154 meningococcal strains isolated from healthy people (carrier strains). Only 25 (16.2%) of the 154 carrier isolates harbored the nadA gene. The commensal species Neisseria lactamica was also found not to have the nadA gene. Eighteen of the carrier strains belonged to the ET-5 and ET-37 hypervirulent clusters, indicating that only the 5.1% of the genuine carrier population actually harbored nadA (7 of 136 strains). Five of the seven strains harbored a novel allele of the nadA gene that was designated nadA4. The NadA4 protein was present on the bacterial surface as heat-stable high-molecular-weight oligomers. Antibodies against the recombinant NadA4 protein were bactericidal against homologous strains, whereas the activity against other NadA alleles was weak. In conclusion, the nadA gene segregates differently in the population of strains isolated from healthy individuals and in the population of strains isolated from patients. The presence of NadA can therefore be used as a tool to study the dynamics of meningococcal infections and understand why this bacterium, which is mostly a commensal, can become a severe pathogen.


Subject(s)
Antigens, Bacterial/immunology , Meningococcal Vaccines/immunology , Neisseria meningitidis/immunology , Alleles , Amino Acid Sequence , Animals , Antibodies/immunology , Antigens, Bacterial/genetics , Base Sequence , Epithelial Cells/immunology , Female , Humans , Meningococcal Vaccines/genetics , Mice , Molecular Sequence Data , Neisseria meningitidis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL