Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Microbes Infect ; 18(10): 627-638, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27373686

ABSTRACT

Neisseria meningitidis (NM) is an opportunistic gram-negative human pathogen that colonizes the human nasopharyngeal epithelium. Asymptomatic carriage is common, but some meningococcal strains can invade nasopharyngeal epithelial cells and proceed to cause severe and often fatal infections. Invasion is predominantly driven by expression of bacterial virulence factors and host cell cognate receptors for bacterial recognition. Porins are among the Neisserial components involved in host cell activation and bacterial internalization processes. Similar to other virulence factors, porins present antigenic and structure variability among strains. Such sequence variability in the surface-exposed loop regions has been correlated to bacterial invasiveness and to variability in host cell responses via Toll-like receptor 2 (TLR2). Here, we examined whether TLR2 signaling by porins influences recovery of intracellular Neisseriae from epithelial cells in vitro. Our results show that TLR2 stimulation, either by the organism or exogenously, generally enhances Neisseriae internalization by epithelial cells. TLR2-driven intracellular signaling via ERK1/2, JNK and particularly NF-κB plays a role in this process. Based on these results, it is possible that expression of porin sequence variants that strongly induce TLR2 activation may be a mechanism to enhance the invasive features of pathogenic Neisseriae strains.


Subject(s)
Endocytosis , Epithelial Cells/microbiology , Host-Pathogen Interactions , Neisseria meningitidis/physiology , Porins/metabolism , Toll-Like Receptor 2/metabolism , Virulence Factors/metabolism , Cells, Cultured , Humans , MAP Kinase Signaling System , NF-kappa B/metabolism
2.
Vaccines (Basel) ; 2(2): 323-53, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-26344622

ABSTRACT

Vaccine efficacy is optimized by addition of immune adjuvants. However, although adjuvants have been used for over a century, to date, only few adjuvants are approved for human use, mostly aimed at improving vaccine efficacy and antigen-specific protective antibody production. The mechanism of action of immune adjuvants is diverse, depending on their chemical and molecular nature, ranging from non-specific effects (i.e., antigen depot at the immunization site) to specific activation of immune cells leading to improved host innate and adaptive responses. Although the detailed molecular mechanism of action of many adjuvants is still elusive, the discovery of Toll-like receptors (TLRs) has provided new critical information on immunostimulatory effect of numerous bacterial components that engage TLRs. These ligands have been shown to improve both the quality and the quantity of host adaptive immune responses when used in vaccine formulations targeted to infectious diseases and cancer that require both humoral and cell-mediated immunity. The potential of such TLR adjuvants in improving the design and the outcomes of several vaccines is continuously evolving, as new agonists are discovered and tested in experimental and clinical models of vaccination. In this review, a summary of the recent progress in development of TLR adjuvants is presented.

3.
J Struct Biol ; 185(3): 440-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24361688

ABSTRACT

Among all Neisseriae species, Neisseria meningitidis and Neisseria gonorrhoeae are the only human pathogens, causative agents of bacterial meningitis and gonorrhoea, respectively. PorB, a pan-Neisseriae trimeric porin that mediates diffusive transport of essential molecules across the bacterial outer membrane, is also known to activate host innate immunity via Toll-like receptor 2 (TLR2)-mediated signaling. The molecular mechanism of PorB binding to TLR2 is not known, but it has been hypothesized that electrostatic interactions contribute to ligand/receptor binding. Strain-specific sequence variability in the surface-exposed loops of PorB which are potentially implicated in TLR2 binding, may explain the difference in TLR2-mediated cell activation in vitro by PorB homologs from the commensal Neisseriae lactamica and the pathogen N. meningitidis. Here, we report a comparative structural analysis of PorB from N. meningitidis serogroup B strain 8765 (63% sequence homology with PorB from N. meningitidis serogroup W135) and a mutant in which amino acid substitutions in the extracellular loop 7 lead to significantly reduced TLR2-dependent activity in vitro. We observe that this mutation both alters the loop conformation and causes dramatic changes of electrostatic surface charge, both of which may affect TLR2 recognition and signaling.


