Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 361
Filter
Add more filters

Publication year range
1.
Microbiol Immunol ; 65(6): 238-244, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33913531

ABSTRACT

The aim of this study was to assess the modulatory effect of TcpA in the expression of CEACAM1 adhesin molecule and IL-1, IL-8, and TNF-α pro-inflammatory cytokines in the Coculture model of Caco-2/PBMC (peripheral blood mononuclear cell) that can mimic the intestinal milieu. The TcpA gene from Vibrio cholerae ATCC14035 was cloned in pET-28a and transformed into Escherichia coli Bl-21. The recombinant TcpA-His6 protein was expressed and purified using Ni-column chromatography. The sequencing of transformed plasmid and Western blot analysis of purified protein confirmed the identity of rTcp. The cytotoxicity of different concentrations of recombinant protein for human colon carcinoma cell line (human colorectal adenocarcinoma cell [Caco-2 cell]) was assessed by MTT assay and showed viability of 92%, 82%, and 70%, for 10 µg/mL of TcpA after 24, 48, and 72 h, respectively. Co-cultures of Caco-2 and PBMCs were used to mimic the intestinal milieu and treated with different concentrations of rTcpA (1, 5, 10, and 50 µg/mL). Our data showed about 2.04-, 3.37-, 3.68-, and 42.7-fold increase in CEACAM1 gene expression, respectively, compared with the nontreated Caco-2/PBMC Coculture. Moreover, the expression of IL-1, IL-8, and TNF-α genes was significantly increased up to 15.75-, 7.04-, and 80.95-folds, respectively. In conclusion, V. cholerae TcpA induces statistically significant dose-dependent stimulatory effect on TNF-α, IL-,1, and IL-8 pro-inflammatory cytokines expression. Of these, TNF-α was much more affected which, consequently, elevated the CEACAM1 expression level in IECs. This suggests that TcpA protein is a critical effector as an inducer of increased adhesion potential of V. cholera as well as inflammatory responses of host intestinal tissue.


Subject(s)
Cholera Toxin/immunology , Cholera , Fimbriae Proteins/immunology , Leukocytes, Mononuclear/immunology , Vibrio cholerae , Antigens, CD/immunology , Caco-2 Cells , Cell Adhesion Molecules/immunology , Coculture Techniques , Cytokines/immunology , Humans , Leukocytes, Mononuclear/microbiology
2.
Infect Immun ; 88(9)2020 08 19.
Article in English | MEDLINE | ID: mdl-32540869

ABSTRACT

Human rhinovirus (hRV) is frequently detected in the upper respiratory tract, and symptomatic infection is associated with an increased nasopharyngeal bacterial load, with subsequent development of secondary bacterial diseases. Nontypeable Haemophilus influenzae (NTHI) is a commensal bacterial species of the human nasopharynx; however, in the context of prior or concurrent upper respiratory tract viral infection, this bacterium commonly causes multiple diseases throughout the upper and lower respiratory tracts. The present study was conducted to determine the mechanism(s) by which hRV infection promotes the development of NTHI-induced diseases. We showed that hRV infection of polarized primary human airway epithelial cells resulted in increased adherence of NTHI, due in part to augmented expression of CEACAM1 and ICAM1, host cell receptors to which NTHI binds via engagement of multiple adhesins. Antibody blockade of these host cell receptors significantly reduced NTHI adherence. With a specific focus on the NTHI type IV pilus (T4P), which we have previously shown binds to ICAM1, an essential adhesin and virulence determinant, we next showed that T4P-directed antibody blockade significantly reduced NTHI adherence to hRV-infected airway cells and, further, that expression of this adhesin was required for the enhanced adherence observed. Collectively, these data provide a mechanism by which "the common cold" promotes diseases due to NTHI, and they add further support for the use of PilA (the majority subunit of T4P) as a vaccine antigen, since antibodies directed against PilA are expected to limit the notably increased bacterial load associated with hRV coinfection and thereby to prevent secondary NTHI-induced diseases of the respiratory tract.


Subject(s)
Adhesins, Bacterial/immunology , Bacterial Adhesion/immunology , Epithelial Cells/immunology , Fimbriae Proteins/immunology , Haemophilus influenzae/immunology , Host-Pathogen Interactions/immunology , Rhinovirus/immunology , Adhesins, Bacterial/genetics , Antibodies, Neutralizing/pharmacology , Antigens, CD/genetics , Antigens, CD/immunology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Epithelial Cells/microbiology , Epithelial Cells/virology , Fimbriae Proteins/genetics , Gene Expression Regulation/immunology , Haemophilus influenzae/growth & development , Host-Pathogen Interactions/genetics , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Primary Cell Culture , Protein Binding , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Respiratory Mucosa/virology , Rhinovirus/growth & development , Signal Transduction
3.
BMC Biotechnol ; 20(1): 28, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32423439

ABSTRACT

BACKGROUND: Legionella pneumophila (L.pneumophila), a Gram-negative small microorganism, causes hospital-acquired pneumonia especially in immunocompromised patients. Vaccination may be an effective method for preventing L.pneumophila infection. Therefore, it is necessary to develop a better vaccine against this disease. In this study, we developed a recombinant peptidoglycan-associated lipoprotein (PAL)/type IV pilin (PilE)/lagellin (FlaA) DNA vaccine and evaluated its immunogenicity and efficacy to protect against L.pneumophila infection. RESULTS: According to the results, the expression of PAL, PilE, FlaA proteins and PAL/PilE/FlaA fusion protein in 293 cells was confirmed. Immunization with PAL/PilE/FlaA DNA vaccine resulted in highest IgG titer and strongest cytotoxic T-lymphocyte (CTL) response. Furthermore, the histopathological changes in lung tissues of mice challenged with a lethal dose of L.pneumophila were alleviated by PAL/PilE/FlaA DNA vaccine immunization. The production of T-helper-1 (Th1) cytokines (IFNγ, TGF-α, and IL-12), and Th2 cytokines (IL-4 and IL-10) were promoted in PAL/PilE/FlaA DNA vaccine group. Finally, immunization with PAL/PilE/FlaA vaccine raised the survival rate of mice to 100% after challenging with a lethal dose of L.pneumophila for 10 consecutive days. CONCLUSIONS: Our study suggests that the newly developed PAL/PilE/FlaA DNA vaccine stimulates strong humoral and cellular immune responses and may be a potential intervention on L.pneumophila infection.


Subject(s)
Bacterial Vaccines/immunology , Fimbriae Proteins/immunology , Flagellin/immunology , Legionella pneumophila/immunology , Legionnaires' Disease/prevention & control , Lipoproteins/immunology , Peptidoglycan/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Bacterial , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Vaccines/genetics , Cytokines/metabolism , Female , Fimbriae Proteins/genetics , Flagellin/genetics , HEK293 Cells , Humans , Immunity, Cellular , Immunization , Legionella pneumophila/genetics , Lipoproteins/genetics , Lung , Mice , Mice, Inbred BALB C , Peptidoglycan/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccines, DNA/genetics
4.
Microb Pathog ; 138: 103697, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31465785

ABSTRACT

Pseudomonas aeruginosa is a common nosocomial pathogen in burn patients, and rapidly achieves antibiotic resistance, and thus, developing an effective vaccine is critically important for combating P. aeruginosa infection. Flagella and pili play important roles in colonization of P. aeruginosa at the burn wound site and its subsequent dissemination to deeper tissue and organs. In the present study, we evaluated protective efficacy of a trivalent vaccine containing flagellins A and B (FlaA + FlaB) + pilin (PilA) in a murine burn model of infection. "FlaA + FlaB + PilA" induced greater protection in P. aeruginosa murine burn model than the single components alone, and it showed broad immune protection against P. aeruginosa strains. Immunization with "FlaA + FlaB + PilA" induced strong opsonophagocytic antibodies and resulted in reduced bacterial loads, systemic IL-12/IL-10 cytokine expression, and increased survival after challenge with three times lethal dose fifty (LD50) of P. eruginosa strains. Moreover, the protective efficacy of "FlaA + FlaB + PilA" vaccination was largely attributed to specific antibodies. Taken together, these data further confirm that the protective effects of "FlaA + FlaB + PilA" vaccine significantly enhance efficacy compared with antibodies against either mono or divalent antigen, and that the former broadens the coverage against P. eruginosa strains that express two of the three antigens.


Subject(s)
Burns/microbiology , Pseudomonas Vaccines , Pseudomonas aeruginosa/immunology , Wound Infection/microbiology , Animals , Disease Models, Animal , Fimbriae Proteins/immunology , Flagellin/immunology , Mice , Pseudomonas Infections/prevention & control , Vaccination
5.
J Bacteriol ; 201(13)2019 07 01.
Article in English | MEDLINE | ID: mdl-30988037

ABSTRACT

The major subunit of the type IV pilus (T4p) of Neisseria gonorrhoeae undergoes antigenic variation (AV) dependent on a guanine quadruplex (G4) DNA structure located upstream of the pilin gene. Since the presence of G4 DNA induces genome instability in both eukaryotic and prokaryotic chromosomes, we tested whether a double-strand break (DSB) at the site of the pilE G4 sequence could substitute for G4-directed pilin AV. The G4 motif was replaced by an I-SceI cut site, and the cut site was also introduced to locations near the origin of replication and the terminus. Expression of the I-SceI endonuclease from an irrelevant chromosomal site confirmed that the endonuclease functions to induce double-strand breaks at all three locations. No antigenic variants were detected when the G4 was replaced with the I-SceI cut site, but there was a growth defect from having a DSB in the chromosome, and suppressor mutations that were mainly deletions of the cut site and/or the entire pilE gene accumulated. Thus, the pilE G4 does not act to promote pilin AV by generating a DSB but requires either a different type of break, a nick, or more complex interactions with other factors to stimulate this programmed recombination system.IMPORTANCENeisseria gonorrhoeae, the causative agent of gonorrhea, possesses a DNA recombination system to change one of its surface-exposed antigens. This recombination system, known as antigenic variation, uses an alternate DNA structure to initiate variation. The guanine quadruplex DNA structure is known to cause nicks or breaks in DNA; however, much remains unknown about how this structure functions in cells. We show that inducing a break by different means does not allow antigenic variation, indicating that the DNA structure may have a more complicated role.


Subject(s)
Antigenic Variation , DNA Breaks, Double-Stranded , Fimbriae Proteins/immunology , Neisseria gonorrhoeae/genetics , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , DNA, Bacterial/genetics , Fimbriae Proteins/genetics , Fimbriae, Bacterial/immunology , G-Quadruplexes , Recombination, Genetic
6.
Infect Immun ; 87(8)2019 08.
Article in English | MEDLINE | ID: mdl-31085711

ABSTRACT

Nontypeable Haemophilus influenzae (NTHi) is a pathogen known for being a frequent cause of acute otitis media in children and respiratory tract infections in adults with chronic obstructive pulmonary disease. In the present study, a vaccine antigen based on the fusion of two known NTHi adhesive proteins, protein E (PE) and a pilin subunit (PilA), was developed. The quality of the combined antigen was investigated through functional, biophysical, and structural analyses. It was shown that the PE and PilA individual structures are not modified in the PE-PilA fusion and that PE-PilA assembles as a dimer in solution, reflecting PE dimerization. PE-PilA was found to bind vitronectin by enzyme-linked immunosorbent assay, as isolated PE does. Disulfide bridges were conserved and homogeneous, which was determined by peptide mapping and top-down analysis of PE, PilA, and PE-PilA molecules. Finally, the PE-PilA crystal showed a PE entity with a three-dimensional (3D) structure similar to that of the recently published isolated PE, while the structure of the PilA entity was similar to that of a 3D model elaborated from two other type 4 pilin subunits. Taken together, our observations suggest that the two tethered proteins behave independently within the chimeric molecule and display structures similar to those of the respective isolated antigens, which are important characteristics for eliciting optimal antibody-mediated immunity. PE and PilA can thus be further developed as a single fusion protein in a vaccine perspective, in the knowledge that tethering the two antigens does not perceptibly compromise the structural attributes offered by the individual antigens.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Fimbriae Proteins/immunology , Haemophilus Vaccines/immunology , Bacterial Proteins/chemistry , Crystallization , Fimbriae Proteins/chemistry , Protein Folding , Vaccines, Synthetic/immunology
7.
Infect Immun ; 87(8)2019 08.
Article in English | MEDLINE | ID: mdl-31109946

ABSTRACT

PE-PilA is a fusion protein composed of immunologically relevant parts of protein E (PE) and the majority subunit of the type IV pilus (PilA), two major antigens of nontypeable Haemophilus influenzae (NTHi). Here we report on the preclinical evaluation of PE-PilA as a vaccine antigen. The immunogenic potential of the PE and PilA within the fusion was compared with that of isolated PE and PilA antigens. When injected intramuscularly into mice, the immunogenicity of PE within the fusion was equivalent to that of isolated PE, except when it was formulated with alum. In contrast, in our murine models PilA was consistently found to be more immunogenic as a subentity of the PE-PilA fusion protein than when it was injected as an isolated antigen. Following immunization with PE-PilA, anti-PE antibodies demonstrated the same capacity to inhibit the binding of PE to vitronectin as those induced after PE immunization. Likewise, PE-PilA-induced anti-PilA antibodies inhibited the formation of NTHi biofilms and disrupted established biofilms in vitro These experiments support the immunogenic equivalence between fused PE-PilA and isolated PE and PilA. Further, the potential of PE-PilA immunization against NTHi-induced disease was evaluated. After intranasal NTHi challenge, colonization of the murine nasopharynx significantly dropped in animals formerly immunized with PE-PilA, and in chinchillas, signs of otitis media were significantly reduced in animals that had received anti-PE-PilA antibodies. Taken together, our data support the use of PE-PilA as an NTHi vaccine antigen.


Subject(s)
Bacterial Proteins/immunology , Fimbriae Proteins/immunology , Haemophilus Vaccines/immunology , Haemophilus influenzae/immunology , Animals , Bacterial Adhesion , Biofilms , Chinchilla , Female , Immunization , Mice , Mice, Inbred BALB C , Nasopharynx/microbiology , Otitis Media/prevention & control , Vaccines, Synthetic/immunology , Vitronectin/metabolism
8.
Infect Immun ; 87(12)2019 12.
Article in English | MEDLINE | ID: mdl-31570556

ABSTRACT

The Porphyromonas gingivalis strain ATCC 33277 (33277) and 381 genomes are nearly identical. However, strain 33277 displays a significantly diminished capacity to stimulate host cell Toll-like receptor 2 (TLR2)-dependent signaling and interleukin-1ß (IL-1ß) production relative to 381, suggesting that there are strain-specific differences in one or more bacterial immune-modulatory factors. Genomic sequencing identified a single nucleotide polymorphism in the 33277 fimB allele (A→T), creating a premature stop codon in the 33277 fimB open reading frame relative to the 381 fimB allele. Gene exchange experiments established that the 33277 fimB allele reduces the immune-stimulatory capacity of this strain. Transcriptome comparisons revealed that multiple genes related to carboxy-terminal domain (CTD) family proteins, including the gingipains, were upregulated in 33277 relative to 381. A gingipain substrate degradation assay demonstrated that cell surface gingipain activity is higher in 33277, and an isogenic mutant strain deficient for the gingipains exhibited an increased ability to induce TLR2 signaling and IL-1ß production. Furthermore, 33277 and 381 mutant strains lacking CTD cell surface proteins were more immune-stimulatory than the parental wild-type strains, consistent with an immune-suppressive role for the gingipains. Our data show that the combination of an intact fimB allele and limited cell surface gingipain activity in P. gingivalis 381 renders this strain more immune-stimulatory. Conversely, a defective fimB allele and high-level cell surface gingipain activity reduce the capacity of P. gingivalis 33277 to stimulate host cell innate immune responses. In summary, genomic and transcriptomic comparisons identified key virulence characteristics that confer divergent host cell innate immune responses to these highly related P. gingivalis strains.


Subject(s)
Fimbriae Proteins/genetics , Fimbriae Proteins/immunology , Gingipain Cysteine Endopeptidases/metabolism , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/immunology , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Cell Line, Tumor , HEK293 Cells , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Interleukin-1beta/metabolism , Polymorphism, Single Nucleotide/genetics , Signal Transduction/immunology , THP-1 Cells , Toll-Like Receptor 2/metabolism
9.
Infect Immun ; 87(12)2019 12.
Article in English | MEDLINE | ID: mdl-31548326

ABSTRACT

The type IV pilus (Tfp) of nontypeable Haemophilus influenzae (NTHI) mediates adherence, colonization, motility, and biofilm formation, and the major protein subunit, PilA, is a promising vaccine candidate. Thus, it is crucial to understand how Tfp expression is regulated within the microenvironments of the human nasopharynx, which NTHI colonizes asymptomatically, and the more distal regions of the respiratory tract where NTHI-induced diseases occur. Here, we examined the effects of coculture of NTHI with human airway epithelial cells and heme availability on Tfp expression at temperatures typical of the human nasopharynx (34°C) or warmer anatomical sites during infection (37°C). Tfp expression was estimated by pilA promoter activity, pilA gene expression, and relative abundances of PilA and pilin protein. The results revealed that at both temperatures, NTHI cocultured with airway epithelial cells demonstrated significantly greater expression of pilA, PilA/pilin protein, and likely, fully assembled Tfp than NTHI cultured on an abiotic surface. Because NTHI is a heme auxotroph, we hypothesized that availability of heme from host cells might be a signal for Tfp expression. Thereby, we cultured NTHI in iron-limited medium, and we observed that supplementation with heme significantly increased pilA promoter activity. Collectively, our data suggested that NTHI Tfp expression was stimulated by soluble factor(s) released by epithelial cells, which are present in all microenvironments of the respiratory tract. The expression of this target antigen under conditions that mimic the human airway strongly supports the rationale for the use of PilA as a vaccine immunogen to prevent NTHI-induced diseases of the respiratory tract.


Subject(s)
Fimbriae Proteins/biosynthesis , Fimbriae Proteins/immunology , Fimbriae, Bacterial/immunology , Haemophilus influenzae/immunology , Nasopharynx/immunology , Bacterial Adhesion/genetics , Bacterial Vaccines/immunology , Cells, Cultured , Coculture Techniques , Epithelial Cells/immunology , Epithelial Cells/metabolism , Fimbriae Proteins/genetics , Fimbriae, Bacterial/metabolism , Haemophilus Infections/immunology , Haemophilus Infections/microbiology , Heme/metabolism , Humans , Nasopharynx/microbiology , Promoter Regions, Genetic/genetics , Respiratory System/cytology
10.
Mol Cell Probes ; 45: 19-25, 2019 06.
Article in English | MEDLINE | ID: mdl-30940544

ABSTRACT

FimH is the adhesin of type I fimbriae expressed on Escherichia coli that can mediate specific adherence to host cells. High binding mutations in FimH are related to the adaptive evolution of bacteria. However, additional roles that these allelic variations may play remain elusive. To investigate novel biological functions of the mutations in FimH, we introduced four different variants of FimH by incorporating single amino acid substitutions at specific sites, namely A25P, G73R, A106, and T158P, respectively. In this study, adjuvant potential of FimH variants was evaluated by investigating their ability to trigger innate immune response to DC2.4 and adaptive immunity to improve immunological characteristics. The data revealed that purified A106 and T158P up-regulated the expression of co-stimulatory molecules critically involved in DC2.4 activation by interaction with TLR4, whereas A25P and G73R did not induce the phenotypic maturation of DC2.4. Besides, the culture of DC2.4 with A106 and T158P enhanced the release of cytokines and protein phagocytosis. When formulated with PAc, T158P elicited more robust PAc-specific IgG and IgA antibody responses compared to PBS, PAc and PAc+K12 groups and inhibited bacteria colonization. Collectively, the results confirmed that the T158P mutation located around the inter-domain interface of the protein induced a specific enhancement effect on adjuvant characteristics.


Subject(s)
Adhesins, Escherichia coli/administration & dosage , Antigens, Surface/administration & dosage , Fimbriae Proteins/administration & dosage , Point Mutation , Streptococcal Vaccines/administration & dosage , Streptococcus mutans/immunology , Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/immunology , Adjuvants, Immunologic/administration & dosage , Amino Acid Substitution , Animals , Antigens, Surface/immunology , Bacterial Proteins/administration & dosage , Bacterial Proteins/immunology , Cell Line , Cytokines/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Fimbriae Proteins/genetics , Fimbriae Proteins/immunology , Mice , Phagocytosis , Streptococcal Vaccines/genetics , Streptococcal Vaccines/immunology
11.
BMC Vet Res ; 15(1): 234, 2019 Jul 08.
Article in English | MEDLINE | ID: mdl-31286936

ABSTRACT

BACKGROUND: Enterotoxigenic Escherichia coli K88 (E. coli K88) are considered as a major cause of diarrhea and death in newly weaned piglets. Oral passive immunization with chicken egg yolk immunoglobulins (IgY) have attracted considerable attention for treatment of gastrointestinal infection due to its high specificity. In this study it was estimated the protective effect of anti-K88 fimbriae IgY against E. coli K88 adhesion to piglet intestinal mucus in vitro and to investigate the potential use of IgY for controlling E. coli-induced diarrhea in weaned piglets in vivo. RESULTS: E. coli K88 was incubated with IgY for 24 h, and the bacterial growth profiles showed that specific IgY with a concentration higher than 5 mg/mL was observed to significantly inhibit the growth of E. coli K88 compared to nonspecific yolk powder in a liquid medium. Moreover, pretreatment with 50 mg/mL of IgY was found to significantly decrease the adhesion ability of E. coli K88 to porcine jejunal and ileal mucus, further supported by the observations from our immunofluorescence microscopic analysis. In vivo, administration of IgY successfully protected piglets from diarrhea caused by E. coli K88 challenge. Additionally, IgY treatment efficiently alleviated E. coli-induced intestinal inflammation in piglets as the gene expression levels of inflammatory cytokines TNF-α, IL-22, IL-6 and IL-1ß in IgY-treated piglets remained unchanged after E. coli K88 infection. Furthermore, IgY significantly prevented E. coli K88 adhering to the jejunal and ileal mucosa of piglets with E. coli infection and significantly decreased E. coli and enterotoxin expression in colonic contents. CONCLUSION: Outcome of the study demonstrated that IgY against the fimbrial antigen K88 was able to significantly inhibit the growth of E. coli K88, block the binding of E. coli to small intestinal mucus, and protect piglets from E. coli-induced diarrhea. These results indicate that passive immunization with IgY may be useful to prevent bacterial colonization and to control enteric diseases due to E. coli infection. The study has great clinical implication to provide alternative therapy to antibiotics in E coli induced diarrhea.


Subject(s)
Bacterial Adhesion/drug effects , Diarrhea/etiology , Diarrhea/prevention & control , Enterotoxigenic Escherichia coli/drug effects , Escherichia coli Infections/complications , Immunoglobulins/pharmacology , Animals , Antigens, Bacterial/immunology , Cytokines/genetics , Diarrhea/microbiology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/pathology , Escherichia coli Infections/prevention & control , Escherichia coli Proteins/immunology , Fimbriae Proteins/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Immunization, Passive , Immunoglobulins/therapeutic use , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Protein Binding/drug effects , Swine
12.
J Infect Chemother ; 25(8): 643-645, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31053536

ABSTRACT

Serological surveillance of pertussis antibodies was performed in 118 children aged 1-12 years. The positivity of pertussis toxin (PT) antibodies was low at 4-6 years and significantly higher at 8-9 years, compared with those at 6 years. Fimbriae 2 (Fim2) antibody showed similar response to the PT antibody. Higher antibody titers against Fim3 were observed among subjects ≥5 years and highest at 8 years. Data demonstrated that the vaccine-induced antibodies decayed by 4-5 years and subclinical pertussis infection was suspected thereafter, suggesting the need for additional dose at around 4-5 years.


Subject(s)
Bordetella pertussis/immunology , Vaccines/immunology , Whooping Cough/immunology , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Child , Child, Preschool , Female , Fimbriae Proteins/immunology , Fimbriae, Bacterial/immunology , Humans , Infant , Male , Pertussis Toxin/immunology , Vaccination/methods , Virulence Factors, Bordetella/immunology
13.
Infect Immun ; 86(3)2018 03.
Article in English | MEDLINE | ID: mdl-29263112

ABSTRACT

Enterotoxigenic Escherichia coli (ETEC) strains are a leading cause of children's diarrhea and travelers' diarrhea. Vaccines inducing antibodies to broadly inhibit bacterial adherence and to neutralize toxin enterotoxicity are expected to be effective against ETEC-associated diarrhea. 6×His-tagged adhesin-toxoid fusion proteins were shown to induce neutralizing antibodies to several adhesins and LT and STa toxins (X. Ruan, D. A. Sack, W. Zhang, PLoS One 10:e0121623, 2015, https://doi.org/10.1371/journal.pone.0121623). However, antibodies derived from His-tagged CFA/I/II/IV-2xSTaA14Q-dmLT or CFA/I/II/IV-2xSTaN12S-dmLT protein were less effective in neutralizing STa enterotoxicity and were not evaluated in vivo for efficacy against ETEC diarrhea. Additionally, His-tagged proteins are considered less desirable for human vaccines. In this study, we produced a tagless adhesin-toxoid MEFA (multiepitope fusion antigen) protein, enhanced anti-STa immunogenicity by including a third copy of STa toxoid STaN12S, and examined antigen immunogenicity in a murine model. Moreover, we immunized pregnant pigs with the tagless adhesin-toxoid MEFA protein and evaluated passive antibody protection against STa+ or LT+ ETEC infection in a pig challenge model. Results showed that tagless adhesin-toxoid MEFA CFA/I/II/IV-3xSTaN12S-mnLTR192G/L211A induced broad antiadhesin and antitoxin antibody responses in the intraperitoneally immunized mice and the intramuscularly immunized pigs. Mouse and pig serum antibodies significantly inhibited adherence of seven colonization factor antigen (CFA) adhesins (CFA/I and CS1 to CS6) and effectively neutralized both toxins. More importantly, suckling piglets born to the immunized mothers acquired antibodies and were protected against STa+ ETEC and LT+ ETEC diarrhea. These results indicated that tagless CFA/I/II/IV-3xSTaN12S-mnLTR192G/L211A induced broadly protective antiadhesin and antitoxin antibodies and demonstrate that this adhesin-toxoid MEFA is a potential antigen for developing broadly protective ETEC vaccines.


Subject(s)
Adhesins, Bacterial/administration & dosage , Diarrhea/prevention & control , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Infections/prevention & control , Escherichia coli Proteins/administration & dosage , Escherichia coli Vaccines/administration & dosage , Toxoids/administration & dosage , Adhesins, Bacterial/genetics , Adhesins, Bacterial/immunology , Animals , Antibodies, Bacterial/immunology , Antibodies, Neutralizing/immunology , Antigens, Surface/administration & dosage , Antigens, Surface/genetics , Antigens, Surface/immunology , Bacterial Adhesion/drug effects , Bacterial Toxins/administration & dosage , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Diarrhea/immunology , Diarrhea/microbiology , Enterotoxigenic Escherichia coli/genetics , Enterotoxigenic Escherichia coli/physiology , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Escherichia coli Vaccines/genetics , Escherichia coli Vaccines/immunology , Female , Fimbriae Proteins/administration & dosage , Fimbriae Proteins/genetics , Fimbriae Proteins/immunology , Mice , Mice, Inbred BALB C , Swine , Toxoids/genetics , Toxoids/immunology
14.
Mol Microbiol ; 103(2): 282-298, 2017 01.
Article in English | MEDLINE | ID: mdl-27741558

ABSTRACT

Group A Streptococcus (GAS), or Streptococcus pyogenes, is a human pathogen that causes diseases ranging from skin and soft tissue infections to severe invasive diseases, such as toxic shock syndrome. Each GAS strain carries a particular pilus type encoded in the variable fibronectin-binding, collagen-binding, T antigen (FCT) genomic region. Here, we describe the functional analysis of the serotype M2 pilus encoded in the FCT-6 region. We found that, in contrast to other investigated GAS pili, the ancillary pilin 1 lacks adhesive properties. Instead, the backbone pilin is important for host cell adhesion and binds several host factors, including fibronectin and fibrinogen. Using a panel of recombinant pilus proteins, GAS gene deletion mutants and Lactococcus lactis gain-of-function mutants we show that, unlike other GAS pili, the FCT-6 pilus also contributes to immune evasion. This was demonstrated by a delay in blood clotting, increased intracellular survival of the bacteria in macrophages, higher bacterial survival rates in human whole blood and greater virulence in a Galleria mellonella infection model in the presence of fully assembled FCT-6 pili.


Subject(s)
Bacterial Adhesion/physiology , Fimbriae Proteins/physiology , Streptococcus pyogenes/physiology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Antigens, Viral, Tumor , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , Bacterial Proteins/metabolism , Biofilms , Fibronectins/metabolism , Fimbriae Proteins/genetics , Fimbriae Proteins/immunology , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Humans , Immune Evasion , Mutation , Sequence Deletion , Serogroup , Streptococcal Infections/microbiology , Streptococcus pyogenes/genetics , Streptococcus pyogenes/immunology , Streptococcus pyogenes/metabolism , Virulence
15.
Microb Pathog ; 123: 426-432, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30075242

ABSTRACT

Considering the increased antibiotic resistance of Pseudomonas aeruginosa, the evaluation of immune response against the antigens of this bacterium seems necessary. In this study, the protective efficacy and immunological properties of P. aeruginosa recombinant PilQ (r-PilQ) and type b-flagellin (FLB) proteins was evaluated in the burn mouse model of infection. The inbred BALB/c mice were immunized with r-PilQ and FLB antigens. To investigate the type of induced immune response, sera were analyzed by ELISA for total IgG, IgG1, and IgG2a isotypes. After the final immunization, the IL-4, IFN-γ, and IL-17 cytokines level were examined in the spleen of non-challenged mice. Fifty days after lethal challenge, the survival rate and bacterial burden in the skin and other internal organs of experimental mice were assessed. The in vivo administration of r-PilQ, FLB and combined antigen resulted in a significant increase in the survival of mice (66%, 75%, and 83%, respectively) infected by the PAO1 strain of P. aeruginosa in the burn model of infection. Immunization of mice with r-PilQ and FLB mixture induced high titers of IL-4 and IL-17 cytokines compared to control groups (P < 0.05). The high titer of antisera raised against combined antigen was able to inhibit the systemic spread of the PAO1 strain from the site of infection to the internal organs. We concluded that the parallel role of IL-4 and IL-17 is necessary for elimination of the bacteria and promotion of survival in the immunized burn mice.


