Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 14: 1397940, 2024.
Article in English | MEDLINE | ID: mdl-38751999

ABSTRACT

Non-typeable Haemophilus influenzae (NTHi) and Moraxella catarrhalis (Mcat) are two common respiratory tract pathogens often associated with acute exacerbations in Chronic Obstructive Pulmonary Disease (COPD) as well as with otitis media (OM) in children. Although there is evidence that these pathogens can adopt persistence mechanisms such as biofilm formation, the precise means through which they contribute to disease severity and chronicity remains incompletely understood, posing challenges for their effective eradication. The identification of potential vaccine candidates frequently entails the characterization of the host-pathogen interplay in vitro even though this approach is limited by the fact that conventional models do not permit long term bacterial infections. In the present work, by using air-liquid-interface (ALI) human airway in vitro models, we aimed to recreate COPD-related persistent bacterial infections. In particular, we explored an alternative use of the ALI system consisting in the assembly of an inverted epithelium grown on the basal part of a transwell membrane with the aim to enable the functionality of natural defense mechanisms such as mucociliary clearance and cellular extrusion that are usually hampered during conventional ALI infection experiments. The inversion of the epithelium did not affect tissue differentiation and considerably delayed NTHi or Mcat infection progression, allowing one to monitor host-pathogen interactions for up to three weeks. Notably, the use of these models, coupled with confocal and transmission electron microscopy, revealed unique features associated with NTHi and Mcat infection, highlighting persistence strategies including the formation of intracellular bacterial communities (IBCs) and surface-associated biofilm-like structures. Overall, this study demonstrates the possibility to perform long term host-pathogen investigations in vitro with the aim to define persistence mechanisms adopted by respiratory pathogens and individuate potential new vaccine targets.


Subject(s)
Biofilms , Haemophilus influenzae , Moraxella catarrhalis , Moraxellaceae Infections , Moraxella catarrhalis/physiology , Humans , Haemophilus influenzae/physiology , Haemophilus influenzae/pathogenicity , Biofilms/growth & development , Moraxellaceae Infections/microbiology , Persistent Infection/microbiology , Host-Pathogen Interactions , Haemophilus Infections/microbiology , Pulmonary Disease, Chronic Obstructive/microbiology , Models, Biological , Respiratory Tract Infections/microbiology , Epithelial Cells/microbiology
2.
Respir Res ; 24(1): 243, 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37798723

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is a lung disease characterised by airflow-limiting inflammation and mucus production. Acute exacerbations are a major cause of COPD-related morbidity and mortality and are mostly associated with bacterial or viral infections. A vaccine targeting non-typeable Haemophilus influenzae (NTHi) and Moraxella catarrhalis (Mcat), the main bacteria associated with exacerbations, was tested in a Phase 2 trial. We assessed "ex-vivo" expression of vaccine candidate and housekeeping genes pd, pe, pilA, gapA, ompP6 of NTHi, and uspA2, parE, polA of Mcat in sputum samples of COPD patients and determined whether expression of the vaccine candidate genes pd, pe, pilA (NTHi) and uspA2 (Mcat) differed between stable and exacerbation samples. METHODS: A single-centre, prospective, observational cohort study was conducted where 123 COPD patients were seen on enrolment, followed monthly for 2 years, and reviewed after onset of acute exacerbations. We selected 69 patients with sputum samples positive for NTHi or Mcat by PCR during at least one stable and one exacerbation visit. mRNA was isolated from the sputum, and expression of NTHi and Mcat genes was analysed with RT-PCR. Statistical analyses compared mRNA concentrations between stable and exacerbation samples and in relationship to COPD severity and exacerbation frequency. RESULTS: The vaccine candidate genes were variably expressed in sputum samples, suggesting they are expressed in the lung. Absolute and relative expression of all NTHi vaccine candidate genes and Mcat uspA2 were similar between exacerbation and stable samples. Expression of pd and pilA was slightly associated with the number of exacerbations in the year before enrolment, and uspA2 with the disease severity status at enrolment. CONCLUSIONS: The NTHi-Mcat vaccine candidate genes were expressed in sputum samples, and each gene had a specific level of expression. No statistically significant differences in gene expression were detectable between stable and exacerbation samples. However, the history of COPD exacerbations was slightly associated with the expression of pd, pilA and uspA2. Trial registration NCT01360398 ( https://www. CLINICALTRIALS: gov ).


Subject(s)
Haemophilus Vaccines , Pulmonary Disease, Chronic Obstructive , Humans , Sputum/microbiology , Prospective Studies , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/genetics , Moraxella catarrhalis , Haemophilus influenzae , RNA, Messenger , RNA
3.
Vaccine ; 40(45): 6520-6527, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36202640

ABSTRACT

Moraxella catarrhalis is an important and common respiratory pathogen that can cause Otitis Media, Community Acquired Pneumonia, and has been associated with an increased risk of exacerbations in chronic obstructive pulmonary disease in adults, leading to morbidity and mortality. Its ubiquitous surface protein A2 (UspA2) has been shown to interact with host structures and extracellular matrix proteins, suggesting a role at an early stage of infection and a contribution to bacterial serum resistance. The UspA proteins are homo-trimeric autotransporters that appear as a lollipop-shaped structure in electron micrographs. They are composed of an N-terminal head with adhesive properties, followed by a stalk, which ends by an amphipathic helix and a C-terminal membrane domain. The three family members UspA1, UspA2 and UspA2H, present different amino acid signatures both at the head and membrane-spanning regions. By combining electron microscopy, hydrogen deuterium exchange mass spectrometry and protein modeling, we identified a shared and repeated epitope recognized by FHUSPA2/10, a potent cross-bactericidal monoclonal antibody raised by UspA2 and deduced key amino acids involved in the binding. The finding strengthens the potential of UspA2 to be incorporated in a vaccine formulation against M. catarrhalis.


Subject(s)
Anti-Bacterial Agents , Antibodies, Monoclonal , Moraxella catarrhalis , Adult , Humans , Amino Acids/metabolism , Antibodies, Monoclonal/pharmacology , Bacterial Outer Membrane Proteins/immunology , Epitopes/metabolism , Extracellular Matrix Proteins/metabolism , Type V Secretion Systems/metabolism , Anti-Bacterial Agents/pharmacology
4.
iScience ; 25(3): 103931, 2022 Mar 18.
Article in English | MEDLINE | ID: mdl-35265810

ABSTRACT

Moraxella catarrhalis and nontypeable Haemophilus influenzae (NTHi) are pathogenic bacteria frequently associated with exacerbation of chronic obstructive pulmonary disease (COPD), whose hallmark is inflammatory oxidative stress. Neutrophils produce reactive oxygen species (ROS) which can boost antimicrobial response by promoting neutrophil extracellular traps (NET) and autophagy. Here, we showed that M. catarrhalis induces less ROS and NET production in differentiated HL-60 cells compared to NTHi. It is also able to actively interfere with these responses in chemically activated cells in a phagocytosis and opsonin-independent and contact-dependent manner, possibly by engaging host immunosuppressive receptors. M. catarrhalis subverts the autophagic pathway of the phagocytic cells and survives intracellularly. It also promotes the survival of NTHi which is otherwise susceptible to the host antimicrobial arsenal. In-depth understanding of the immune evasion strategies exploited by these two human pathogens could suggest medical interventions to tackle COPD and potentially other diseases in which they co-exist.

5.
Front Immunol ; 12: 749432, 2021.
Article in English | MEDLINE | ID: mdl-34819932

ABSTRACT

Staphylococcus aureus is a common human commensal and the leading cause of diverse infections. To identify distinctive parameters associated with infection and colonization, we compared the immune and inflammatory responses of patients with a diagnosis of invasive S. aureus disease to healthy donors. We analyzed the inflammatory responses founding a pattern of distinctive cytokines significantly higher in the patients with invasive disease. The measure of antibody levels revealed a wide antibody responsiveness from all subjects to most of the antigens, with significantly higher response for some antigens in the invasive patients compared to control. Moreover, functional antibodies against toxins distinctively associated with the invasive disease. Finally, we examined the genomic variability of isolates, showing no major differences in genetic distribution compared to a panel of representative strains. Overall, our study shows specific signatures of cytokines and functional antibodies in patients with different primary invasive diseases caused by S. aureus. These data provide insight into human responses towards invasive staphylococcal infections and are important for guiding the identification of novel preventive and therapeutic interventions against S. aureus.


Subject(s)
Staphylococcal Infections/immunology , Adult , Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Child , Cytokines/blood , Humans , Immunoglobulin G/blood , Protein Array Analysis , Staphylococcal Infections/blood , Staphylococcal Infections/genetics , Staphylococcus aureus/immunology , Virulence Factors/immunology
6.
PLoS One ; 14(8): e0203234, 2019.
Article in English | MEDLINE | ID: mdl-31369555

ABSTRACT

Neisserial Heparin Binding Antigen (NHBA) is a surface-exposed lipoprotein of Neisseria meningitidis and a component of the Bexsero vaccine. NHBA is characterized by the presence of a highly conserved Arg-rich region involved in binding to heparin and heparan sulphate proteoglycans present on the surface of host epithelial cells, suggesting a possible role of NHBA during N. meningitidis colonization. NHBA has been shown to be cleaved by the meningococcal protease NalP and by human lactoferrin (hLF), a host protease presents in different body fluids (saliva, breast milk and serum). Cleavage occurs upstream or downstream the Arg-rich region. Since the human nasopharynx is the only known reservoir of infection, we further investigated the susceptibility of NHBA to human proteases present in the saliva to assess whether proteolytic cleavage could happen during the initial steps of colonization. Here we show that human saliva proteolytically cleaves NHBA, and identified human kallikrein 1 (hK1), a serine protease, as responsible for this cleavage. Kallikrein-related peptidases (KLKs) have a distinct domain structure and exist as a family of 15 genes which are differentially expressed in many tissues and in the central nervous system. They are present in plasma, lymph, urine, saliva, pancreatic juices, and other body fluids where they catalyze the proteolysis of several human proteins. Here we report the characterization of NHBA cleavage by the tissue kallikrein, expressed in saliva and the identification of the cleavage site on NHBA both, as recombinant protein or as native protein, when expressed on live bacteria. Overall, these findings provide new insights on NHBA as target of host proteases, highlights thepotential role of NHBA in the Neisseria meningitidis nasopharyngeal colonization, and of kallikrein as a defensive agent against meningococcal infection.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/metabolism , Meningococcal Infections/microbiology , Proteolysis , Saliva/chemistry , Tissue Kallikreins/metabolism , Amino Acid Sequence , Humans , Neisseria meningitidis/physiology , Proteomics
7.
PLoS One ; 13(3): e0194662, 2018.
Article in English | MEDLINE | ID: mdl-29579105

ABSTRACT

Neisserial Heparin Binding Antigen (NHBA) is a surface-exposed lipoprotein specific for Neisseria and constitutes one of the three main protein antigens of the Bexsero vaccine. Meningococcal and human proteases, cleave NHBA protein upstream or downstream of a conserved Arg-rich region, respectively. The cleavage results in the release of the C-terminal portion of the protein. The C-terminal fragment originating from the processing of meningococcal proteases, referred to as C2 fragment, exerts a toxic effect on endothelial cells altering the endothelial permeability. In this work, we reported that recombinant C2 fragment has no influence on the integrity of human airway epithelial cell monolayers, consistent with previous findings showing that Neisseria meningitidis traverses the epithelial barrier without disrupting the junctional structures. We showed that epithelial cells constantly secrete proteases responsible for a rapid processing of C2 fragment, generating a new fragment that does not contain the Arg-rich region, a putative docking domain reported to be essential for C2-mediated toxic effect. Moreover, we found that the C3-convertase of the alternative complement pathway is one of the proteases responsible for this processing. Overall, our data provide new insights on the cleavage of NHBA protein during meningococcal infection. NHBA cleavage may occur at different stages of the infection, and it likely has a different role depending on the environment the bacterium is interacting with.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/metabolism , Complement C3 Convertase, Alternative Pathway/metabolism , Neisseria/metabolism , Amino Acid Sequence , Antigens, Bacterial/chemistry , Bacterial Outer Membrane Proteins/chemistry , Carrier Proteins/chemistry , Cell Line , Edetic Acid/pharmacology , Epithelial Cells/cytology , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Humans , Magnesium/chemistry , Magnesium/metabolism , Peptide Hydrolases/metabolism , Proteolysis/drug effects , Proteomics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Zinc/chemistry , Zinc/metabolism
8.
PLoS One ; 11(10): e0162878, 2016.
Article in English | MEDLINE | ID: mdl-27780200