Subject(s)
Neisseria meningitidis/metabolism , Porins/chemistry , Porins/metabolism , Toll-Like Receptor 2/chemistry , Toll-Like Receptor 2/metabolism , Amino Acid Sequence , Humans , Molecular Sequence Data , Protein Structure, Secondary , Sequence Homology, Amino Acid
4.
Microbes Infect ; 15(13): 920-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23999313

ABSTRACT

Vaccines formulated with the Chlamydia muridarum native major outer membrane protein (nMOMP) have so far been shown to elicit the most robust protection against this pathogen. nMOMP is a membrane protein and therefore, detergents are used to keep it in solution. Detergents however, have toxic effects. To address this limitation, we tested a nMOMP proteosome vaccine and compared its ability to elicit protection against nMOMP solubilized in the detergent Z3-14. The two preparations were formulated with or without CpG + Montanide (C/M). As a control antigen we used ovalbumin. Mice vaccinated with nMOMP developed strong humoral and cell mediated Chlamydia-specific immune responses. Based on the IgG2a/IgG1 levels in serum and amounts of IFN-γ in splenocytes supernatants the immune responses were predominantly Th1-biased. The animals were subsequently challenged intranasally with 2 × 10(3)Chlamydia inclusion forming units (IFU) and the course of the infection was followed for 10 days when the mice were euthanized. Based on changes in body weight, weight of the lungs and number of IFU recovered from the lungs, mice immunized with nMOMP-Ps and nMOMP + Z3-14 adjuvanted with C/M showed the most robust protection. In summary, nMOMP-Ps should be considered as Chlamydia vaccine candidates.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Chlamydia muridarum/immunology , Pneumonia, Bacterial/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Bacterial/blood , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Vaccines/administration & dosage , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Disease Models, Animal , Immunoglobulin G/blood , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Mannitol/administration & dosage , Mannitol/analogs & derivatives , Mice , Oleic Acids/administration & dosage , Oligodeoxyribonucleotides/administration & dosage , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Vaccination/methods
5.
Infect Immun ; 81(1): 303-10, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23132491

ABSTRACT

Chlamydia trachomatis is the most common sexually transmitted bacterial pathogen and the etiologic agent of blinding trachoma. Intracellular signaling pathways leading to host cell inflammation and innate immunity to Chlamydia include those mediated by Toll-like receptors (TLRs) and nucleotide binding oligomerization domain 1 (Nod1) protein. In epithelial cells, TLR-dependent signaling contributes to local immune responses via induction of inflammatory mediators. There is evidence that TLR3, TLR4, and, particularly, TLR2 are critical for Chlamydia-mediated host cell activation and pathology. Despite the importance of TLR2, major chlamydial TLR2 antigens have not been identified so far. Numerous bacterial porins are known TLR2 agonists, i.e., porins from Neisseriae, Shigella, Salmonella, Haemophilus influenzae, and Fusobacterium nucleatum, which share structural and functional similarities with the chlamydial major outer membrane protein (MOMP), a strong antigen candidate for a potential vaccine against C. trachomatis. We describe the ability of purified, detergent-free MOMP to signal via TLR2 in vitro in TLR-overexpressing cells and TLR2-competent human reproductive tract epithelial cell lines. Using MOMP formed in pure protein micelles (proteosomes), we show the induction of TLR2-dependent interleukin-8 (IL-8) and IL-6 secretion in vitro, the involvement of TLR1 as a TLR2 coreceptor, and the activation of both NF-κB and mitogen-activated protein (MAP) kinase intracellular pathways. Interestingly, MOMP proteosomes induce cytokine secretion in endocervical epithelial cells (End/E6E7) but not in urethral epithelial cells (THUECs). A detailed understanding of the TLR2-dependent molecular mechanisms that characterize the effect of MOMP proteosomes on host cells may provide new insights for its successful development as an immunotherapeutic target against Chlamydia.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Chlamydia Infections/metabolism , Chlamydia trachomatis/metabolism , Proteasome Endopeptidase Complex/metabolism , Toll-Like Receptor 2/metabolism , Cell Line , Cell Line, Tumor , Chlamydia Infections/microbiology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , HEK293 Cells , HeLa Cells , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Micelles , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Porins/metabolism , Signal Transduction , Toll-Like Receptor 1/metabolism
6.
Infect Immun ; 80(10): 3417-28, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22825445

ABSTRACT

Toll-like receptors (TLRs) play a major role in host mucosal and systemic defense mechanisms by recognizing a diverse array of conserved pathogen-associated molecular patterns (PAMPs). TLR2, with TLR1 and TLR6, recognizes structurally diverse bacterial products such as lipidated factors (lipoproteins and peptidoglycans) and nonlipidated proteins, i.e., bacterial porins. PorB is a pan-neisserial porin expressed regardless of organisms' pathogenicity. However, commensal Neisseria lactamica organisms and purified N. lactamica PorB (published elsewhere as Nlac PorB) induce TLR2-dependent proinflammatory responses of lower magnitude than N. meningitidis organisms and N. meningitidis PorB (published elsewhere as Nme PorB). Both PorB types bind to TLR2 in vitro but with different apparent specificities. The structural and molecular details of PorB-TLR2 interaction are only beginning to be unraveled and may be due to electrostatic attraction. PorB molecules have significant strain-specific sequence variability within surface-exposed regions (loops) putatively involved in TLR2 interaction. By constructing chimeric recombinant PorB loop mutants in which surface-exposed loop residues have been switched between N. lactamica PorB and N. meningitidis PorB, we identified residues in loop 5 and loop 7 that influence TLR2-dependent cell activation using HEK cells and BEAS-2B cells. These loops are not uniquely responsible for PorB interaction with TLR2, but NF-κB and MAP kinases signaling downstream of TLR2 recognition are likely influenced by a hypothetical "TLR2-binding signature" within the sequence of PorB surface-exposed loops. Consistent with the effect of purified PorB in vitro, a chimeric N. meningitidis strain expressing N. lactamica PorB induces lower levels of interleukin 8 (IL-8) secretion than wild-type N. meningitidis, suggesting a role for PorB in induction of host cell activation by whole bacteria.


Subject(s)
Neisseria lactamica/metabolism , Porins/metabolism , Toll-Like Receptor 2/metabolism , Amino Acid Sequence , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , HEK293 Cells , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Models, Molecular , Mutation , Neisseria lactamica/genetics , Porins/chemistry , Porins/genetics , Protein Binding , Protein Conformation , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
7.
Clin Vaccine Immunol ; 19(7): 1093-101, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22623652

ABSTRACT

Many bacterial components selectively activate immune and nonhematopoietic target cells via Toll-like receptor (TLR) signaling; modulation of such host responses defines the immune adjuvant properties of these bacterial products. For example, the outer membrane protein porins from Neisseria, Salmonella, and Shigella are known TLR2 agonists with established systemic and mucosal immune adjuvanticity. Early work indicated that the FomA porin from Fusobacterium nucleatum has immune adjuvant activity in mice. Using a purified recombinant FomA, we have verified its immune stimulatory properties and have defined a role for TLR2 signaling in its in vitro and in vivo activity. FomA induces interleukin 8 (IL-8) secretion and NF-κB-dependent luciferase activity in HEK cells expressing TLR2, IL-6 secretion, and cell surface upregulation of CD86 and major histocompatibility complex (MHC) II in primary B cells from wild-type mice, but it fails to activate cells from TLR2 knockout mice. Accordingly, the immune adjuvant activity of FomA is also TLR2 dependent. In a mouse model of immunization with ovalbumin (OVA), FomA induces enhanced production of OVA-specific IgM and IgG, including IgG1 and IgG2b antibodies, as well as enhanced secretion of IL-10 and IL-6, consistent with a Th2-type adjuvant effect. We also observe a moderate production of anti-FomA antibodies, suggesting that FomA is also immunogenic, a quality that is also TLR2 dependent. Therefore, modulation of host immune responses by FomA may be effective for targeting general host immunity not only to pathogens (as a novel TLR2 adjuvant) but also to F. nucleatum itself (as an antigen), expanding its use as a self-adjuvanted antigen in an immunization strategy against polymicrobial infections, including those by F. nucleatum.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bacterial Outer Membrane Proteins/pharmacology , Toll-Like Receptor 2/agonists , Animals , Antibodies/blood , B7-2 Antigen/biosynthesis , Cells, Cultured , Female , Histocompatibility Antigens Class II/biosynthesis , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Interleukin-10/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Ovalbumin/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...