Subject(s)
Bacterial Vaccines/immunology , Burns/immunology , Fimbriae Proteins/immunology , Flagellin/immunology , Pseudomonas Infections/immunology , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/immunology , Wound Infection/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Burns/microbiology , Cytokines/metabolism , Disease Models, Animal , Female , Fimbriae Proteins/administration & dosage , Fimbriae Proteins/genetics , Flagellin/administration & dosage , Flagellin/genetics , Immunity, Humoral , Immunization , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunologic Factors/metabolism , Interferon-gamma/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred BALB C , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Recombinant Proteins , Spleen/immunology , Survival Rate , Wound Infection/microbiology , Wound Infection/prevention & control
16.
Microb Pathog ; 114: 99-106, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29138084

ABSTRACT

Proteus mirabilis is common cause of urinary tract infections (UTIs) especially in complicated UTIs which are resistant to antibiotic therapy, Consequently, an ideal vaccine is inevitably required. The N-terminal domain of MrpH (Truncated form of MrpH) lies between the most critical antigens of P. mirabilis to consider as vaccine candidate. FliC of Salmonella typhimurium induces several pathways of immunity system, which leads to produce antibody and cytokines. In this study, adjuvant properties of FliC and efficacy of truncated MrpH as important antigen, in tMrpH.FliC were determined in in vitro and in vivo circumstances. Three proteins including: FliC, MrpH and tMrpH.FliC were injected to mice and subsequently sera and supernatant of cell culture were collected to evaluate different immune responses. According to our findings, tMrpH.FliC could stimulate both humoral and cellular immune responses, so that serum IgG, urine IgA, IL.4, IFN-γ and IL.17 were increased significantly in comparison to MrpH and FliC alone, this augmentation was considerable. Results showed significant decrease of bacterial load in all of the challenged groups compared to the control group, although this protective effect was the highest in mice vaccinated with tMrpH.FliC. Our results showed truncated MrpH, without an unwanted domain is an ideal vaccine target and FliC, as adjuvant, increases its immunogenic property. Thus, fusion protein tMrpH.FliC can be considered as promising vaccine against P. mirabilis.


Subject(s)
Adhesins, Bacterial/immunology , Adjuvants, Immunologic , Fimbriae Proteins/immunology , Flagellin/immunology , Immunogenicity, Vaccine/immunology , Proteus Infections/immunology , Proteus mirabilis/pathogenicity , Urinary Tract Infections/prevention & control , Adhesins, Bacterial/genetics , Animals , Antibodies, Bacterial/blood , Antibody Formation , Cloning, Molecular , Cytokines/metabolism , DNA, Bacterial , Female , Fimbriae Proteins/genetics , Flagellin/genetics , Gene Fusion , Immunity, Cellular , Immunity, Humoral , Immunoglobulin A/urine , Immunoglobulin G/blood , Interferon-gamma/metabolism , Interferon-gamma/urine , Interleukin-17/metabolism , Interleukin-4/metabolism , Kidney/immunology , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Protein Interaction Domains and Motifs , Proteus Infections/microbiology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Salmonella typhimurium/metabolism , Urinary Bladder/immunology , Urinary Tract Infections/microbiology
17.
Vet Res ; 49(1): 23, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29482635

ABSTRACT

The binding of F4+ enterotoxigenic Escherichia coli (ETEC) and the specific receptor on porcine intestinal epithelial cells is the initial step in F4+ ETEC infection. Porcine aminopeptidase N (APN) is a newly discovered receptor for F4 fimbriae that binds directly to FaeG adhesin, which is the major subunit of the F4 fimbriae variants F4ab, F4ac, and F4ad. We used overlapping peptide assays to map the APN-FaeG binding sites, which has facilitated in the identifying the APN-binding amino acids that are located in the same region of FaeG variants, thereby limiting the major binding regions of APN to 13 peptides. To determine the core sequence motif, a panel of FaeG peptides with point mutations and FaeG mutants were constructed. Pull-down and binding reactivity assays using piglet intestines determined that the amino acids G159 of F4ab, N209 and L212 of F4ac, and A200 of F4ad were the critical residues for APN binding of FaeG. We further show using ELISA and confocal microscopy assay that amino acids 553-568, and 652-670 of the APN comprise the linear epitope for FaeG binding in all three F4 fimbriae variants.


Subject(s)
Adhesins, Escherichia coli/immunology , Antigens, Bacterial/immunology , CD13 Antigens/metabolism , Enterotoxigenic Escherichia coli/physiology , Epitopes/immunology , Escherichia coli Proteins/immunology , Fimbriae Proteins/immunology , Fimbriae, Bacterial/immunology , Animals , Binding Sites , Escherichia coli Infections/immunology , Intestinal Mucosa/immunology , Swine
18.
J Infect Dis ; 216(1): 7-13, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28541500

ABSTRACT

Background: Tip-localized adhesive proteins of bacterial fimbriae from diverse pathogens confer protection in animal models, but efficacy in humans has not been reported. Enterotoxigenic Escherichia coli (ETEC) commonly elaborate colonization factors comprising a minor tip adhesin and major stalk-forming subunit. We assessed the efficacy of antiadhesin bovine colostral IgG (bIgG) antibodies against ETEC challenge in volunteers. Methods: Adults were randomly assigned (1:1:1) to take oral hyperimmune bIgG raised against CFA/I minor pilin subunit (CfaE) tip adhesin or colonization factor I (CFA/I) fimbraie (positive control) or placebo. Two days before challenge, volunteers began a thrice-daily, 7-day course of investigational product administered in sodium bicarbonate 15 minutes after each meal. On day 3, subjects drank 1 × 109 colony-forming units of colonization factor I (CFA/I)-ETEC strain H10407 with buffer. The primary efficacy endpoint was diarrhea within 120 hours of challenge. Results: After enrollment and randomization, 31 volunteers received product, underwent ETEC challenge, and were included in the per protocol efficacy analysis. Nine of 11 placebos developed diarrhea, 7 experiencing moderate to severe disease. Protective efficacy of 63% (P = .03) and 88% (P = .002) was observed in the antiadhesin bIgG and positive control groups, respectively. Conclusions: Oral administration of anti-CFA/I minor pilin subunit (CfaE) antibodies conferred significant protection against ETEC, providing the first clinical evidence that fimbrial tip adhesins function as protective antigens.