ABSTRACT

Neisserial Heparin Binding Antigen (NHBA) is a surface-exposed lipoprotein ubiquitously expressed by Neisseria meningitidis strains and an antigen of the Bexsero® vaccine. NHBA binds heparin through a conserved Arg-rich region that is the target of two proteases, the meningococcal NalP and human lactoferrin (hLf). In this work, in vitro studies showed that recombinant NHBA protein was able to bind epithelial cells and mutations of the Arg-rich tract abrogated this binding. All N-terminal and C-terminal fragments generated by NalP or hLf cleavage, regardless of the presence or absence of the Arg-rich region, did not bind to cells, indicating that a correct positioning of the Arg-rich region within the full length protein is crucial. Moreover, binding was abolished when cells were treated with heparinase III, suggesting that this interaction is mediated by heparan sulfate proteoglycans (HSPGs). N. meningitidis nhba knockout strains showed a significant reduction in adhesion to epithelial cells with respect to isogenic wild-type strains and adhesion of the wild-type strain was inhibited by anti-NHBA antibodies in a dose-dependent manner. Overall, the results demonstrate that NHBA contributes to meningococcal adhesion to epithelial cells through binding to HSPGs and suggest a possible role of anti-Bexsero® antibodies in the prevention of colonization.


Subject(s)
Antibodies, Bacterial/genetics , Antibodies, Bacterial/metabolism , Bacterial Adhesion , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/metabolism , Epithelial Cells/microbiology , Meningococcal Infections/drug therapy , Neisseria meningitidis/physiology , Antibodies, Bacterial/chemistry , Binding Sites , Cell Line , Gene Knockout Techniques , Heparan Sulfate Proteoglycans/metabolism , Heparin/metabolism , Humans , Lactoferrin/metabolism , Membrane Transport Proteins/metabolism , Meningococcal Infections/microbiology , Meningococcal Infections/prevention & control , Mutation , Neisseria meningitidis/immunology , Recombinant Proteins/metabolism , Serine Endopeptidases/metabolism
9.
PLoS One ; 11(1): e0147767, 2016.
Article in English | MEDLINE | ID: mdl-26812180

ABSTRACT

A rapidly acting, single dose vaccine against Staphylococcus aureus would be highly beneficial for patients scheduled for major surgeries or in intensive care units. Here we show that one immunization with a multicomponent S. aureus candidate vaccine, 4C-Staph, formulated with a novel TLR7-dependent adjuvant, T7-alum, readily protected mice from death and from bacterial dissemination, both in kidney abscess and peritonitis models, outperforming alum-formulated vaccine. This increased efficacy was paralleled by higher vaccine-specific and α-hemolysin-neutralizing antibody titers and Th1/Th17 cell responses. Antibodies played a crucial protective role, as shown by the lack of protection of 4C-Staph/T7-alum vaccine in B-cell-deficient mice and by serum transfer experiments. Depletion of effector CD4+ T cells not only reduced survival but also increased S. aureus load in kidneys of mice immunized with 4C-Staph/T7-alum. The role of IL-17A in the control of bacterial dissemination in 4C-Staph/T7-alum vaccinated mice was indicated by in vivo neutralization experiments. We conclude that single dose 4C-Staph/T7-alum vaccine promptly and efficiently protected mice against S. aureus through the combined actions of antibodies, CD4+ effector T cells, and IL-17A. These data suggest that inclusion of an adjuvant that induces not only fast antibody responses but also IL-17-producing cell-mediated effector responses could efficaciously protect patients scheduled for major surgeries or in intensive care units.


Subject(s)
Antibodies, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , Interleukin-17/metabolism , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Toll-Like Receptor 7/metabolism , Adjuvants, Immunologic , Animals , Antibodies, Neutralizing/immunology , CD4-Positive T-Lymphocytes/cytology , Cytokines/metabolism , Female , Mice , Mice, Inbred C57BL , Spleen/metabolism , Spleen/pathology , Staphylococcal Infections/immunology , Staphylococcal Infections/mortality , Staphylococcus aureus/genetics , Survival Rate , Th1 Cells/immunology , Th17 Cells/immunology , Toll-Like Receptor 7/immunology
10.
Front Immunol ; 6: 439, 2015.
Article in English | MEDLINE | ID: mdl-26441955

ABSTRACT

Staphylococcus aureus (S. aureus) is an important opportunistic pathogen that may cause invasive life-threatening infections, like sepsis and pneumonia. Due to the increasing antibiotic resistance, the development of an effective vaccine against S. aureus is needed. Although a correlate of protection against staphylococcal diseases is not yet established, several findings suggest that both antibodies and CD4 T cells might contribute to optimal immunity. In this study, we show that adjuvanting a multivalent vaccine (4C-Staph) with MF59, an oil-in-water emulsion licensed in human vaccines, further potentiated antigen-specific IgG titers and CD4 T-cell responses compared to alum and conferred protection in the peritonitis model of S. aureus infection. Moreover, we showed that MF59- and alum-adjuvanted 4C-Staph vaccines induced persistent antigen-specific humoral and T-cell responses, and protected mice from infection up to 4 months after immunization. Furthermore, 4C-Staph formulated with MF59 was used to investigate which immune compartment is involved in vaccine-induced protection. Using CD4 T cell-depleted mice or B cell-deficient mice, we demonstrated that both T and B-cell responses contributed to 4C-Staph vaccine-mediated protective immunity. However, the role of CD4 T cells seemed more evident in the presence of low-antibody responses. This study provides preclinical data further supporting the use of the adjuvanted 4C-Staph vaccines against S. aureus diseases, and provides critical insights on the correlates of protective immunity necessary to combat this pathogen.

11.
J Immunol ; 195(4): 1617-27, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26170383

ABSTRACT

Induction of persistent protective immune responses is a key attribute of a successful vaccine formulation. MF59 adjuvant, an oil-in-water emulsion used in human vaccines, is known to induce persistent high-affinity functional Ab titers and memory B cells, but how it really shapes the Ag-specific B cell compartment is poorly documented. In this study, we characterized the Ab- and Ag-specific B cell compartment in wild-type mice immunized with HlaH35L, a Staphylococcus aureus Ag known to induce measurable functional Ab responses, formulated with MF59 or aluminum salts, focusing on germinal centers (GC) in secondary lymphoid organs. Taking advantage of single-cell flow cytometry analyses, HlaH35L-specific B cells were characterized for the expression of CD38 and GL-7, markers of memory and GC, respectively, and for CD80 and CD73 activation markers. We demonstrated that immunization with MF59-, but not aluminum salt-adjuvanted HlaH35L, induced expanded Ag-specific CD73(+)CD80(-) GC B cells in proximal- and distal-draining lymph nodes, and promoted the persistence of GC B cells, detected up to 4 mo after immunization. In addition to increasing GC B cells, MF59-adjuvanted HlaH35L also increased the frequency of T follicular helper cells. This work extends previous knowledge regarding adaptive immune responses to MF59-adjuvanted vaccines, and, to our knowledge, for the first time an adjuvant used in human licensed products is shown to promote strong and persistent Ag-specific GC responses that might benefit the rational design of new vaccination strategies.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation , Germinal Center/cytology , Germinal Center/immunology , Polysorbates , Squalene , Vaccination , Adjuvants, Immunologic , Animals , Antibodies, Bacterial/immunology , Antibody Specificity/immunology , Antigens, CD/metabolism , B-Lymphocytes/metabolism , Bacterial Toxins/immunology , Chemotaxis, Leukocyte/immunology , Female , Hemolysin Proteins/immunology , Immunophenotyping , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Mice , Phenotype , Squalene/immunology , Staphylococcal Vaccines
12.
PLoS One ; 10(5): e0127614, 2015.
Article in English | MEDLINE | ID: mdl-25996923

ABSTRACT

Many pathogenic bacteria utilize ADP-ribosylating toxins to modify and impair essential functions of eukaryotic cells. It has been previously reported that Neisseria meningitidis possesses an ADP-ribosyltransferase enzyme, NarE, retaining the capacity to hydrolyse NAD and to transfer ADP-ribose moiety to arginine residues in target acceptor proteins. Here we show that upon internalization into human epithelial cells, NarE gains access to the cytoplasm and, through its ADP-ribosylating activity, targets host cell proteins. Notably, we observed that these events trigger the disruption of the epithelial monolayer integrity and the activation of the apoptotic pathway. Overall, our findings provide, for the first time, evidence for a biological activity of NarE on host cells, suggesting its possible involvement in Neisseria pathogenesis.


Subject(s)
ADP Ribose Transferases/metabolism , Epithelial Cells/metabolism , Epithelial Cells/virology , Neisseria meningitidis/metabolism , Actins/metabolism , Animals , Apoptosis , Endocytosis , Epithelial Cells/pathology , HeLa Cells , Humans , Intracellular Space/metabolism , Mice , Protein Binding , Protein Transport
13.
BMC Microbiol ; 15: 87, 2015 Apr 18.
Article in English | MEDLINE | ID: mdl-25927946

ABSTRACT

BACKGROUND: Non-typeable Haemophilus influenzae (NTHi) is a Gram negative microorganism residing in the human nasopharyngeal mucosa and occasionally causing infections of both middle ear and lower respiratory airways. A broadly protective vaccine against NTHi has been a long-unmet medical need, as the high genetic variability of this bacterium has posed great challenges. RESULTS: In this study, we developed a robust serum bactericidal assay (SBA) to optimize the selection of protective antigens against NTHi. SBA takes advantage of the complement-mediated lysis of bacterial cells and is a key in vitro method for measuring the functional activity of antibodies. As a proof of concept, we assessed the bactericidal activity of antibodies directed against antigens known to elicit a protective response, including protein D used as carrier protein in the Synflorix pneumococcal polysaccharide conjugate vaccine. Prior to SBA screening, the accessibility of antigens to antibodies and the capacity of the latter to induce C3 complement deposition was verified by flow cytometry. Using baby rabbit serum as a source of complement, the proposed assay not only confirmed the bactericidal activity of the antibodies against the selected vaccine candidates, but also showed a significant reproducibility. CONCLUSIONS: Considering the rapidity and cost-effectiveness of this novel SBA protocol, we conclude that it is likely to become an important tool to prove the capability of antibodies directed against recombinant antigens to induce NTHi in vitro killing and to both select new protective vaccine candidates, and predict vaccine efficacy.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Blood Bactericidal Activity , Haemophilus influenzae/immunology , Haemophilus influenzae/physiology , Microbial Viability/drug effects , Animals , Guinea Pigs , Haemophilus influenzae/drug effects , Immunoassay/methods , Mice , Rabbits
14.
Cell Microbiol ; 17(9): 1365-75, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25801707

ABSTRACT

Translocation of the nasopharyngeal barrier by Neisseria meningitidis occurs via an intracellular microtubule-dependent pathway and represents a crucial step in its pathogenesis. Despite this fact, the interaction of invasive meningococci with host subcellular compartments and the resulting impact on their organization and function have not been investigated. The influence of serogroup B strain MC58 on host cell polarity and intracellular trafficking system was assessed by confocal microscopy visualization of different plasma membrane-associated components (such as E-cadherin, ZO-1 and transferrin receptor) and evaluation of the transferrin uptake and recycling in infected Calu-3 monolayers. Additionally, the association of N. meningitidis with different endosomal compartments was evaluated through the concomitant staining of bacteria and markers specific for Rab11, Rab22a, Rab25 and Rab3 followed by confocal microscopy imaging. Subversion of the host cell architecture and intracellular trafficking system, denoted by mis-targeting of cell plasma membrane components and perturbations of transferrin transport, was shown to occur in response to N. meningitidis infection. Notably, the appearance of all of these events seems to positively correlate with the efficiency of N. meningitidis to cross the epithelial barrier. Our data reveal for the first time that N. meningitidis is able to modulate the host cell architecture and function, which might serve as a strategy of this pathogen for overcoming the nasopharyngeal barrier without affecting the monolayer integrity.


Subject(s)
Cell Polarity , Epithelial Cells/microbiology , Epithelial Cells/physiology , Host-Pathogen Interactions , Neisseria meningitidis, Serogroup B/physiology , Transcytosis , Cell Line , Humans , Microscopy, Confocal
15.
Proc Natl Acad Sci U S A ; 112(12): 3680-5, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25775551

ABSTRACT

Both active and passive immunization strategies against Staphylococcus aureus have thus far failed to show efficacy in humans. With the attempt to develop an effective S. aureus vaccine, we selected five conserved antigens known to have different roles in S. aureus pathogenesis. They include the secreted factors α-hemolysin (Hla), ess extracellular A (EsxA), and ess extracellular B (EsxB) and the two surface proteins ferric hydroxamate uptake D2 and conserved staphylococcal antigen 1A. The combined vaccine antigens formulated with aluminum hydroxide induced antibodies with opsonophagocytic and functional activities and provided consistent protection in four mouse models when challenged with a panel of epidemiologically relevant S. aureus strains. The importance of antibodies in protection was demonstrated by passive transfer experiments. Furthermore, when formulated with a toll-like receptor 7-dependent (TLR7) agonist recently designed and developed in our laboratories (SMIP.7-10) adsorbed to alum, the five antigens provided close to 100% protection against four different staphylococcal strains. The new formulation induced not only high antibody titers but also a Th1 skewed immune response as judged by antibody isotype and cytokine profiles. In addition, low frequencies of IL-17-secreting T cells were also observed. Altogether, our data demonstrate that the rational selection of mixtures of conserved antigens combined with Th1/Th17 adjuvants can lead to promising vaccine formulations against S. aureus.


Subject(s)
Adjuvants, Immunologic/pharmacology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/chemistry , Toll-Like Receptor 7/chemistry , Abscess/pathology , Adaptive Immunity , Animals , Anti-Bacterial Agents/chemistry , Antibodies, Bacterial/immunology , Antigens/immunology , Humans , Mice , Models, Animal , Staphylococcal Infections/immunology , Staphylococcus aureus , Th1 Cells/immunology
16.
PLoS One ; 10(3): e0117486, 2015.
Article in English | MEDLINE | ID: mdl-25789808

ABSTRACT

SslE is a zinc-metalloprotease involved in the degradation of mucin substrates and recently proposed as a potential vaccine candidate against pathogenic E. coli. In this paper, by exploiting a human in vitro model of mucus-secreting cells, we demonstrated that bacteria expressing SslE have a metabolic benefit which results in an increased growth rate postulating the importance of this antigen in enhancing E. coli fitness. We also observed that SslE expression facilitates E. coli penetration of the mucus favouring bacteria adhesion to host cells. Moreover, we found that SslE-mediated opening of the mucosae contributed to the activation of pro-inflammatory events. Indeed, intestinal cells infected with SslE-secreting bacteria showed an increased production of IL-8 contributing to neutrophil recruitment. The results presented in this paper conclusively designate SslE as an important colonization factor favouring E. coli access to both metabolic substrates and target cells.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/pathogenicity , Intestinal Mucosa/microbiology , Polysaccharide-Lyases/metabolism , Virulence Factors/metabolism , Escherichia coli/enzymology , Humans , Virulence
17.
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
18.
mBio ; 4(4)2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23882011

ABSTRACT

UNLABELLED: In this study, we have characterized the functional properties of a novel Escherichia coli antigen named EsiB (E. coli secretory immunoglobulin A-binding protein), recently reported to protect mice from sepsis. Gene distribution analysis of a panel of 267 strains representative of different E. coli pathotypes revealed that esiB is preferentially associated with extraintestinal strains, while the gene is rarely found in either intestinal or nonpathogenic strains. These findings were supported by the presence of anti-EsiB antibodies in the sera of patients affected by urinary tract infections (UTIs). By solving its crystal structure, we observed that EsiB adopts a superhelical fold composed of Sel1-like repeats (SLRs), a feature often associated with bacterial proteins possessing immunomodulatory functions. Indeed, we found that EsiB interacts with secretory immunoglobulin A (SIgA) through a specific motif identified by an immunocapturing approach. Functional assays showed that EsiB binding to SIgA is likely to interfere with productive FcαRI signaling, by inhibiting both SIgA-induced neutrophil chemotaxis and respiratory burst. Indeed, EsiB hampers SIgA-mediated signaling events by reducing the phosphorylation status of key signal-transducer cytosolic proteins, including mitogen-activated kinases. We propose that the interference with such immune events could contribute to the capacity of the bacterium to avoid clearance by neutrophils, as well as reducing the recruitment of immune cells to the infection site. IMPORTANCE: Pathogenic Escherichia coli infections have recently been exacerbated by increasing antibiotic resistance and the number of recurrent contagions. Attempts to develop preventive strategies against E. coli have not been successful, mainly due to the large antigenic and genetic variability of virulence factors, but also due to the complexity of the mechanisms used by the pathogen to evade the immune system. In this work, we elucidated the function of a recently discovered protective antigen, named EsiB, and described its capacity to interact with secretory immunoglobulin A (SIgA) and impair effector functions. This work unravels a novel strategy used by E. coli to subvert the host immune response and avoid neutrophil-dependent clearance.


Subject(s)
Antigens, Bacterial/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/immunology , Escherichia coli/pathogenicity , Immunoglobulin A, Secretory/metabolism , Neutrophil Activation , Virulence Factors/metabolism , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Gene Knockout Techniques , Humans , Immune Evasion , Mice , Models, Molecular , Protein Conformation , Virulence Factors/chemistry , Virulence Factors/genetics
19.
Mol Aspects Med ; 30(6): 456-66, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19560486

ABSTRACT

Successful colonization of the host by bacterial pathogens relies on their capacity to evade the complex and powerful defenses opposed by the host immune system, at least in the initial phases of infection. The two toxins of Bacillus anthracis, lethal toxin and edema toxin, appear to have been shaped by evolution to assist the microorganism in this crucial function, in addition to act as general toxins acting on almost all cell types. Edema toxin causes a consistent elevation of cAMP, an important second messenger the production of which is normally strictly controlled in mammalian cells, whereas lethal toxin cleaves most isoforms of mitogen-activated protein kinase kinases. By disrupting or subverting central modules common to all the principal signaling networks which control immune cell activation, effector function and migration, the anthrax toxins effectively and systematically dismantle both the innate and the adaptive immune defenses of the host. Here, we review the specific effects of the lethal and edema toxins of B. anthracis on the activation and function of phagocytes, dendritic cells and lymphocytes. We also discuss some open issues which should be addressed to gain a comprehensive insight into the complex relationship that B. anthracis establishes with the host.


Subject(s)
Adaptive Immunity , Antigens, Bacterial/immunology , Bacillus anthracis , Bacterial Toxins/immunology , Immunity, Innate , Virulence Factors/immunology , Animals , Antibodies/metabolism , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Cell Proliferation , Chemotaxis , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Lymphocytes/immunology , Macrophages/immunology , Monocytes/immunology , Neutrophils/immunology , Phagocytes/immunology , Signal Transduction/immunology
20.
PLoS Pathog ; 5(3): e1000325, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19266022

ABSTRACT

The adjuvanticity of bacterial adenylate cyclase toxins has been ascribed to their capacity, largely mediated by cAMP, to modulate APC activation, resulting in the expression of Th2-driving cytokines. On the other hand, cAMP has been demonstrated to induce a Th2 bias when present during T cell priming, suggesting that bacterial cAMP elevating toxins may directly affect the Th1/Th2 balance. Here we have investigated the effects on human CD4(+) T cell differentiation of two adenylate cyclase toxins, Bacillus anthracis edema toxin (ET) and Bordetella pertussis CyaA, which differ in structure, mode of cell entry, and subcellular localization. We show that low concentrations of ET and CyaA, but not of their genetically detoxified adenylate cyclase defective counterparts, potently promote Th2 cell differentiation by inducing expression of the master Th2 transcription factors, c-maf and GATA-3. We also present evidence that the Th2-polarizing concentrations of ET and CyaA selectively inhibit TCR-dependent activation of Akt1, which is required for Th1 cell differentiation, while enhancing the activation of two TCR-signaling mediators, Vav1 and p38, implicated in Th2 cell differentiation. This is at variance from the immunosuppressive toxin concentrations, which interfere with the earliest step in TCR signaling, activation of the tyrosine kinase Lck, resulting in impaired CD3zeta phosphorylation and inhibition of TCR coupling to ZAP-70 and Erk activation. These results demonstrate that, notwithstanding their differences in their intracellular localization, which result in focalized cAMP production, both toxins directly affect the Th1/Th2 balance by interfering with the same steps in TCR signaling, and suggest that their adjuvanticity is likely to result from their combined effects on APC and CD4(+) T cells. Furthermore, our results strongly support the key role of cAMP in the adjuvanticity of these toxins.


Subject(s)
Adenylate Cyclase Toxin/pharmacology , Antigens, Bacterial/pharmacology , Bacillus anthracis/enzymology , Bacterial Toxins/pharmacology , Bordetella pertussis/enzymology , Cell Differentiation/drug effects , Receptors, Antigen, T-Cell/metabolism , Adenylate Cyclase Toxin/genetics , Antigens, Bacterial/genetics , Bacillus anthracis/genetics , Bacterial Toxins/genetics , Bordetella pertussis/genetics , CD4-Positive T-Lymphocytes/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , GATA3 Transcription Factor/metabolism , Gene Expression Regulation , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-maf/metabolism , Signal Transduction/drug effects , Th2 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...