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










Publication year range
1.
Autophagy ; 11(8): 1293-307, 2015.
Article in English | MEDLINE | ID: mdl-26151773

ABSTRACT

Burkholderia pseudomallei is the causative agent of melioidosis, a disease with high mortality, which is prevalent in tropical regions of the world. A recent study shows that B. pseudomallei can survive inside mammalian cells because of its ability to actively evade cell autophagy. However, the underlying mechanisms remain unclear. In the present study, based on microarray screening, we found that ATG10 was downregulated following B. pseudomallei infection in A549 human lung epithelial cells. Forced expression of ATG10 accelerated the elimination of intracellular B. pseudomallei by enhancing the process of autophagy. Moreover, MIR4458, MIR4667-5p, and MIR4668-5p were found, by microarray screening, to be upregulated in response to B. pseudomallei infection. These 3 novel miRNAs, MIR4458, MIR4667-5p, and MIR4668-5p, targeted to the 3'-untranslated region of ATG10 in different time-course and spatial manners. Upregulation of these miRNAs reduced the level of ATG10 and inhibited autophagy, leading to increasing survival rate of intracellular B. pseudomallei. Furthermore, the increase of these miRNAs was correlated with the reduced promoter methylation status in A549 cells in response to B. pseudomallei infection. Our results reveal that 3 novel miRNAs regulate autophagy-mediated elimination of B. pseudomallei by targeting ATG10, and provide potential targets for clinical treatment.


Subject(s)
Burkholderia pseudomallei/pathogenicity , Epithelial Cells/cytology , Lung/microbiology , MicroRNAs/metabolism , Vesicular Transport Proteins/metabolism , 3' Untranslated Regions , Autophagy-Related Proteins , Base Sequence , Cell Line , Cell Line, Tumor , Cell Survival , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Epithelial Cells/microbiology , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Lung/cytology , Lung/pathology , MicroRNAs/genetics , Microscopy, Confocal , Microscopy, Electron, Transmission , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Plasmids/metabolism , Promoter Regions, Genetic , Up-Regulation
2.
Lancet ; 386(10002): 1457-64, 2015 Oct 10.
Article in English | MEDLINE | ID: mdl-26142048

ABSTRACT

BACKGROUND: Helicobacter pylori is one of the most common gastric pathogens, affecting at least half the world's population, and is strongly associated with gastritis, peptic ulcer, gastric adenocarcinoma, and lymphoma. We aimed to assess the efficacy, safety, and immunogenicity of a three-dose oral recombinant H pylori vaccine in children in China. METHODS: We did this randomised, double-blind, placebo-controlled, phase 3 trial at one centre in Ganyu County, Jiangsu Province, China. Healthy children aged 6-15 years without past or present H pylori infection were randomly assigned (1:1), via computer-generated randomisation codes in blocks of ten, to receive the H pylori vaccine or placebo. Participants, their guardians, and study investigators were masked to treatment allocation. The primary efficacy endpoint was the occurrence of H pylori infection within 1 year after vaccination. We did analysis in the per-protocol population. This trial is registered with ClinicalTrials.gov, number NCT02302170. FINDINGS: Between Dec 2, 2004, and March 19, 2005, we randomly assigned 4464 participants to either the vaccine group (n=2232) or the placebo group (n=2232), of whom 4403 (99%) participants completed the three-dose vaccination schedule and were included in the per-protocol efficacy analysis. We extended follow-up to 3 years. We recorded 64 events of H pylori infection within the first year (14 events in 2074·3 person-years at risk in the vaccine group vs 50 events in 2089·6 person-years at risk in the placebo group), resulting in a vaccine efficacy of 71·8% (95% CI 48·2-85·6). 157 (7%) participants in the vaccine group and 161 (7%) participants in the placebo group reported at least one adverse reaction. Serious adverse events were reported in five (<1%) participants in the vaccine group and seven (<1%) participants in the placebo group, but none was considered to be vaccination related. INTERPRETATION: The oral recombinant H pylori vaccine was effective, safe, and immunogenic in H pylori-naive children. This vaccine could substantially reduce the incidence of H pylori infection; however, follow up over a longer period is needed to confirm the protection of the vaccine against H pylori-associated diseases. FUNDING: Chongqing Kangwei Biological Technology.


Subject(s)
Bacterial Vaccines/administration & dosage , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Administration, Oral , Adolescent , Age Factors , Bacterial Vaccines/adverse effects , Bacterial Vaccines/immunology , Child , Double-Blind Method , Female , Helicobacter Infections/immunology , Humans , Immunity, Active/immunology , Male , Recombinant Proteins , Sex Factors , Treatment Outcome
3.
Autophagy ; 11(2): 344-54, 2015.
Article in English | MEDLINE | ID: mdl-25831014

ABSTRACT

Shiga toxins (Stxs) are a family of cytotoxic proteins that lead to the development of bloody diarrhea, hemolytic-uremic syndrome, and central nervous system complications caused by bacteria such as S. dysenteriae, E. coli O157:H7 and E. coli O104:H4. Increasing evidence indicates that macroautophagy (autophagy) is a key factor in the cell death induced by Stxs. However, the associated mechanisms are not yet clear. This study showed that Stx2 induces autophagic cell death in Caco-2 cells, a cultured line model of human enterocytes. Inhibition of autophagy using pharmacological inhibitors, such as 3-methyladenine and bafilomycin A1, or silencing of the autophagy genes ATG12 or BECN1 decreased the Stx2-induced death in Caco-2 cells. Furthermore, there were numerous instances of dilated endoplasmic reticulum (ER) in the Stx2-treated Caco-2 cells, and repression of ER stress due to the depletion of viable candidates of DDIT3 and NUPR1. These processes led to Stx2-induced autophagy and cell death. Finally, the data showed that the pseudokinase TRIB3-mediated DDIT3 expression and AKT1 dephosphorylation upon ER stress were triggered by Stx2. Thus, the data indicate that Stx2 causes autophagic cell death via the ER stress pathway in intestinal epithelial cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Epithelial Cells/drug effects , Shiga Toxins/pharmacology , Animals , