Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 238
Filter
Add more filters

Publication year range
1.
Int Immunol ; 33(9): 491-504, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34297096

ABSTRACT

As a key virulence factor for persistent colonization, urease B subunit (UreB) is considered to be an ideal vaccine antigen against Helicobacter pylori infection. However, the role and molecular mechanisms of UreB involved in immune microenvironment dysregulation still remain largely unknown. In the present study, we evaluated the effects of UreB on macrophage activation and found that UreB induced PD-L1 accumulation on bone marrow-derived macrophages (BMDMs). Co-culture assays further revealed that UreB-induced PD-L1 expression on BMDMs significantly decreased the proliferation and secretion of cytolytic molecules (granzyme B and perforin) of splenic CD8+ T cells isolated from inactivated H. pylori-immunized mice. More importantly, using liquid chromatography-tandem mass spectrometry (LC-MS/MS) and co-immunoprecipitation techniques, it has been confirmed that myosin heavy chain 9 (Myh9) is a direct membrane receptor for UreB and is required for PD-L1 up-regulation on BMDMs. Molecular studies further demonstrated that the interaction between UreB and Myh9 decreased GCN2 autophosphorylation and enhanced the intracellular pool of amino acids, leading to the up-regulation of S6K phosphorylation, a commonly used marker for monitoring activation of mTORC1 signaling activity. Furthermore, blocking mTORC1 activation with its inhibitor Temsirolimus reversed the UreB-induced PD-L1 up-regulation and the subsequent inhibitory effects of BMDMs on activation of cytotoxic CD8+ T-cell responses. Overall, our data unveil a novel immunosuppressive mechanism of UreB during H. pylori infection, which may provide valuable clues for the optimization of H. pylori vaccine.


Subject(s)
B7-H1 Antigen/immunology , Bacterial Proteins/immunology , CD8-Positive T-Lymphocytes/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Myosin Heavy Chains/immunology , Urease/immunology , Animals , Cell Line , Cell Line, Tumor , Humans , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , RAW 264.7 Cells , THP-1 Cells
2.
Helicobacter ; 26(1): e12758, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33259676

ABSTRACT

BACKGROUND: Helicobacter pylori is a gram-negative bacterium involved in many gastric pathologies such as ulcers and cancers. Although the treatment for this infection has existed for several years, the development of a vaccine is nevertheless necessary to reduce the severe forms of the disease. For more than three decades, many advances have been made particularly in the understanding of virulence factors as well as the pathogenesis of gastric diseases caused by H. pylori. Among these key virulence factors, specific antigens have been identified: Urease, Vacuolating cytotoxin A (VacA), Cytotoxin-associated gene A (CagA), Blood group antigen-binding adhesin (BabA), H. pylori adhesin A (HpaA), and others. OBJECTIVES: This review will focus on H. pylori adhesins, in particular, on HpaA and on the current knowledge of H. pylori vaccines. METHODS: All of the information included in this review was retrieved from published studies on H. pylori adhesins in H. pylori infections. RESULTS: These proteins, used in their native or recombinant forms, induce protection against H. pylori in experimental animal models. CONCLUSION: H. pylori adhesins are known to be promising candidate vaccines against H. pylori. Future research should be carried out on adhesins, in particular, on HpaA.


Subject(s)
Adhesins, Bacterial/immunology , Bacterial Vaccines/immunology , Helicobacter Infections , Helicobacter pylori , Animals , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Urease/immunology , Virulence Factors/immunology
3.
Appl Microbiol Biotechnol ; 103(8): 3407-3420, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30810777

ABSTRACT

Helicobacter pylori bacteria are involved in gastroduodenal disorders, including gastric adenocarcinoma. Since the current therapies encounter with some significant shortcomings, much attention has been paid to the development of new alternative diagnostic and treatment modalities such as immunomedicines to target H. pylori. Having used phage display technology, we isolated fully humane small antibody (Ab) fragment (VL) against the Flap region of urease enzyme of H. pylori to suppress its enzymatic activity. Solution biopanning (SPB) and screening process against a customized biotinylated peptide corresponding to the enzyme Flap region resulted in the selection of VL single domain Abs confirmed by the enzyme-linked immunosorbent assay (ELISA), sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), and Western blotting. The selected Ab fragments showed a high affinity with a KD value of 97.8 × 10-9 and specificity to the enzyme with high inhibitory impact. For the first time, a VL single domain Ab was isolated by SPB process against a critical segment of H. pylori urease using a diverse semi-synthetic library. Based on our findings, the selected VL Ab fragments can be used for the diagnosis, imaging, targeting, and/or immunotherapy of H. pylori. Further, Flap region shows great potential for vaccine therapy.


Subject(s)
Antibodies, Bacterial/immunology , Bacterial Proteins/immunology , Helicobacter pylori/enzymology , Single-Domain Antibodies/immunology , Urease/immunology , Antibody Affinity , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Cell Surface Display Techniques , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/immunology , Humans , Peptide Library , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics , Urease/antagonists & inhibitors , Urease/chemistry
4.
Helicobacter ; 22(5)2017 Oct.
Article in English | MEDLINE | ID: mdl-28805287

ABSTRACT

BACKGROUND: Gram-positive enhancer matrix particles (GEM) produced by Lactococcus lactis can enhance vaccine-induced immune response. However, the mechanism under which this adjuvant mounts the efficacy of orally administered vaccines remains unexplored. MATERIALS AND METHODS: We used a prophylactic mice model to investigate the mechanism of GEM-adjuvanted vaccination. Helicobacter pylori urease-specific antibody response was monitored and detected in murine serum by ELISA. Urease-specific splenic cytokine profile was examined. Gastric inflammatory responses were measured on day 43 or 71 by quantitative real-time PCR, flow cytometry and histology. RESULTS: We found that GEM enhanced the efficiency of oral H. pylori vaccine by promoting innate immunity. The vaccine CUE-GEM composed of GEM particles and recombinant antigen CTB-UE provided protection of immunized mice against H. pylori insult. The protective response was associated with induction of postimmunization gastritis and local Th1/Th17 cell-medicated immune response. We showed that innate inflammatory responses including neutrophil chemokines CXCL1-2, neutrophils, and antimicrobial proteins S100A8 and MUC1 were significantly elevated. Within all infected mice, S100A8 and MUC1 levels were negatively correlated with H. pylori burden. Strikingly, mice receiving GEM also show reduction of colonization, possibly through natural host response pathways to recruit CD4+ T cells and promote S100A8 expression. CONCLUSIONS: These findings suggest that GEM-based vaccine may impact Th1/Th17 immunity to orchestrate innate immune response against H. pylori infection.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Vaccines/immunology , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Immunity, Innate , Lactococcus lactis/immunology , Animals , Antibodies, Bacterial/blood , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/isolation & purification , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gastric Mucosa/pathology , Helicobacter Infections/pathology , Leukocytes, Mononuclear/immunology , Male , Mice, Inbred BALB C , Urease/immunology
5.
Helicobacter ; 22(6)2017 Dec.
Article in English | MEDLINE | ID: mdl-28851031

ABSTRACT

BACKGROUND: Therapeutic vaccination is a desirable alternative for controlling Helicobacter pylori (H. pylori) infection. Attachment to the gastric mucosa is the first step in establishing bacterial colonization, and adhesins, which are on the surface of H. pylori, play a pivotal role in binding to human gastric mucosa. MATERIALS AND METHODS: In the present study, we constructed a multivalent epitope-based vaccine named CFAdE with seven carefully selected antigenic fragments from four H. pylori adhesins (urease, Lpp20, HpaA and CagL). The specificity, immunogenicity and ability to produce neutralizing antibodies of CFAdE were evaluated in BALB/c mice. After that, its therapeutic efficacy and protective immune mechanisms were explored in H. pylori-infected Mongolian gerbils. RESULTS: The results indicated that CFAdE could induce comparatively high levels of specific antibodies against urease, Lpp20, HpaA and CagL. Additionally, oral therapeutic immunization with CFAdE plus polysaccharide adjuvant (PA) significantly decreased H. pylori colonization compared with oral immunization with urease plus PA, and the protection was correlated with IgG and sIgA antibody and antigen-specific CD4+ T cells. CONCLUSIONS: This study indicated that the multivalent epitope-based vaccine, which targeted multiple adhesins in adherence of H. pylori to the gastric mucosa, is more effective than the univalent vaccine targeting urease only. This multivalent epitope-based vaccine may be a promising therapeutic candidate vaccine against H. pylori infection.


Subject(s)
Adhesins, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Helicobacter Infections/therapy , Helicobacter pylori/immunology , Lipoproteins/immunology , Urease/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Bacterial Vaccines/administration & dosage , Disease Models, Animal , Epitopes/immunology , Gerbillinae , Immunoglobulin A/blood , Immunoglobulin G/blood , Male , Mice, Inbred BALB C , Treatment Outcome
6.
J Korean Med Sci ; 32(7): 1139-1146, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28581271

ABSTRACT

To identify the Helicobacter pylori antigens operating during early infection in sera from infected infants using proteomics and immunoblot analysis. Two-dimensional (2D) large and small gel electrophoresis was performed using H. pylori strain 51. We performed 2D immunoglobulin G (IgG), immunoglobulin A (IgA), and immunoglobulin M (IgM) antibody immunoblotting using small gels on sera collected at the Gyeongsang National University Hospital from 4-11-month-old infants confirmed with H. pylori infection by pre-embedding immunoelectron microscopy. Immunoblot spots appearing to represent early infection markers in infant sera were compared to those of the large 2D gel for H. pylori strain 51. Corresponding spots were analyzed by matrix-assisted laser desorption/ionization time of flight-mass spectrometry (MALDI-TOF-MS). The peptide fingerprints obtained were searched in the National Center for Biotechnology Information (NCBI) database. Eight infant patients were confirmed with H. pylori infection based on urease tests, histopathologic examinations, and pre-embedding immunoelectron microscopy. One infant showed a 2D IgM immunoblot pattern that seemed to represent early infection. Immunoblot spots were compared with those from whole-cell extracts of H. pylori strain 51 and 18 spots were excised, digested in gel, and analyzed by MALDI-TOF-MS. Of the 10 peptide fingerprints obtained, the H. pylori proteins flagellin A (FlaA), urease ß subunit (UreB), pyruvate ferredoxin oxidoreductase (POR), and translation elongation factor Ts (EF-Ts) were identified and appeared to be active during the early infection periods. These results might aid identification of serological markers for the serodiagnosis of early H. pylori infection in infants.


Subject(s)
Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Bacterial Proteins/analysis , Helicobacter Infections/diagnosis , Helicobacter pylori/immunology , Hydro-Lyases/analysis , Oxidoreductases/analysis , Peptide Elongation Factors/analysis , Pyruvate Synthase/analysis , Urease/analysis , Bacterial Proteins/immunology , Biomarkers/analysis , Female , Humans , Hydro-Lyases/immunology , Immunoblotting , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Infant , Male , Oxidoreductases/immunology , Peptide Elongation Factors/immunology , Peptide Mapping , Pyruvate Synthase/immunology , Serologic Tests , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Urease/immunology
7.
Appl Microbiol Biotechnol ; 100(14): 6219-6229, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26846746

ABSTRACT

The gastric bacterial pathogen Helicobacter pylori persistently colonizes the gastric mucosa of humans and plays a critical role in the development of gastritis, peptic ulceration and gastric adenocarcinoma. Consequently, the eradication of H. pylori might contribute to the prevention of H. pylori-associated gastric diseases. In this study, a multi-epitope vaccine CTB-UE (CUE) was displayed on the surface of non-genetically modified Lactococcus lactis particles (GEM) to enhance immunogenicity. This particulate vaccine CUE-GEM induced serum and mucosal specific antibody responses against native H. pylori urease and provided potent protection to eliminate H. pylori colonization and relieve gastritis in an H. pylori-infected BALB/c mouse model. The immuno-protective mechanisms are highly associated with CD4(+) Th cell-mediated and humoral immunity, especially local immunity. There might be two main aspects of this association. One aspect is related to the suppression of urease activity by promotion of the production of specific mucosal neutralizing antibody. The other aspect is correlated with alleviating gastritis by regulating the gastric pro-inflammatory cytokine profile, especially IFN-γ and IL-17. These results demonstrated that conjugating antigen vaccines with GEM particles could lead to promising oral therapeutic vaccine formulations against H. pylori infection.


Subject(s)
Bacterial Vaccines/immunology , Helicobacter Infections/prevention & control , Immunization , Lactococcus lactis/immunology , Administration, Oral , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes/immunology , Gastritis/immunology , Gastritis/microbiology , Gastritis/prevention & control , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Helicobacter Infections/immunology , Helicobacter pylori/enzymology , Helicobacter pylori/immunology , Immunity, Cellular , Immunity, Humoral , Interferon-gamma/blood , Interleukin-17/blood , Interleukin-4/blood , Interleukin-6/blood , Lactococcus lactis/metabolism , Mice , Mice, Inbred BALB C , Urease/immunology , Urease/metabolism
8.
Biochem Biophys Res Commun ; 462(3): 269-74, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-25957472

ABSTRACT

Urease is considered as an excellent vaccine candidate antigen against Helicobacter pylori (H. pylori) infection. Our previous study reported a novel multi-epitope vaccine CTB-UE which was composed of the mucosal adjuvant cholera toxin B subunit (CTB) and five cell epitopes from urease subunits. Murine experiments indicated that it could induce cellular and humoral immune responses intensively and attenuate H. pylori infection effectively in mice model. However, the body expression and lack of suitable adjuvant of this epitope vaccine restricted its application. In this study, new recombinant Escherichia coli strains was established to increase the solubility by fusing thioredoxin (Trx) and the combination adjuvants which composed of the chitosan and CpG were adopted to enhance the immunogenicity of CTB-UE for oral immunization. The experimental results indicated that the levels of IgG2a, IgG1 and IgA in the serum and the levels of sIgA in stomach, intestine and feces were significantly higher in the vaccinated group compared with the model control group. Additionally, chitosan-CpG combination adjuvants changed the ratio of IgG2a/IgG1 and conferred Th1/Th17-mediated protective immune responses. These results demonstrate that the oral vaccine with chitosan-CpG as combination adjuvants may be a promising vaccine candidate against H. pylori infection.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Chitosan/administration & dosage , Chitosan/immunology , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , CpG Islands/immunology , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Urease/administration & dosage , Urease/immunology , Administration, Oral , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/blood , Antibody Specificity , Bacterial Vaccines/genetics , Cytokines/biosynthesis , Epitopes/administration & dosage , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter pylori/enzymology , Helicobacter pylori/genetics , Immunity, Mucosal , Immunoglobulin A, Secretory/biosynthesis , Male , Mice , Mice, Inbred BALB C , Urease/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
9.
BMC Vet Res ; 11: 94, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25889568

ABSTRACT

BACKGROUND: Ureolytic activity of rumen bacteria leads to rapid urea conversion to ammonia in the rumen of dairy cows, resulting possible toxicity, excessive ammonia excretion to the environment, and poor nitrogen utilization. The present study investigated immunization of dairy cows against urease in the rumen as an approach to mitigate bacterial ureolytic activity therein. RESULTS: Most alpha subunit of rumen urease (UreC) proteins shared very similar amino acid sequences, which were also highly similar to that of H. pylori. Anti-urease titers in the serum and the saliva of the immunized cows were evaluated following repeated immunization with the UreC of H. pylori as the vaccine. After the fourth booster, the vaccinated cows had a significantly reduced urease activity (by 17%) in the rumen than the control cows that were mock immunized cows. The anti-urease antibody significantly reduced ureolysis and corresponding ammonia formation in rumen fluid in vitro. Western blotting revealed that the H. pylori UreC had high immunological homology with the UreC from rumen bacteria. CONCLUSIONS: Vaccine developed based on UreC of H. pylori can be a useful approach to decrease bacterial ureolysis in the rumen.


Subject(s)
Bacteria/enzymology , Bacterial Vaccines/immunology , Cattle/microbiology , Rumen/enzymology , Urea/metabolism , Urease/immunology , Animals , Bacteria/classification , Gene Expression Regulation, Bacterial/immunology , Gene Expression Regulation, Enzymologic/immunology , Immunization , Urease/metabolism
10.
Hepatogastroenterology ; 62(138): 503-6, 2015.
Article in English | MEDLINE | ID: mdl-25916090

ABSTRACT

BACKGROUND/AIMS: We aimed to observe the changes in the anti-Helicobacter pylori (Hp) serum antibodies to Hp virulence factors after eradication therapy and evaluate the potential application value of protein microarray in detecting Hp antibodies after eradication therapy. METHODOLOGY: A total of 107 Hp-positive patients with peptic ulcers (55) and chronic gastritis (52) were recruited. Serum antibodies to Hp urease (Ure), cytotoxin-associated protein (CagA), vacuolating cytotoxin (VacA), heat shock protein 60 (Hsp60), and anti-RdxA nitroreductase were measured. Four weeks after treatment, a 13C-urea breath test (13C- UBT) was applied to assess the Hp eradication state and to analyze correlations between the Hp eradication rate and the five antibodies. Six months after the therapy, protein microarray analysis was used to study the changes in these five serum antibodies. RESULTS: The overall Hp eradication rate was 86.0%There was no significant difference in the rate among the groups that tested positive and negative for the remaining four virulence factors. CONCLUSION: The disease type and serum anti-CagA antibody levels affect the therapeutic outcome of Hp eradication therapy. Protein microarray detection of Hp-related antibodies did not have significant application value for the long-term follow-up of Hp infection after eradication therapy.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Antibodies, Bacterial/blood , Helicobacter Infections/drug therapy , Helicobacter pylori/drug effects , Histamine H2 Antagonists/therapeutic use , Protein Array Analysis , Proton Pump Inhibitors/therapeutic use , Virulence Factors/immunology , Adult , Aged , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Breath Tests , Chronic Disease , Duodenal Ulcer/drug therapy , Duodenal Ulcer/microbiology , Female , Follow-Up Studies , Gastritis/drug therapy , Gastritis/microbiology , Helicobacter Infections/diagnosis , Helicobacter Infections/microbiology , Helicobacter pylori/immunology , Helicobacter pylori/pathogenicity , Humans , Male , Middle Aged , Remission Induction , Stomach Ulcer/drug therapy , Stomach Ulcer/microbiology , Time Factors , Treatment Outcome , Urease/immunology , Virulence
11.
Mol Cell Probes ; 28(1): 25-30, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24036137

ABSTRACT

Helicobacter pylori (H. pylori) infection remains a significant global public health problem. Vaccine, especially edible vaccine, is considered to be effective in the management of H. pylori infections. By using recombinant technology, Lactococcus lactis (L. lactis) could serve as an antigen-delivering vehicle for the development of edible vaccine. The aim of this study was to produce edible UreB (urease B) vaccine derived from L. lactis against H. pylori. The UreB subunit is the most effective and common immunogen of all strains of H. pylori. The UreB was produced as a chimeric protein fused with IL-2 (human interleukin 2) as the mucosal adjuvant. Mucosal immunization of mice with recombinant L. lactis NZ9000 containing the UreB-IL-2 protein elicited more anti-UreB antibody that specifically bounded to the purified bacterial UreB protein and more cytokines such as IFN-γ, IL-4, and IL-17, and had a lower H. pylori burden and urease activity than control mice. These results suggest that the recombinant L. lactis expressing UreB-IL-2 can be potentially used as an edible vaccine for controlling H. pylori infection.


Subject(s)
Bacterial Vaccines/administration & dosage , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Interleukin-2/immunology , Urease/immunology , Administration, Oral , Animals , Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Cytokines/immunology , Gene Expression , Genetic Vectors , Helicobacter Infections/immunology , Humans , Interleukin-2/metabolism , Lactococcus lactis/genetics , Lactococcus lactis/immunology , Male , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/immunology , Urease/genetics , Urease/metabolism , Vaccines, Edible/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
12.
Appl Microbiol Biotechnol ; 98(8): 3495-507, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24370888

ABSTRACT

Epitope vaccine is a promising option for prophylactic and therapeutic vaccination against Helicobacter pylori infection. Urease is an essential virulence factor and colonization factor for H. pylori. In this study, we constructed a multi-epitope vaccine named CTB-UE with mucosal adjuvant cholera toxin B subunit (CTB) and tandem copies of Th and B cell epitopes from H. pylori urease A and B subunits. The immunogenicity, specificity, ability to induce neutralizing antibodies against H. pylori urease, and prophylactic and therapeutic efficacy of the CTB-UE vaccine were evaluated in BALB/c mice model after purification. The experimental results indicated that CTB-UE could induce comparatively high levels of specific antibodies against native H. pylori urease, UreA, UreB, or the selected B cell epitopes UreA183₋203 and UreB327₋334 involved with the active site of urease and showed an effectively inhibitory effect on the enzymatic activity of urease. Besides, oral prophylactic or therapeutic immunization with CTB-UE significantly decreased H. pylori colonization compared with oral immunization with rUreB or PBS, and the protection was correlated with antigen-specific CD4⁺ T cells and IgG, IgA, and mucosal sIgA antibody responses. This CTB-UE vaccine may be a promising vaccine candidate for the control of H. pylori infection.


Subject(s)
Bacterial Vaccines/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Helicobacter pylori/immunology , Urease/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Oral , Animals , Antibodies, Bacterial/blood , Bacterial Load , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , CD4-Positive T-Lymphocytes/immunology , Cholera Toxin/administration & dosage , Disease Models, Animal , Epitopes, B-Lymphocyte/genetics , Epitopes, T-Lymphocyte/genetics , Helicobacter pylori/genetics , Immunity, Mucosal , Immunoglobulin A/analysis , Immunoglobulin A/blood , Immunoglobulin A, Secretory , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Stomach/microbiology , Urease/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
13.
Virulence ; 15(1): 2367783, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38937901

ABSTRACT

Helicobacter pylori causes globally prevalent infections that are highly related to chronic gastritis and even development of gastric carcinomas. With the increase of antibiotic resistance, scientists have begun to search for better vaccine design strategies to eradicate H. pylori colonization. However, while current strategies prefer to formulate vaccines with a single H. pylori antigen, their potential has not yet been fully realized. Outer membrane vesicles (OMVs) are a potential platform since they could deliver multiple antigens. In this study, we engineered three crucial H. pylori antigen proteins (UreB, CagA, and VacA) onto the surface of OMVs derived from Salmonella enterica serovar Typhimurium (S. Typhimurium) mutant strains using the hemoglobin protease (Hbp) autotransporter system. In various knockout strategies, we found that OMVs isolated from the ΔrfbP ΔfliC ΔfljB ΔompA mutants could cause distinct increases in immunoglobulin G (IgG) and A (IgA) levels and effectively trigger T helper 1- and 17-biased cellular immune responses, which perform a vital role in protecting against H. pylori. Next, OMVs derived from ΔrfbP ΔfliC ΔfljB ΔompA mutants were used as a vector to deliver different combinations of H. pylori antigens. The antibody and cytokine levels and challenge experiments in mice model indicated that co-delivering UreB and CagA could protect against H. pylori and antigen-specific T cell responses. In summary, OMVs derived from the S. Typhimurium ΔrfbP ΔfliC ΔfljB ΔompA mutant strain as the vector while importing H. pylori UreB and CagA as antigenic proteins using the Hbp autotransporter system would greatly benefit controlling H. pylori infection.


Outer membrane vesicles (OMVs), as a novel antigen delivery platform, has been used in vaccine design for various pathogens and even tumors. Salmonella enterica serovar Typhimurium (S. Typhimurium), as a bacterium that is easy to engineer and has both adjuvant efficacy and immune stimulation capacity, has become the preferred bacterial vector for purifying OMVs after Escherichia coli. This study focuses on the design of Helicobacter pylori ;(H. pylori) vaccines, utilizing genetically modified Salmonella OMVs to present several major antigens of H. pylori, including UreB, VacA and CagA. The optimal Salmonella OMV delivery vector and antigen combinations are screened and identified, providing new ideas for the development of H. pylori vaccines and an integrated antigen delivery platform for other difficult to develop vaccines for bacteria, viruses, and even tumors.


Subject(s)
Antigens, Bacterial , Bacterial Proteins , Helicobacter Infections , Helicobacter pylori , Salmonella typhimurium , Animals , Helicobacter Infections/prevention & control , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Helicobacter pylori/immunology , Helicobacter pylori/genetics , Mice , Salmonella typhimurium/immunology , Salmonella typhimurium/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Female , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Immunoglobulin G , Genetic Engineering , Urease/immunology , Urease/genetics , Disease Models, Animal
14.
Sci Rep ; 14(1): 17910, 2024 08 02.
Article in English | MEDLINE | ID: mdl-39095538

ABSTRACT

Helicobacter pylori (H. pylori) is responsible for various chronic or acute diseases, such as stomach ulcers, dyspepsia, peptic ulcers, gastroesophageal reflux, gastritis, lymphoma, and stomach cancers. Although specific drugs are available to treat the bacterium's harmful effects, there is an urgent need to develop a preventive or therapeutic vaccine. Therefore, the current study aims to create a multi-epitope vaccine against H. pylori using lipid nanoparticles. Five epitopes from five target proteins of H. pylori, namely, Urease, CagA, HopE, SabA, and BabA, were used. Immunogenicity, MHC (Major Histocompatibility Complex) bonding, allergenicity, toxicity, physicochemical analysis, and global population coverage of the entire epitopes and final construct were carefully examined. The study involved using various bioinformatic web tools to accomplish the following tasks: modeling the three-dimensional structure of a set of epitopes and the final construct and docking them with Toll-Like Receptor 4 (TLR4). In the experimental phase, the final multi-epitope construct was synthesized using the solid phase method, and it was then enclosed in lipid nanoparticles. After synthesizing the construct, its loading, average size distribution, and nanoliposome shape were checked using Nanodrop at 280 nm, dynamic light scattering (DLS), and atomic force microscope (AFM). The designed vaccine has been confirmed to be non-toxic and anti-allergic. It can bind with different MHC alleles at a rate of 99.05%. The construct loading was determined to be about 91%, with an average size of 54 nm. Spherical shapes were also observed in the AFM images. Further laboratory tests are necessary to confirm the safety and immunogenicity of the multi-epitope vaccine.


Subject(s)
Bacterial Vaccines , Computational Biology , Helicobacter pylori , Nanoparticles , Helicobacter pylori/immunology , Nanoparticles/chemistry , Bacterial Vaccines/immunology , Bacterial Vaccines/chemistry , Computational Biology/methods , Humans , Bacterial Proteins/immunology , Bacterial Proteins/chemistry , Epitopes/immunology , Epitopes/chemistry , Molecular Docking Simulation , Antigens, Bacterial/immunology , Antigens, Bacterial/chemistry , Helicobacter Infections/prevention & control , Helicobacter Infections/immunology , Toll-Like Receptor 4/immunology , Urease/immunology , Urease/chemistry , Immunoinformatics , Liposomes
15.
Appl Microbiol Biotechnol ; 97(6): 2367-78, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23064457

ABSTRACT

Urease is an essential virulence factor and colonization factor for Helicobacter pylori, of which the urease B subunit (UreB) is considered as an excellent vaccine candidate antigen. In previous study, an epitope vaccine with cholera toxin B subunit (CTB) and an epitope (UreB321-339) named CtUBE was constructed and the mice were protected significantly after intragastric vaccination with the CtUBE liposome vaccine. However, the fusion protein CtUBE was expressed as inclusion bodies and was difficultly purified. Besides, the immunogenicity and specificity of the CtUBE vaccine was not investigated in a fairly wide and detailed way. In this study, the fusion peptide CtUBE was reconstructed and expressed as a soluble protein with pectinase signal peptide at the N terminus and the 6-his tag at its C-terminal, and then the immunogenicity, specificity, prophylactic, and therapeutic efficacy of the reconstructed CtUBE (rCtUBE) vaccine were evaluated in BALB/c mice model after purification. The experimental results indicated that mice immunized with rCtUBE could produce comparatively high level of specific antibodies which could respond to natural H. pylori urease, UreB, or the minimal epitope UreB327-334 involved with the active site of urease, and showed effectively inhibitory effect on the enzymatic activity of urease. Besides, oral prophylactic or therapeutic immunization with rCtUBE significantly decreased H. pylori colonization compared with oral immunization with rCTB or PBS, and the protection was correlated with antigen-specific IgG, IgA, and mucosal sIgA antibody responses, and a Th2 cells response. This rCtUBE vaccine may be a promising vaccine candidate for the control of H. pylori infection.


Subject(s)
Bacterial Vaccines/immunology , Epitopes/immunology , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Urease/immunology , Administration, Oral , Animals , Antibodies, Bacterial/analysis , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Cholera Toxin/genetics , Cholera Toxin/immunology , Disease Models, Animal , Epitopes/genetics , Helicobacter Infections/therapy , Helicobacter pylori/genetics , Immunity, Mucosal , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Serum/immunology , Urease/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Virulence Factors/genetics , Virulence Factors/immunology
16.
Vet Res ; 43: 72, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23101660

ABSTRACT

Helicobacter (H.) suis is a porcine and human gastric pathogen. Previous studies in mice showed that an H. suis infection does not result in protective immunity, whereas immunization with H. suis whole-cell lysate (lysate) protects against a subsequent experimental infection. Therefore, two-dimensional gel electrophoresis of H. suis proteins was performed followed by immunoblotting with pooled sera from H. suis- infected mice or mice immunized with lysate. Weak reactivity against H. suis proteins was observed in post-infection sera. Sera from lysate-immunized mice, however, showed immunoreactivity against a total of 19 protein spots which were identified using LC-MS/MS. The H. suis urease subunit B (UreB) showed most pronounced reactivity against sera from lysate-immunized mice and was not detected with sera from infected mice. None of the pooled sera detected H. suis neutrophil-activating protein A (NapA). The protective efficacy of intranasal vaccination of BALB/c mice with H. suis UreB and NapA, both recombinantly expressed in Escherichia coli (rUreB and rNapA, respectively), was compared with that of H. suis lysate. All vaccines contained choleratoxin as adjuvant. Immunization of mice with rUreB and lysate induced a significant reduction of H. suis colonization compared to non-vaccinated H. suis-infected controls, whereas rNapA had no significant protective effect. Probably, a combination of local Th1 and Th17 responses, complemented by antibody responses play a role in the protective immunity against H. suis infections.


Subject(s)
Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Helicobacter Infections/prevention & control , Helicobacter heilmannii/enzymology , Urease/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Antibody Formation , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Vaccines/administration & dosage , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Chromatography, Liquid/veterinary , Cytokines/immunology , Electrophoresis, Gel, Two-Dimensional/veterinary , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Immunoblotting/veterinary , Mice , Mice, Inbred BALB C , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Stomach/immunology , Stomach/microbiology , Stomach/pathology , Tandem Mass Spectrometry/veterinary , Urease/genetics
17.
Appl Microbiol Biotechnol ; 93(5): 1937-45, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22134639

ABSTRACT

Epitope vaccine based on urease of Helicobacter pylori is a promising option for prophylactic and therapeutic vaccination against H. pylori infection. In this study, we constructed an epitope vaccine with mucosal adjuvant cholera toxin B subunit (CTB) and an epitope (UreA(183-203)) of H. pylori urease A subunit named CTB-UA. The CTB-UA fusion protein was expressed in Escherichia coli, and the purified protein was used for intraperitoneal immunization experiments in BALB/c mice. The experimental results indicated that anti-CTB-UA antibody could recognize both H. pylori urease A subunit (UreA) and urease B subunit (UreB). Besides, the CTB-UA epitope vaccine had good immunogenicity and immunoreactivity and could induce specific neutralizing antibodies which showed effectively inhibitory effect on the enzymatic activity of H. pylori urease. CTB-UA is a promising molecule to be investigated as H. pylori vaccine antigen candidate.


Subject(s)
Bacterial Vaccines/immunology , Cholera Toxin/immunology , Epitopes, B-Lymphocyte/immunology , Helicobacter pylori/enzymology , Helicobacter pylori/immunology , Urease/antagonists & inhibitors , Urease/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Bacterial Vaccines/genetics , Cholera Toxin/genetics , Epitopes, B-Lymphocyte/genetics , Escherichia coli/genetics , Gene Expression , Helicobacter Infections/prevention & control , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
18.
Appl Microbiol Biotechnol ; 95(6): 1437-44, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22569640

ABSTRACT

Epitope vaccine based on the enzyme urease of Helicobacter pylori is a promising option for prophylactic and therapeutic vaccination against H. pylori infection. In our previous study, the epitope vaccine CTB-UA, which was composed of the mucosal adjuvant cholera toxin B subunit (CTB) and an epitope (UreA183₋203) from the H. pylori urease A subunit (UreA) was constructed. This particular vaccine was shown to have good immunogenicity and immunoreactivity and could induce specific neutralizing antibodies, which exhibited effectively inhibitory effects on the enzymatic activity of H. pylori urease. In this study, the prophylactic and therapeutic efficacy of the epitope vaccine CTB-UA was evaluated in a BALB/c mice model. The experimental results indicated that oral prophylactic or therapeutic immunization with CTB-UA significantly decreased H. pylori colonization compared with oral immunization with PBS. The results also revealed that the protection was correlated with antigen-specific IgG, IgA, and mucosal secretory IgA antibody responses. CTB-UA may be a promising vaccine candidate for the control of H. pylori infection.


Subject(s)
Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Cholera Toxin/immunology , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Urease/immunology , Animals , Antibodies, Bacterial/immunology , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Cholera Toxin/administration & dosage , Cholera Toxin/genetics , Disease Models, Animal , Helicobacter Infections/drug therapy , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter pylori/enzymology , Helicobacter pylori/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Urease/administration & dosage , Urease/genetics , Vaccination
19.
BMC Microbiol ; 11: 183, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21843372

ABSTRACT

BACKGROUND: Nontypeable Haemophilus influenzae is a common cause of otitis media in children and lower respiratory tract infection in adults with chronic obstructive pulmonary disease (COPD). Prior studies have shown that H. influenzae expresses abundant urease during growth in the middle ear of the chinchilla and in pooled human sputum, suggesting that expression of urease is important for colonization and infection in the hostile environments of the middle ear and in the airways in adults. Virtually nothing else is known about the urease of H. influenzae, which was characterized in the present study. RESULTS: Analysis by reverse transcriptase PCR revealed that the ure gene cluster is expressed as a single transcript. Knockout mutants of a urease structural gene (ureC) and of the entire ure operon demonstrated no detectable urease activity indicating that this operon is the only one encoding an active urease. The ure operon is present in all strains tested, including clinical isolates from otitis media and COPD. Urease activity decreased as nitrogen availability increased. To test the hypothesis that urease is expressed during human infection, purified recombinant urease C was used in ELISA with pre acquisition and post infection serum from adults with COPD who experienced infections caused by H. influenzae. A total of 28% of patients developed new antibodies following infection indicating that H. influenzae expresses urease during airway infection. Bacterial viability assays performed at varying pH indicate that urease mediates survival of H. influenzae in an acid environment. CONCLUSIONS: The H. influenzae genome contains a single urease operon that mediates urease expression and that is present in all clinical isolates tested. Nitrogen availability is a determinant of urease expression. H. influenzae expresses urease during human respiratory tract infection and urease is a target of the human antibody response. Expression of urease enhances viability in an acid environment. Taken together, these observations suggest that urease is important for survival and replication of H. influenzae in the human respiratory tract.


Subject(s)
Acids/toxicity , Haemophilus Infections/microbiology , Haemophilus influenzae/enzymology , Haemophilus influenzae/pathogenicity , Microbial Viability/drug effects , Respiratory Tract Infections/microbiology , Urease/biosynthesis , Adult , Animals , Antibodies, Bacterial/blood , Enzyme-Linked Immunosorbent Assay , Gene Deletion , Gene Expression , Gene Expression Profiling , Haemophilus Infections/immunology , Haemophilus influenzae/immunology , Humans , Multigene Family , Respiratory Tract Infections/immunology , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Urease/genetics , Urease/immunology
20.
Mol Biol Rep ; 38(5): 3173-80, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20127514

ABSTRACT

For mass production of urease B subunit (UreB) and heat shock protein A subunit (HspA) of Helicobacter pylori with Bombyx mori nuclear polyhedrosis virus (BmNPV) baculovirus expression system (BES) and to determine whether they could be used as an oral vaccine against H. pylori, besides, to determine the time course of expressed recombinant protein and the optimum acquisition time directly through green fluorescence, HspA and enhanced green fluorescence protein (EGFP) genes were cloned into vector pFastBacDual to form donor vector pFastBacDual-(EGFP) (HspA), UreB gene was cloned into vector pFastBacDual to form donor vector pFastBacDual-UreB,then they were transformed into E. coli BmDH10Bac to obtain the recombinant Bacmid-(EGFP) (HspA) and Bacmid-UreB respectively. They were used to transfect BmN cells and generated the recombinant baculovirus BmNPV-(EGFP) (HspA) and BmNPV-UreB. Using these recombinant baculovirus BmNPV-(EGFP) (HspA) and BmNPV-UreB inoculated the silkworm pupae, a recombinant HspA and UreB protein were expressed in silkworm pupae, which were around 13 and 62 kDa in sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and western blot analysis. After oral immunization of mice, serum specific IgG antibodies against HspA and UreB in vaccine group were much higher than that in mock and native silkworm powder control groups. The results indicated that the expressed recombinant HspA and UreB in silkworm pupae would possess good immunogenicity. In addition, when EGFP and HspA proteins were expressed, a direct correlation between the increase in intensity of fluorescence and HspA concentration.


Subject(s)
Bacterial Proteins/immunology , Bombyx/microbiology , Heat-Shock Proteins/immunology , Helicobacter pylori/immunology , Protein Subunits/immunology , Pupa/microbiology , Urease/immunology , Animals , Bacterial Proteins/genetics , Bombyx/growth & development , Bombyx/immunology , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heat-Shock Proteins/genetics , Helicobacter pylori/chemistry , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Protein Subunits/chemistry , Protein Subunits/genetics , Pupa/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Urease/chemistry , Urease/genetics
SELECTION OF CITATIONS
SEARCH DETAIL