Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Med Rep ; 15(1): 65-74, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27922699

ABSTRACT

The interaction between Streptococcus pneumoniae (S. pneumoniae) and the mucosal epithelial cells of its host is a prerequisite for pneumococcal disease development, yet the specificity of this interaction between different respiratory cells is not fully understood. In the present study, three areas were examined: i) The capability of the encapsulated S. pneumoniae serotype 3 strain (WU2) to adhere to and invade primary nasal­derived epithelial cells in comparison to primary oral­derived epithelial cells, A549 adenocarcinoma cells and BEAS­2B viral transformed bronchial cells; ii) the capability of the unencapsulated 3.8DW strain (a WU2 derivative) to adhere to and invade the same cells over time; and iii) the ability of various genetically­unrelated encapsulated and unencapsulated S. pneumoniae strains to adhere to and invade A549 lung epithelial cells. The results of the present study demonstrated that the encapsulated WU2 strain adhesion to and invasion of primary nasal epithelial cells was greatest, followed by BEAS­2B, A549 and primary oral epithelial cells. By contrast, the unencapsulated 3.8­DW strain invaded oral epithelial cells significantly more efficiently when compared to the nasal epithelial cells. In addition, unencapsulated S. pneumoniae strains adhered to and invaded the A459 cells significantly more efficiently than the encapsulated strains; this is consistent with previously published data. In conclusion, the findings presented in the current study indicated that the adhesion and invasion of the WU2 strain to primary nasal epithelial cells was more efficient compared with the other cultured respiratory epithelial cells tested, which corresponds to the natural course of S. pneumoniae infection and disease development. The target cell preference of unencapsulated strains was different from that of the encapsulated strains, which may be due to the exposure of cell wall proteins.


Subject(s)
Bacterial Adhesion , Pneumococcal Infections/pathology , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , Streptococcus pneumoniae/physiology , Cell Line , Cells, Cultured , Epithelial Cells/microbiology , Epithelial Cells/pathology , Humans , Mouth Mucosa/cytology , Mouth Mucosa/microbiology , Mouth Mucosa/pathology , Nasal Mucosa/cytology , Nasal Mucosa/microbiology , Nasal Mucosa/pathology , Pneumococcal Infections/microbiology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/pathology
2.
PLoS One ; 11(8): e0160779, 2016.
Article in English | MEDLINE | ID: mdl-27580126

ABSTRACT

The function of NCR1 was studied in a model of experimental asthma, classified as a type 1 hypersensitivity reaction, in mice. IgE levels were significantly increased in the serum of OVA immunized NCR1 deficient (NCR1gfp/gfp) mice in comparison to OVA immunized wild type (NCR1+/+) and adjuvant immunized mice. Histological analysis of OVA immunized NCR1gfp/gfp mice revealed no preservation of the lung structure and overwhelming peribronchial and perivascular granulocytes together with mononuclear cells infiltration. OVA immunized NCR+/+ mice demonstrated preserved lung structure and peribronchial and perivascular immune cell infiltration to a lower extent than that in NCR1gfp/gfp mice. Adjuvant immunized mice demonstrated lung structure preservation and no immune cell infiltration. OVA immunization caused an increase in PAS production independently of NCR1 presence. Bronchoalveolar lavage (BAL) revealed NCR1 dependent decreased percentages of eosinophils and increased percentages of lymphocytes and macrophages following OVA immunization. In the OVA immunized NCR1gfp/gfp mice the protein levels of eosinophils' (CCL24) and Th2 CD4+ T-cells' chemoattractants (CCL17, and CCL24) in the BAL are increased in comparison with OVA immunized NCR+/+ mice. In the presence of NCR1, OVA immunization caused an increase in NK cells numbers and decreased NCR1 ligand expression on CD11c+GR1+ cells and decreased NCR1 mRNA expression in the BAL. OVA immunization resulted in significantly increased IL-13, IL-4 and CCL17 mRNA expression in NCR1+/+ and NCR1gfp/gfp mice. IL-17 and TNFα expression increased only in OVA-immunized NCR1+/+mice. IL-6 mRNA increased only in OVA immunized NCR1gfp/gfp mice. Collectively, it is demonstrated that NCR1 dampens allergic eosinophilic airway inflammation.


Subject(s)
Antigens, Ly/immunology , Asthma/immunology , Lung/immunology , Natural Cytotoxicity Triggering Receptor 1/immunology , Th2 Cells/immunology , Animals , Antigens, Ly/genetics , Asthma/chemically induced , Asthma/genetics , Asthma/pathology , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Lung/pathology , Mice , Mice, Transgenic , Natural Cytotoxicity Triggering Receptor 1/genetics , Th2 Cells/pathology
3.
PLoS One ; 11(3): e0150320, 2016.
Article in English | MEDLINE | ID: mdl-26990554

ABSTRACT

In Streptococcus pneumonia, phosphoenolpyruvate protein phosphotransferase (PtsA) is an intracellular protein of the monosaccharide phosphotransferase systems. Biochemical and immunostaining methods were applied to show that PtsA also localizes to the bacterial cell-wall. Thus, it was suspected that PtsA has functions other than its main cytoplasmic enzymatic role. Indeed, recombinant PtsA and anti-rPtsA antiserum were shown to inhibit adhesion of S. pneumoniae to cultured human lung adenocarcinoma A549 cells. Screening of a combinatorial peptide library expressed in a filamentous phage with rPtsA identified epitopes that were capable of inhibiting S. pneumoniae adhesion to A549 cells. The insert peptides in the phages were sequenced, and homologous sequences were found in human BMPER, multimerin1, protocadherin19, integrinß4, epsin1 and collagen type VIIα1 proteins, all of which can be found in A549 cells except the latter. Six peptides, synthesized according to the homologous sequences in the human proteins, specifically bound rPtsA in the micromolar range and significantly inhibited pneumococcal adhesion in vitro to lung- and tracheal-derived cell lines. In addition, the tested peptides inhibited lung colonization after intranasal inoculation of mice with S. pneumoniae. Immunization with rPtsA protected the mice against a sublethal intranasal and a lethal intravenous pneumococcal challenge. In addition, mouse anti rPtsA antiserum reduced bacterial virulence in the intravenous inoculation mouse model. These findings showed that the surface-localized PtsA functions as an adhesin, PtsA binding peptides derived from its putative target molecules can be considered for future development of therapeutics, and rPtsA should be regarded as a candidate for vaccine development.


Subject(s)
Cell Wall/enzymology , Phosphoenolpyruvate Sugar Phosphotransferase System/metabolism , Phosphotransferases (Nitrogenous Group Acceptor)/metabolism , Pneumococcal Vaccines/immunology , Streptococcus pneumoniae/enzymology , Adhesins, Bacterial/physiology , Cell Line, Tumor , Child, Preschool , Flow Cytometry , Humans , Streptococcus pneumoniae/immunology
4.
Int J Mol Med ; 37(4): 1127-38, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26935978

ABSTRACT

Streptococcus pneumoniae (S. pneumoniae) is a major pathogen worldwide. The currently available polysaccharide-based vaccines significantly reduce morbidity and mortality. However, the inherent disadvantages of the currently available polysaccharide-based vaccines have motivated the search for other bacterial immunogens capable of eliciting a protective immune response against S. pneumoniae. Fructose-1,6-bisphosphate aldolase (FBA) is a glycolytic enzyme, which was found to localize to the bacterial surface, where it functions as an adhesin. Previously, immunizing mice with recombinant FBA (rFBA) in the presence of alum elicited a protective immune response against a lethal challenge with S. pneumoniae. Thus, the aim of the present study was to determine the cytokine responses that are indicative of protective immunity following immunization with rFBA. The protective effects against pneumococcal challenge in mice immunized with rFBA with complete Freund's adjuvant (CFA) in the initial immunization and with incomplete Freund's adjuvant (IFA) in booster immunizations surpassed the protective effects observed following immunization with either rFBA + alum or pVACfba. CD4+ T-cells obtained from the rFBA/CFA/IFA/IFA-immunized mice co-cultured with rFBA-pulsed antigen-presenting cells (APCs), exhibited a significantly greater proliferative ability than CD4+ T-cells obtained from the adjuvant-immunized mice co-cultured with rFBA­pulsed APCs. The levels of the Th1-type cytokines, interferon (IFN)-γ, interleukin (IL)-2, tumor necrosis factor (TNF)-α and IL-12, the Th2-type cytokines, IL-4, IL-5 and IL-10, and the Th17-type cytokine, IL-17A, significantly increased within 72 h of the initiation of co-culture with CD4+ T-cells obtained from the rFBA­immunized mice, in comparison with the co-cultures with CD4+ T-cells obtained from the adjuvant-immunized mice. Immunizing mice with rFBA resulted in an IgG1/IgG2 ratio of 41, indicating a Th2 response with substantial Th1 involvement. In addition, rabbit and mouse anti-rFBA antisera significantly protected the mice against a lethal S. pneumoniae challenge in comparison with preimmune sera. Our results emphasize the mixed involvement of the Th1, Th2 and Th17 arms of the immune system in response to immunization with pneumococcal rFBA, a potential vaccine candidate.


Subject(s)
Cytokines/immunology , Fructose-Bisphosphate Aldolase/therapeutic use , Pneumococcal Infections/prevention & control , Streptococcal Vaccines/therapeutic use , Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Animals , Female , Freund's Adjuvant/immunology , Freund's Adjuvant/therapeutic use , Fructose-Bisphosphate Aldolase/immunology , Immunization , Lipids/immunology , Lipids/therapeutic use , Mice , Mice, Inbred BALB C , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Rabbits , Streptococcal Vaccines/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology
5.
PLoS One ; 6(8): e23472, 2011.
Article in English | MEDLINE | ID: mdl-21887255

ABSTRACT

Natural killer (NK) cells serve as a crucial first line of defense against tumors, viral and bacterial infections. We studied the involvement of a principal activating natural killer cell receptor, natural cytotoxicity receptor 1 (NCR1), in the innate immune response to S. pneumoniae infection. Our results demonstrate that the presence of the NCR1 receptor is imperative for the early clearance of S. pneumoniae. We tied the ends in vivo by showing that deficiency in NCR1 resulted in reduced lung NK cell activation and lung IFNγ production at the early stages of S. pneumoniae infection. NCR1 did not mediate direct recognition of S. pneumoniae. Therefore, we studied the involvement of lung macrophages and dendritic cells (DC) as the mediators of NK-expressed NCR1 involvement in response to S. pneumoniae. In vitro, wild type BM-derived macrophages and DC expressed ligands to NCR1 and co-incubation of S. pneumoniae-infected macrophages/DC with NCR1-deficient NK cells resulted in significantly lesser IFNγ levels compared to NCR1-expressing NK cells. In vivo, ablation of lung macrophages and DC was detrimental to the early clearance of S. pneumoniae. NCR1-expressing mice had more potent alveolar macrophages as compared to NCR1-deficient mice. This result correlated with the higher fraction of NCR1-ligand(high) lung macrophages, in NCR1-expressing mice, that had better phagocytic activity compared to NCR1-ligand(dull) macrophages. Overall, our results point to the essential contribution of NK-expressed NCR1 in early response to S. pneumoniae infection and to NCR1-mediated interaction of NK and S. pneumoniae infected-macrophages and -DC.


Subject(s)
Antigens, Ly/metabolism , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 1/metabolism , Streptococcus pneumoniae/immunology , Administration, Intranasal , Animals , Bacterial Load/immunology , Bone Marrow Cells/cytology , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/microbiology , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Ligands , Lung/immunology , Lung/microbiology , Lung/pathology , Lymphocyte Activation/immunology , Macrophage Activation/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis
6.
Int Immunol ; 20(9): 1139-46, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18596024

ABSTRACT

The role of IL-1 in susceptibility to Streptococcus pneumoniae infection was studied in mice deficient in genes of the IL-1 family [i.e. IL-1alpha-/-, IL-1beta-/-, IL-1alpha/beta-/- and IL-1R antagonist (IL-1Ra)-/- mice] following intra-nasal inoculation. Intra-nasal inoculation of S. pneumoniae of IL-1beta-/- and IL-1alpha/beta-/- mice displayed significantly lower survival rates and higher nasopharyngeal and lung bacterial load as compared with control, IL-1alpha-/- and IL-1Ra-/- mice. Treatment of IL-1beta-/- mice with rIL-1beta significantly improved their survival. A significant increase in blood neutrophils was found in control, IL-1alpha-/- and IL-1Ra-/- but not in IL-1beta-/- and IL-1alpha/beta-/- mice. Local infiltrates of neutrophils and relatively preserved organ architecture were observed in the lungs of IL-1alpha-/- and control mice. However, S. pneumoniae-infected IL-1beta-/-, IL-1alpha/beta-/- and IL-1Ra-/- mice demonstrated diffuse pneumonia and tissue damage. Altogether, all three isoforms contribute to protection against S. pneumoniae; our results point to differential role of IL-1alpha and IL-1beta in the pathogenesis and control of S. pneumoniae infection and suggest that IL-1beta has a major role in resistance to primary pneumococcal infection while the role of IL-1alpha is less important.


Subject(s)
Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Pneumonia, Pneumococcal/immunology , Streptococcus pneumoniae/pathogenicity , Animals , Female , Humans , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1alpha/administration & dosage , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Interleukin-1beta/administration & dosage , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Nasopharynx/microbiology , Pneumonia, Pneumococcal/drug therapy , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/mortality , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Streptococcus pneumoniae/isolation & purification
7.
J Infect Dis ; 196(6): 945-53, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17703427

ABSTRACT

Glutamyl tRNA synthetase (GtS) has been found to be among the Streptococcus pneumoniae cell wall-derived proteins that have age-dependent immunogenicity in children. Here, GtS was cloned, expressed, and purified and then was used to immunize 7-week-old BALB/c OlaHsd mice. Serum obtained from mice immunized with recombinant (r) GtS cross-reacted with a 55.9-kDa protein, identified as GtS, in the cell wall fraction derived from genetically and capsularly unrelated strains of S. pneumoniae. Surface localization of GtS was further confirmed using flow cytometry analysis. The rGtS and anti-rGtS antiserum significantly inhibited the adhesion of 3 pairs of encapsulated and unencapsulated strains of S. pneumoniae to A549 cells. Thirty-nine percent of rGtS-immunized mice survived a lethal bacterial challenge, whereas no control mice survived. These results suggest that GtS, an age-dependent S. pneumoniae antigen, is a surface-located adhesin that is capable of inducing a partially protective immune response against S. pneumoniae in mice.


Subject(s)
Bacterial Proteins/immunology , Glutamate-tRNA Ligase/immunology , Membrane Proteins/immunology , Pneumococcal Vaccines/immunology , Streptococcal Infections/prevention & control , Streptococcus pneumoniae/immunology , Adult , Animals , Antibodies, Bacterial/blood , Cell Adhesion/immunology , Cell Line , Child, Preschool , Cloning, Molecular , Female , Gene Expression , Humans , Infant , Mice , Mice, Inbred BALB C , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Streptococcus pneumoniae/enzymology , Survival Analysis , Vaccines, Subunit/immunology
8.
J Infect Dis ; 195(12): 1828-37, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17492599

ABSTRACT

Streptococcus pneumoniae fructose bisphosphate aldolase (FBA) is a cell wall-localized lectin. We demonstrate that recombinant (r) FBA and anti-rFBA antibodies inhibit encapsulated and unencapsulated S. pneumoniae serotype 3 adherence to A549 type II lung carcinoma epithelial cells. A random combinatorial peptide library expressed by filamentous phage was screened with rFBA. Eleven of 30 rFBA-binding phages inhibited 90% of S. pneumoniae adhesion to A549 cells. The insert peptide sequence of 9 of these phages matched the Flamingo cadherin receptor (FCR) when aligned against the human genome. A peptide comprising a putative FBA-binding region of FCR (FCRP) inhibited 2 genetically and capsularly unrelated pairs of encapsulated and unencapsulated S. pneumoniae strains from binding to A549 cells. Moreover, FCRP inhibited S. pneumoniae nasopharyngeal and lung colonization and, possibly, pneumonia development in the mouse intranasal inoculation model system. These data indicate that FBA is an S. pneumoniae adhesin and that FCR is its host receptor.


Subject(s)
Adhesins, Bacterial/isolation & purification , Bacterial Adhesion/physiology , Cadherins/metabolism , Fructose-Bisphosphate Aldolase/immunology , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/metabolism , Adhesins, Bacterial/metabolism , Animals , Antibodies, Bacterial/metabolism , Cadherins/isolation & purification , Cell Line, Tumor , Female , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/metabolism , Humans , Mice , Mice, Inbred BALB C , Nasopharynx/microbiology , Pneumococcal Infections/prevention & control , Rabbits , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Streptococcus pneumoniae/genetics , Time Factors , Virus Attachment
9.
Vaccine ; 24(11): 1868-73, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16316712

ABSTRACT

We have fractionated S. pneumoniae surface proteins into lectin and non-lectin fractions and tested their ability to elicit protective immune responses in the mouse model system. The total cell wall protein fraction (CW-T) was separated into lectin (CW-L), and non-lectin (CW-NL) fractions and used for immunization of mice. Immunized mice were challenged intranasally or intraperitoneally with S. pneumoniae strain WU2 (serotype 3). CW-T, CW-NL and CW-L and adjuvant only vaccination protected 55%, 43%, 44% and 0% of the intranasally challenged mice, respectively and 67%, 86%, 11% and 0% of mice challenged intraperitoneally, respectively. Immunogenic proteins in each fraction were sequenced and identified using MALDI-TOF. CW-L proteins provided a significantly better protection against intranasal inoculation and CW-NL proteins provided a significantly better protection from intraperitoneal inoculation. Proteins identified by sera from mice immunized with the cell-wall derived fractions may constitute candidates for future development of anti S. pneumoniae vaccines.


Subject(s)
Bacterial Proteins/immunology , Lectins/immunology , Membrane Proteins/immunology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/immunology , Streptococcus pneumoniae/immunology , Animals , Bacterial Proteins/administration & dosage , Bacterial Proteins/isolation & purification , Cell Wall/chemistry , Electrophoresis, Gel, Two-Dimensional , Immunoblotting , Lectins/administration & dosage , Lectins/isolation & purification , Membrane Proteins/administration & dosage , Membrane Proteins/isolation & purification , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Survival Analysis
10.
J Infect Dis ; 188(11): 1752-6, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14639547

ABSTRACT

We studied cytokine mRNA expression in Streptococcus pneumoniae carriage, pneumonia, and sepsis in 3-week-old, inbred C57BL/6 and BALB/c mice. Mice were inoculated intranasally with S. pneumoniae serotype 6B, 14, or 3. Survival, bacterial load in the nasopharynx (NP) and lungs, and mRNA levels of tumor necrosis factor (TNF)-alpha, transforming growth factor (TGF)-beta, interleukin (IL)-10, and IL-12 in the spleen were analyzed. No baseline mRNA expression levels were found, except for TNF-alpha in C57BL/6 mice. Serotype 6B caused NP colonization only, a significant (P<.05) increase in the levels of TNF-alpha, and induction of TGF-beta and IL-12 mRNA. Serotype 14 caused NP and nonlethal lung colonization and induction of TGF-beta, IL-10, and IL-12. Serotype 3 caused NP colonization, pneumonia, 35% mortality, and no alteration in the mRNA expression of tested cytokines. Although activation of the immune system culminated in nonlethal disease, evasion of the immune system was associated with detrimental disease.


Subject(s)
Bacteremia/immunology , Carrier State/immunology , Cytokines/genetics , Pneumonia, Pneumococcal/immunology , RNA, Messenger/analysis , Animals , Interleukin-12/genetics , Lung/microbiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nasopharynx/microbiology , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...