Subject(s)
Antibodies, Bacterial/therapeutic use , Colostrum/immunology , Diarrhea/drug therapy , Enterotoxigenic Escherichia coli , Escherichia coli Infections/drug therapy , Immunoglobulin G/therapeutic use , Adhesins, Bacterial/immunology , Administration, Oral , Adult , Animals , Antigens, Bacterial/immunology , Cattle , Colony Count, Microbial , Diarrhea/microbiology , Double-Blind Method , Female , Fimbriae Proteins/immunology , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Humans , Male , Reproducibility of Results , Young Adult
19.
Immunology ; 151(1): 98-109, 2017 05.
Article in English | MEDLINE | ID: mdl-28066900

ABSTRACT

Burkholderia pseudomallei is the causative agent of melioidosis, an infectious disease with high incidence and mortality in South East Asia and northern Australia. To date there is no protective vaccine and antibiotic treatment is prolonged and not always effective. Most people living in endemic areas have been exposed to the bacteria and have developed some immunity, which may have helped to prevent disease. Here, we used a humanized mouse model (hu-PBL-SCID), reconstituted with human peripheral blood mononuclear cells from seropositive donors, to illustrate the potential of three known antigens (FliC, OmpA and N-PilO2) for boosting both T-cell and B-cell immune responses. All three antigens boosted the production of specific antibodies in vivo, and increased the number of antibody and interferon-γ-secreting cells, and induced antibody affinity maturation. Moreover, antigen-specific antibodies isolated from either seropositive individuals or boosted mice, were found to enhance phagocytosis and oxidative burst activities from human polymorphonuclear cells. Our study demonstrates that FliC, OmpA and N-PilO2 can stimulate human memory T and B cells and highlight the potential of the hu-PBL-SCID system for screening and evaluation of novel protein antigens for inclusion in future vaccine trials against melioidosis.


Subject(s)
B-Lymphocytes/immunology , Bacterial Vaccines/immunology , Burkholderia pseudomallei/immunology , Melioidosis/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Antibodies, Bacterial/blood , B-Lymphocytes/microbiology , Bacterial Outer Membrane Proteins/immunology , Cells, Cultured , Endemic Diseases , Fimbriae Proteins/immunology , Flagellin/immunology , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Melioidosis/epidemiology , Mice , Mice, SCID , T-Lymphocytes/microbiology , Thailand
20.
Microb Pathog ; 105: 177-184, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28215587

ABSTRACT

Helicobacter pylori infection can cause peptic ulceration and is associated with gastric adenocarcinoma. This study aimed to construct and characterize a non-virulent Vibrio cholerae O1 strain, which grows more rapidly than H. pylori, as vector for H. pylori antigens for possible use as a vaccine strain against H. pylori. This was done by recombinant expression of the H. pylori adhesion antigen HpaA alone or, as a proof of principle, together with different colonization factor (CF) antigens of enterotoxigenic Escherichia coli (ETEC) which may enhance immune responses against HpaA. A recombinant V. cholerae strain co-expressing HpaA and a fimbrial CF antigens CFA/I or CS5, but not the non-fimbrial CF protein CS6, was shown to express larger amounts of HpaA on the surface when compared with the same V. cholerae strain expressing HpaA alone. Mutations in the CFA/I operon showed that the chaperon, possibly together with the usher, was involved in enhancing the surface expression of HpaA. Oral immunization of mice with formaldehyde-inactivated recombinant V. cholerae expressing HpaA alone or together with CFA/I induced significantly higher serum antibody responses against HpaA than mice similarly immunized with inactivated HpaA-expressing H. pylori bacteria. Our results demonstrate that a non-virulent V. cholerae strain can be engineered to allow strong surface expression of HpaA, and that the expression can be further increased by co-expressing it with ETEC fimbrial antigens. Such recombinant V. cholerae strains expressing HpaA, and possibly also other H. pylori antigens, may have the potential as oral inactivated vaccine candidates against H. pylori.


Subject(s)
Adhesins, Bacterial/immunology , Escherichia coli Proteins/genetics , Fimbriae Proteins/immunology , Helicobacter pylori/immunology , Vibrio cholerae/immunology , Adhesins, Bacterial/biosynthesis , Adhesins, Bacterial/genetics , Animals , Antibodies, Bacterial/immunology , Antibody Formation , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , Bacterial Outer Membrane Proteins , Bacterial Vaccines/immunology , DNA, Bacterial , Enterotoxigenic Escherichia coli/genetics , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/immunology , Female , Fimbriae Proteins/biosynthesis , Fimbriae Proteins/genetics , Fimbriae, Bacterial/genetics , Gene Expression Regulation, Bacterial , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter Infections/prevention & control , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Immunity, Heterologous/genetics , Immunity, Heterologous/immunology , Mice , Mice, Inbred C57BL , Protein Engineering , Recombinant Proteins/biosynthesis , Vaccines, Synthetic/immunology , Vibrio cholerae/genetics , Vibrio cholerae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL