Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Clin Vaccine Immunol ; 20(9): 1360-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23803905

ABSTRACT

The poor immunogenicity of the meningococcal serogroup B (MenB) capsule has led to the development of vaccines targeting subcapsular antigens, in particular the immunodominant and diverse outer membrane porin, PorA. These vaccines are largely strain specific; however, they offer limited protection against the diverse MenB-associated diseases observed in many industrialized nations. To broaden the scope of its protection, the multicomponent vaccine (4CMenB) incorporates a PorA-containing outer membrane vesicle (OMV) alongside relatively conserved recombinant protein components, including factor H-binding protein (fHbp), Neisseria adhesin A (NadA), and neisserial heparin-binding antigen (NHBA). The expression of PorA is unique to meningococci (Neisseria meningitidis); however, many subcapsular antigens are shared with nonpathogenic members of the genus Neisseria that also inhabit the nasopharynx. These organisms may elicit cross-protective immunity against meningococci and/or occupy a niche that might otherwise accommodate pathogens. The potential for 4CMenB responses to impact such species (and vice versa) was investigated by determining the genetic distribution of the primary 4CMenB antigens among diverse members of the common childhood commensal, Neisseria lactamica. All the isolates possessed nhba but were devoid of fhbp and nadA. The nhba alleles were mainly distinct from but closely related to those observed among a representative panel of invasive MenB isolates from the same broad geographic region. We made similar findings for the immunogenic typing antigen, FetA, which constitutes a major part of the 4CMenB OMV. Thus, 4CMenB vaccine responses may impact or be impacted by nasopharyngeal carriage of commensal neisseriae. This highlights an area for further research and surveillance should the vaccine be routinely implemented.


Subject(s)
Antigens, Bacterial/immunology , Meningococcal Vaccines/immunology , Neisseria lactamica/immunology , Neisseria meningitidis, Serogroup B/immunology , Adolescent , Adult , Antigens, Bacterial/genetics , Carrier State/immunology , Carrier State/microbiology , Child , Child, Preschool , Cross Protection , Cross Reactions , Female , Humans , Infant , Male , Meningococcal Vaccines/genetics , Middle Aged , Nasopharynx/microbiology , Neisseria lactamica/genetics , Neisseria meningitidis, Serogroup B/genetics , Young Adult
2.
Hum Vaccin Immunother ; 9(3): 572-81, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23296384

ABSTRACT

Colonization of the nasopharynx by non-pathogenic Neisseria species, including N. lactamica, has been suggested to lead to the acquisition of natural immunity against Neisseria meningitidis in young children. The aim of this study was to identify a model complex of antigens and adjuvant for immunological preparation against N. meningitidis B, based on cross reactivity with N. lactamica outer membrane vesicles (OMV) antigens and the (DDA-BF) adjuvant. Complexes of 25 µg of OMV in 0.1 mM of DDA-BF were colloidally stable, exhibiting a mean diameter and charge optimal for antigen presentation. Immunogenicity tests for these complexes were performed in mice. A single dose of OMV/DDA-BF was sufficient to induce a (DTH) response, while the same result was achieved only after two doses of OMV/alum. In addition, to achieve total IgG levels that are similar to a single immunization with OMV/DDA-BF, it was necessary to give the mice a second dose of OMV/alum. Moreover, the antibodies induced from a single immunization with OMV/DDA-BF had an intermediate avidity, but antibodies with a similar avidity were only induced by OMV/alum after two immunizations. The use of this novel cationic adjuvant for the first time with a N. lactamica OMV preparation revealed good potential for future vaccine design.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Vaccines/immunology , Cations/administration & dosage , Neisseria lactamica/immunology , Adjuvants, Immunologic/metabolism , Animals , Antibodies, Bacterial/blood , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/isolation & purification , Cations/metabolism , Female , Immunoglobulin G/blood , Meningococcal Infections/prevention & control , Mice , Secretory Vesicles/immunology , Secretory Vesicles/metabolism
3.
São Paulo; s.n; 2012. 107 p.
Thesis in Portuguese | LILACS, Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP, SESSP-IBACERVO | ID: biblio-1080929

ABSTRACT

Neisseria lactamica está envolvida na aquisição da imunidade natural contra Neisseria meningitidis, causadora da doença meningocócica. Vesículas de membrana externa (OMV) de N. lactamica são antígenos potenciais contra N. meningitidis. Analisou-se a cinética de biomassa, de produção de OMV, da fonte de carbono (lactato), e dos metabólitos, para maximizar a produção de OMV. Realizaram-se ensaios em biorreator, em batelada simples, batelada alimentada com lactato, com ou sem pulsos de aminoácidos e extrato de levedura (YE). Utilizou-se o meio de Catlin (MC) como meio mínimo, e analisaram-se efeitos das concentrações do lactato, aminoácidos e YE. Lactato foi consumido e citrato e acetato produzidos. Os melhores resultados obtidos foram no meio com adição de 2 g/L de YE e concentrações dobradas de lactato e 5 aminoácidos constitutivos do MC, em batelada alimentada com pulsos. O lactato apresentou efeito positivo sobre o rendimento de OMV e o YE sobre a biomassa. Os 5 aminoácidos constitutivos do MC foram necessários para obtenção de biomassa e rendimento de OMV.


Neisseria lactamica is involved with the acquisition of natural immunity to Neisseria meningitidis. N. lactamica outer membrane vesicles (OMV) are potential antigens against N. meningitidis, the pathogen of meningococcal disease. The objective of this work was to analyze the kinetics of bacterial growth, OMV production, the carbon source (lactate), and products of metabolism, to improve growing conditions and OMV production. Groups were studied in batch process, fed-batch process with lactate, fed-batch process with pulses of amino acids and YE. MC was considered as minimal medium and it was analyzed the effect of lactate, amino acids and YE. Lactate was consumed and citrate and acetate increased in the medium. The best results were in fed-batch with pulses, in MC with the double concentrations of lactate and amino acids, added with 2 g/L of YE. The lactate had a positive effect over OMV yield and YE had a positive effect over biomass. 5 amino acids of MC were necessary to obtain biomass and OMV yield.


Subject(s)
Humans , Neisseria , Neisseria lactamica/immunology , Vaccines/immunology , Neisseria meningitidis/immunology
4.
Hum Vaccin ; 7(4): 398-401, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21795847

ABSTRACT

From pertussis to meningococcal disease and back represents nearly 30 years of research at Porton, first at the Centre for Applied Microbiology and Research and latterly as part of the Health Protection Agency. I joined the group lead by Andy Robinson developing an acellular pertussis vaccine and was part of an exciting period that encompassed basic antigen characterisation and pathogenesis studies with the development of an acellular vaccine containing fimbriae. Research then changed to focus on serogroup B meningococcal disease, studying the vaccine potential of iron-regulated proteins and then Neisseria lactamica. The resurgence of pertussis seen in some countries alerted me to the lack of understanding of protective immune responses to Bordetella pertussis infection and disease and this is now an active area of research.


Subject(s)
Meningitis, Meningococcal/epidemiology , Meningitis, Meningococcal/prevention & control , Meningococcal Vaccines/immunology , Pertussis Vaccine/immunology , Whooping Cough/epidemiology , Whooping Cough/prevention & control , Bordetella pertussis/genetics , Bordetella pertussis/immunology , Bordetella pertussis/pathogenicity , History, 20th Century , History, 21st Century , Humans , Meningitis, Meningococcal/immunology , Neisseria lactamica/genetics , Neisseria lactamica/immunology , Neisseria lactamica/pathogenicity , Whooping Cough/immunology
5.
Clin Infect Dis ; 52(1): 70-7, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21148522

ABSTRACT

BACKGROUND: Natural immunity to Neisseria meningitidis may result from nasopharyngeal carriage of closely related commensals, such as Neisseria lactamica. METHODS: We enrolled 61 students with no current carriage of Neisseria species and inoculated them intranasally with 10,000 colony-forming units of Neisseria lactamica or sham control. Colonization was monitored in oropharyngeal samples over 6 months. We measured specific mucosal and systemic antibody responses to N. lactamica and serum bactericidal antibody (SBA) and opsonophagocytic antibodies to a panel of N. meningitidis serogroup B strains. We also inoculated an additional cohort following vaccination with N. lactamica outer-membrane vesicles (OMV) produced from the same strain. RESULTS: Twenty-six (63.4%) of 41 inoculated individuals became colonized with N. lactamica; 85% remained colonized at 12 weeks. Noncarriers were resistant to rechallenge, and carriers who terminated carriage were relatively resistant to rechallenge. No carriers acquired N. meningitidis carriage over 24 weeks, compared with 3 control subjects (15%). Carriers developed serum IgG and salivary IgA antibodies to the inoculated N. lactamica strain by 4 weeks; noncarriers and control subjects did not. Cross-reactive serum bactericidal antibody responses to N.meningitidis were negligible in carriers, but they developed broad opsonophagocytic antimeningococcal antibodies. OMV vaccinees developed systemic and mucosal anti-N. lactamica antibodies and were relatively resistant to N. lactamica carriage but not to natural acquisition of N. meningitidis. CONCLUSIONS: Carriers of N. lactamica develop mucosal and systemic humoral immunity to N. lactamica together with cross-reacting systemic opsonophagocytic but not bactericidal antibodies to N. meningitidis. Possession of humoral immunity to N. lactamica inhibits acquisition of N. lactamica but not of N. meningitidis. Some individuals are intrinsically resistant to N. lactamica carriage, independent of humoral immunity.


Subject(s)
Carrier State/immunology , Nasopharynx/microbiology , Neisseria lactamica/immunology , Neisseria meningitidis, Serogroup B/immunology , Neisseriaceae Infections/immunology , Adolescent , Adult , Antibodies, Bacterial/blood , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Blood Bactericidal Activity , Carrier State/microbiology , Cross Reactions , Female , Humans , Immunity, Mucosal , Male , Middle Aged , Neisseria lactamica/isolation & purification , Neisseriaceae Infections/microbiology , Opsonin Proteins/immunology , Phagocytosis , Secretory Vesicles/immunology , Serum Bactericidal Antibody Assay , Young Adult
6.
Cell Microbiol ; 13(4): 554-68, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21105983

ABSTRACT

The upper respiratory tract commensal Neisseria lactamica (Nlac) induces protective humoral immunity against pathogenic Nmen serogroup B (Nmen), but whether it also affords anti-inflammatory mucosal protection, as reported for several gut commensals, has not been investigated. Here we demonstrate for the first time that Nlac weakly induces inflammatory responses compared with Nmen in the nasopharyngeal epithelial cell line, Detroit 562, and that Nlac achieves this by attenuation of secretory cytokine (TNF-α and IL-6) and to a lesser extent chemokine (IL-8 and RANTES) responses. Culture of Detroit cells with Nlac inhibited the induction of cytokine-chemokine mRNA by Nmen, reduced Nmen-induced NF-κß activity and increased constitutive PPAR-γ protein expression. Pretreatment of Detroit cells with a PPAR-γ antagonist abrogated the attenuation of inflammatory IL-6 by Nlac, as did heat-killing of the organisms and preventing their invasion with cytochalasin D. Inflammatory responses from Detroit cells were readily attenuated by Nlac following stimulation with pathogenic Nmen but more specifically following stimulation with the TLR-1/2 agonist Pam3Cys and pro-inflammatory cytokines (IL-1ß, TNF-α) but not LTA or LPS. These results indicate that Nlac plays an important role in suppressing pathogen-induced inflammation in the nasopharyngeal mucosa, mediated through TLR-1/2 stimulation, by activating PPAR-γ and inhibiting NF-κß activity.


Subject(s)
Cytokines/immunology , Epithelial Cells/microbiology , Nasopharynx/cytology , Neisseria lactamica/immunology , PPAR gamma/metabolism , Toll-Like Receptor 1/immunology , Toll-Like Receptor 2/immunology , Animals , Cell Line , Chemokines/immunology , Chemokines/metabolism , Cytokines/metabolism , Epithelial Cells/cytology , Epithelial Cells/immunology , Host-Pathogen Interactions , Humans , NF-kappa B/metabolism
7.
Infect Immun ; 78(12): 5314-23, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20937766

ABSTRACT

The human airway epithelium is constantly exposed to microbial products from colonizing organisms. Regulation of Toll-like receptor (TLR) expression and specific interactions with bacterial ligands is thought to mitigate exacerbation of inflammatory processes induced by the commensal flora in these cells. The genus Neisseria comprises pathogenic and commensal organisms that colonize the human nasopharynx. Neisseria lactamica is not associated with disease, but N. meningitidis occasionally invades the host, causing meningococcal disease and septicemia. Upon colonization of the airway epithelium, specific host cell receptors interact with numerous Neisseria components, including the PorB porin, at the immediate bacterial-host cell interface. This major outer membrane protein is expressed by all Neisseria strains, regardless of pathogenicity, but its amino acid sequence varies among strains, particularly in the surface-exposed regions. The interaction of Neisseria PorB with TLR2 is essential for driving TLR2/TLR1-dependent cellular responses and is thought to occur via the porin's surface-exposed loop regions. Our studies show that N. lactamica PorB is a TLR2 ligand but its binding specificity for TLR2 is different from that of meningococcal PorB. Furthermore, N. lactamica PorB is a poor inducer of proinflammatory mediators and of TLR2 expression in human airway epithelial cells. These effects are reproduced by whole N. lactamica organisms. Since the responsiveness of human airway epithelial cells to colonizing bacteria is in part regulated via TLR2 expression and signaling, commensal organisms such as N. lactamica would benefit from expressing a product that induces low TLR2-dependent local inflammation, likely delaying or avoiding clearance by the host.


Subject(s)
Neisseria lactamica/immunology , Neisseriaceae Infections/immunology , Porins/immunology , Respiratory Mucosa/microbiology , Toll-Like Receptor 2/immunology , Cell Line , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Bacterial/physiology , Humans , Immunity, Cellular/immunology , Immunity, Cellular/physiology , Interleukin-8/immunology , Interleukin-8/physiology , Porins/physiology , Respiratory Mucosa/immunology , Signal Transduction/immunology , Signal Transduction/physiology , Toll-Like Receptor 1/immunology , Toll-Like Receptor 1/physiology , Toll-Like Receptor 2/physiology
8.
J Immunol ; 185(6): 3652-60, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20709949

ABSTRACT

Neisseria lactamica is a commensal bacteria that colonizes the human upper respiratory tract mucosa during early childhood. In contrast to the closely related opportunistic pathogen Neisseria meningitidis, there is an absence of adaptive cell-mediated immunity to N. lactamica during the peak age of carriage. Instead, outer membrane vesicles derived from N. lactamica mediate a B cell-dependent proliferative response in mucosal mononuclear cells that is associated with the production of polyclonal IgM. We demonstrate in this study that this is a mitogenic human B cell response that occurs independently of T cell help and any other accessory cell population. The ability to drive B cell proliferation is a highly conserved property and is present in N. lactamica strains derived from diverse clonal complexes. CFSE staining of purified human tonsillar B cells demonstrated that naive IgD(+) and CD27(-) B cells are selectively induced to proliferate by outer membrane vesicles, including the innate CD5(+) subset. Neither purified lipooligosaccharide nor PorB from N. lactamica is likely to be responsible for this activity. Prior treatment of B cells with pronase to remove cell-surface Ig or treatment with BCR-specific Abs abrogated the proliferative response to N. lactamica outer membrane vesicles, suggesting that this mitogenic response is dependent upon the BCR.


Subject(s)
Antibodies, Bacterial/physiology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Bacterial Outer Membrane Proteins/physiology , Cell Proliferation , Neisseria lactamica/immunology , Receptors, Antigen, B-Cell/physiology , Resting Phase, Cell Cycle/immunology , Adaptive Immunity/immunology , Adolescent , Adult , B-Lymphocyte Subsets/cytology , Cells, Cultured , Child , Child, Preschool , Humans , Immunoglobulin D/physiology , Immunoglobulin M/physiology , Neisseria meningitidis/immunology , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Palatine Tonsil/microbiology , Porins/physiology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology
9.
Clin Vaccine Immunol ; 16(8): 1113-20, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19553555

ABSTRACT

Natural immunity to meningococcal disease in young children is associated epidemiologically with carriage of commensal Neisseria species, including Neisseria lactamica. We have previously demonstrated that outer membrane vesicles (OMVs) from N. lactamica provide protection against lethal challenge in a mouse model of meningococcal septicemia. We evaluated the safety and immunogenicity of an N. lactamica OMV vaccine in a phase I placebo-controlled, double-blinded clinical trial. Ninety-seven healthy young adult male volunteers were randomized to receive three doses of either an OMV vaccine or an Alhydrogel control. Subsequently, some subjects who had received the OMV vaccine also received a fourth dose of OMV vaccine, 6 months after the third dose. Injection site reactions were more frequent in the OMV-receiving group, but all reactions were mild or moderate in intensity. The OMV vaccine was immunogenic, eliciting rises in titers of immunoglobulin G (IgG) against the vaccine OMVs, together with a significant booster response, as determined by an enzyme-linked immunosorbent assay. Additionally, the vaccine induced modest cross-reactive immunity to six diverse strains of serogroup B Neisseria meningitidis, including IgG against meningococcal OMVs, serum bactericidal antibodies, and opsonophagocytic activity. The percentages of subjects showing > or =4-fold rises in bactericidal antibody titer obtained were similar to those previously reported for the Norwegian meningococcal OMV vaccine against the same heterologous meningococcal strain panel. In conclusion, this N. lactamica OMV vaccine is safe and induces a weak but broad humoral immune response to N. meningitidis.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Meningococcal Vaccines/adverse effects , Meningococcal Vaccines/immunology , Neisseria lactamica/immunology , Secretory Vesicles/immunology , Adjuvants, Immunologic/administration & dosage , Adolescent , Adult , Aluminum Hydroxide/administration & dosage , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Cross Reactions , Double-Blind Method , Enzyme-Linked Immunosorbent Assay/methods , Human Experimentation , Humans , Immunization, Secondary/methods , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Mice , Middle Aged , Neisseria meningitidis/immunology , Opsonin Proteins/blood , Opsonin Proteins/immunology , Placebos/administration & dosage , Young Adult
10.
Vaccine ; 27(25-26): 3422-8, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19460600

ABSTRACT

The aim of the present study was to investigate the immune response to native outer membrane vesicles (NOMVs) of Neisseria lactamica with and without Bordetella pertussis (BP) as adjuvant in intranasal (i.n./i.m) immunization. N. lactamica NOMVs delivered intranasally (i.n) to BALB/c mice in a final volume of 5microl that was gradually introduced with a micropipette, Animals received 1, 2, 3, or 4 doses of antigens at 3, 7, 9 and 12 days after birth. On the 35th day, the animals were immunized intramuscularly (i.m.) with (NOMV) of N. lactamica. The prime-booster strategy using NOMV of N. lactamica with BP as adjuvant in the primer (i.n.) and booster (i.m.) is an effective immunization protocol for inducing humoral immune responses producing IgG antibodies of intermediate to high avidity.


Subject(s)
Bacterial Vaccines/immunology , Immunization , Neisseria lactamica/immunology , Animals , Animals, Newborn , Antibodies, Bacterial/blood , Bacterial Outer Membrane Proteins/immunology , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Female , Immunization, Secondary , Mice , Mice, Inbred BALB C
11.
Vaccine ; 27(25-26): 3429-34, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19460601

ABSTRACT

We consider a model for the transmission dynamics of Neisseria meningitidis which incorporates the humoral immunity hypothesis in an explicit way. The "humoral immunity" hypothesis states that individuals will experience significantly different risks of invasive disease depending on whether they are experiencing their first infection episode, or a subsequent one. The model is fitted to the Stonehouse-Danbury carriage data and to UK disease data. For serogroup C (B) the risk of disease during the first infection episode results to be 100 (400) times higher compared to subsequent ones. Moreover the best-fit corresponds to the situation where N. lactamica essentially always confers cross-protection.


Subject(s)
Antibodies, Bacterial/blood , Meningococcal Infections/immunology , Meningococcal Infections/transmission , Adolescent , Adult , Age Factors , Aged , Child , Child, Preschool , Humans , Infant , Infant, Newborn , Meningococcal Infections/etiology , Middle Aged , Models, Theoretical , Neisseria lactamica/immunology
12.
J Immunol ; 182(4): 2231-40, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19201877

ABSTRACT

The normal flora that colonizes the mucosal epithelia has evolved diverse strategies to evade, modulate, or suppress the immune system and avoid clearance. Neisseria lactamica and Neisseria meningitidis are closely related obligate inhabitants of the human upper respiratory tract. N. lactamica is a commensal but N. meningitidis is an opportunistic pathogen that occasionally causes invasive disease such as meningitis and septicemia. We demonstrate that unlike N. meningitidis, N. lactamica does not prime the development of mucosal T or B cell memory during the peak period of colonization. This cannot be explained by the induction of peripheral tolerance or regulatory CD4(+)CD25(+) T cell activity. Instead, N. lactamica mediates a B cell-dependent mitogenic proliferative response that is absent to N. meningitidis. This mitogenic response is associated with the production of T cell-independent polyclonal IgM that we propose functions by shielding colonizing N. lactamica from the adaptive immune system, maintaining immunological ignorance in the host. We conclude that, in contrast to N. meningitidis, N. lactamica maintains a commensal relationship with the host in the absence of an adaptive immune response. This may prolong the period of susceptibility to colonization by both pathogenic and nonpathogenic Neisseria species.


Subject(s)
Immunity, Mucosal/immunology , Nasopharynx/immunology , Nasopharynx/microbiology , Neisseria lactamica/immunology , Neisseria meningitidis/immunology , Adolescent , Antigens, CD19/immunology , B-Lymphocytes/immunology , Cell Proliferation , Child , Child, Preschool , Flow Cytometry , Humans , Immunologic Memory/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Leukocyte Common Antigens/immunology , Nasal Mucosa/immunology , Nasal Mucosa/microbiology , Neisseriaceae Infections/immunology , Respiratory Tract Infections/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology
13.
Hybridoma (Larchmt) ; 27(5): 387-93, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18803504

ABSTRACT

Abstract Neisseria lactamica, a commensal bacterium that is non-pathogenic to humans and is usually found in the upper respiratory tract of children, is closely related to the pathogenic species Neisseria meningitidis. Colonization by Neisseria lactamica can be responsible for the development of natural immunity to meningococcal infection in childhood, when rates of meningococcal carriers are low. These features suggest that N. lactamica components can be key elements in the production of a new vaccine for N. meningitidis. The production of monoclonal antibodies for N. lactamica is an important tool in the selection of new antigens for the preparation of a vaccine for N. meningitidis B.


Subject(s)
Antibodies, Monoclonal/immunology , Neisseria lactamica/immunology , Neisseria meningitidis/immunology , Vaccines/immunology , Animals , Humans , Mice
14.
Microbiology (Reading) ; 154(Pt 5): 1525-1534, 2008 May.
Article in English | MEDLINE | ID: mdl-18451061

ABSTRACT

One potential vaccine strategy in the fight against meningococcal disease involves the exploitation of outer-membrane components of Neisseria lactamica, a commensal bacterium closely related to the meningococcus, Neisseria meningitidis. Although N. lactamica shares many surface structures with the meningococcus, little is known about the antigenic diversity of this commensal bacterium or the antigenic relationships between N. lactamica and N. meningitidis. Here, the N. lactamica porin protein (Por) was examined and compared to the related PorB antigens of N. meningitidis, to investigate potential involvement in anti-meningococcal immunity. Relationships among porin sequences were determined using distance-based methods and F(ST), and maximum-likelihood analyses were used to compare the selection pressures acting on the encoded proteins. These analyses demonstrated that the N. lactamica porin was less diverse than meningococcal PorB and although it was subject to positive selection, this was not as strong as the positive selection pressures acting on the meningococcal porin. In addition, the N. lactamica porin gene sequences and the protein sequences of the loop regions predicted to be exposed to the human immune system were dissimilar to the corresponding sequences in the meningococcus. This suggests that N. lactamica Por, contrary to previous suggestions, may have limited involvement in the development of natural immunity to meningococcal disease and might not be effective as a meningococcal vaccine component.


Subject(s)
Neisseria lactamica/genetics , Neisseria meningitidis/genetics , Polymorphism, Genetic , Porins/genetics , Amino Acid Sequence , Cluster Analysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Female , Humans , Infant , Male , Models, Molecular , Molecular Sequence Data , Neisseria lactamica/immunology , Neisseria lactamica/isolation & purification , Neisseria meningitidis/immunology , Pharynx/microbiology , Porins/immunology , Selection, Genetic , Sequence Analysis, DNA , Sequence Homology, Amino Acid
15.
Vaccine ; 26(6): 786-96, 2008 Feb 06.
Article in English | MEDLINE | ID: mdl-18191311

ABSTRACT

Porins from pathogenic Neisseriae are among several bacterial products with immune adjuvant activity. Neisseria meningitidis (Nme) PorB, has been shown to induce immune cells activation in a TLR2-dependent manner and acts as a vaccine immune adjuvant. The PorB porin from Neisseria lactamica (Nlac), a common nasopharyngeal commensal, shares significant structural and functional similarities with Nme PorB. In this work we ask whether the immune adjuvant ability of porins from pathogenic Neisserial strains is a characteristic shared with porins from non-pathogenic Neisserial species or whether it is unique for bacterial products derived from microorganisms capable of inducing inflammation and disease. We evaluated the potential immune adjuvant effect of Nlac PorB in mice using ovalbumin (OVA) as a prototype antigen. Immunization with Nlac PorB/OVA induced high OVA-specific IgG and IgM titers compared to OVA alone, similar to other adjuvants such as Nme PorB and alum. High titers of IgG1 and IgG2b were detected as well as production of IL-4, IL-10, IL-12 and INF-gamma in response to Nlac PorB, consistent with induction of both a Th1-type and a Th2-type immune response. OVA-specific proliferation was also determined in splenocytes from Nlac PorB/OVA-immunized mice. In addition, B cell activation in vitro and cytokine production in response to Nlac PorB was found to be mediated by TLR2, in a similar manner to Nme PorB.


Subject(s)
Adjuvants, Immunologic , Antibodies/blood , Immunization , Neisseria lactamica/immunology , Ovalbumin/immunology , Porins/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies/immunology , Antibody Specificity , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Immunization Schedule , Immunoglobulin G/blood , Immunoglobulin G/immunology , Injections, Subcutaneous , Interferon-gamma/biosynthesis , Interleukins/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Ovalbumin/administration & dosage , Porins/administration & dosage , Spleen/immunology , Th1 Cells/metabolism , Th2 Cells/metabolism
16.
Hum Vaccin ; 4(1): 23-30, 2008.
Article in English | MEDLINE | ID: mdl-17921703

ABSTRACT

Serogroup B strains are now responsible for over 80% of meningococcal disease in the UK and no suitable vaccine is available that confers universal protection against all serogroup B strains. Neisseria lactamica shares many antigens with the meningococcus, except capsule and the surface protein PorA. Many of these antigens are thought to be responsible for providing cross-protective immunity to meningococcal disease. We have developed an N. lactamica vaccine using methods developed for meningococcal outer membrane vesicle (OMV) vaccines. The major antigenic components were identified by excision of 11 major protein bands from an SDS-PAGE gel, followed by mass spectrometric identification. These bands contained at least 22 proteins identified from an unassembled N. lactamica genome, 15 of which having orthologues in published pathogenic Neisseria genomes. Western blotting revealed that most of these bands were immunogenic, and antibodies to these proteins generally cross-reacted with N. meningitidis proteins. Sera from mice and rabbits immunized with either N. lactamica or N. meningitidis OMVs produced comparable cross-reactive ELISA titres against OMVs prepared from a panel of diverse meningococcal strains. Mice immunized with either N. meningitidis or N. lactamica OMVs showed no detectable serum bactericidal activity against the panel of target strains except N. meningitidis OMV sera against the homologous strain. Similarly, rabbit antisera to N. lactamica OMVs elicited little or no bactericidal antibodies against the panel of serogroup B meningococcal strains. However, such antisera did mediate opsonophagocytosis, suggestingthat this may did mediate opsonophagocytosis, suggesting that this may be a mechanism by which this vaccine protects in a mouse model of meningococcal bacteraemia.


Subject(s)
Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Immunity, Mucosal/drug effects , Meningococcal Vaccines/immunology , Neisseria lactamica/genetics , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Humans , Immunity, Mucosal/immunology , Meningococcal Infections/prevention & control , Mice , Neisseria lactamica/immunology , Rabbits
17.
Infect Immun ; 75(9): 4449-55, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17620353

ABSTRACT

Pathogenic Neisseria bacteria naturally liberate outer membrane "blebs," which are presumed to contribute to pathology, and the detergent-extracted outer membrane vesicles (OMVs) from Neisseria meningitidis are currently employed as meningococcal vaccines in humans. While the composition of these vesicles reflects the bacteria from which they are derived, the functions of many of their constituent proteins remain unexplored. The neisserial colony opacity-associated Opa proteins function as adhesins, the majority of which mediate bacterial attachment to human carcinoembryonic antigen-related cellular adhesion molecules (CEACAMs). Herein, we demonstrate that the Opa proteins within OMV preparations retain the capacity to bind the immunoreceptor tyrosine-based inhibitory motif-containing coinhibitory receptor CEACAM1. When CD4(+) T lymphocytes were exposed to OMVs from Opa-expressing bacteria, their activation and proliferation in response to a variety of stimuli were effectively halted. This potent immunosuppressive effect suggests that localized infection will generate a "zone of inhibition" resulting from the diffusion of membrane blebs into the surrounding tissues. Moreover, it demonstrates that OMV-based vaccines must be developed from strains that lack CEACAM1-binding Opa variants.


Subject(s)
Antigens, CD/metabolism , Bacterial Adhesion , Bacterial Outer Membrane Proteins/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/metabolism , Immunosuppression Therapy , Neisseria meningitidis/immunology , Receptors, Immunologic/metabolism , Amino Acid Motifs/immunology , Antigens, CD/physiology , Bacterial Outer Membrane Proteins/physiology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Adhesion Molecules/physiology , Cell Wall/physiology , Cells, Cultured , Growth Inhibitors/metabolism , Growth Inhibitors/physiology , Humans , Jurkat Cells , Neisseria lactamica/immunology , Receptors, Immunologic/physiology , Tyrosine/metabolism
18.
Hum Vaccin ; 2(2): 68-73, 2006.
Article in English | MEDLINE | ID: mdl-17012888

ABSTRACT

The UK meningococcal serogroup C conjugate (MCC) vaccine program has successfully controlled serogroup C disease, due to high vaccine effectiveness and substantial herd immunity. However, children immunised at 2, 3 and 4 months of age receive only short-term direct protection and may be at risk of disease 15 months after vaccination. To investigate this we applied a mathematical model to predict the future epidemiology of serogroup C disease, with and without changes to the immunization schedule. Only a few cases of serogroup C disease were predicted to occur over the next few years because of persisting herd immunity, even without a change to the vaccine schedule. The inclusion of a booster dose is likely to improve the impact of the MCC program and reducing the number of doses in infancy will improve cost-effectiveness and create space in the schedule for the addition of other vaccines.


Subject(s)
Meningococcal Infections/immunology , Meningococcal Infections/prevention & control , Meningococcal Vaccines/immunology , Adolescent , Algorithms , Carrier State/epidemiology , Carrier State/immunology , Cost-Benefit Analysis , England/epidemiology , Female , Forecasting , Humans , Immunity, Herd , Immunization Schedule , Male , Meningococcal Infections/epidemiology , Meningococcal Vaccines/economics , Models, Statistical , Neisseria lactamica/immunology , Vaccines, Conjugate/economics , Vaccines, Conjugate/immunology , Wales/epidemiology
19.
Infect Immun ; 74(11): 6467-78, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16954390

ABSTRACT

The rationale for the present study was to determine how different species of bacteria interact with cells of the human meninges in order to gain information that would have broad relevance to understanding aspects of the innate immune response in the brain. Neisseria lactamica is an occasional cause of meningitis in humans, and in this study we investigated the in vitro interactions between N. lactamica and cells derived from the leptomeninges in comparison with the closely related organism Neisseria meningitidis, a major cause of meningitis worldwide. N. lactamica adhered specifically to meningioma cells, but the levels of adherence were generally lower than those with N. meningitidis. Meningioma cells challenged with N. lactamica and N. meningitidis secreted significant amounts of the proinflammatory cytokine interleukin-6 (IL-6), the C-X-C chemokine IL-8, and the C-C chemokines monocyte chemoattractant protein 1 (MCP-1) and RANTES, but it secreted very low levels of the cytokine growth factor granulocyte-macrophage colony-stimulating factor (GM-CSF). Thus, meningeal cells are involved in the innate host response to Neisseria species that are capable of entering the cerebrospinal fluid. The levels of IL-8 and MCP-1 secretion induced by both bacteria were essentially similar. By contrast, N. lactamica induced significantly lower levels of IL-6 than N. meningitidis. Challenge with the highest concentration of N. lactamica (10(8) CFU) induced a small but significant down-regulation of RANTES secretion, which was not observed with lower concentrations of bacteria. N. meningitidis (10(6) to 10(8) CFU) also down-regulated RANTES secretion, but this effect was significantly greater than that observed with N. lactamica. Although both bacteria were unable to invade meningeal cells directly, host cells remained viable on prolonged challenge with N. lactamica, whereas N. meningitidis induced death; the mechanism was overwhelming necrosis with no significant apoptosis. It is likely that differential expression of modulins between N. lactamica and N. meningitidis contributes to these observed differences in pathogenic potential.


Subject(s)
Meninges/immunology , Meninges/microbiology , Neisseria lactamica/immunology , Neisseria meningitidis/immunology , Cells, Cultured , Inflammation/immunology , Inflammation/microbiology , Meningeal Neoplasms/immunology , Meningeal Neoplasms/microbiology , Meningeal Neoplasms/pathology , Meninges/pathology , Meningioma/immunology , Meningioma/microbiology , Meningioma/pathology
20.
Infect Immun ; 74(11): 6348-55, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16966413

ABSTRACT

Natural immunity against Neisseria meningitidis is thought to develop following nasopharyngeal colonization with this bacterium or other microbes expressing cross-reactive antigens. Neisseria lactamica is a commensal of the upper respiratory tract which is often carried by infants and young children; epidemiological evidence indicates that colonization with this bacterium can elicit serum bactericidal activity (SBA) against Neisseria meningitidis, the most validated correlate of protective immunity. Here we demonstrate experimentally that immunization of mice with live N. lactamica protects animals against lethal meningococcal challenge and that some, but not all, strains of N. lactamica elicit detectable SBA in immunized animals regardless of the serogroup of N. meningitidis. While it is unlikely that immunization with live N. lactamica will be implemented as a vaccine against meningococcal disease, understanding the basis for the induction of cross-protective immunity and SBA should be valuable in the design of subunit vaccines for the prevention of this important human infection.


Subject(s)
Antibodies, Bacterial/blood , Meningococcal Infections/immunology , Meningococcal Infections/prevention & control , Meningococcal Vaccines/immunology , Neisseria lactamica/immunology , Serum Bactericidal Test , Animals , Antibodies, Bacterial/biosynthesis , Disease Models, Animal , Female , HL-60 Cells , Humans , Meningococcal Infections/blood , Meningococcal Infections/microbiology , Meningococcal Vaccines/administration & dosage , Mice , Mice, Inbred BALB C , Neisseria meningitidis/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...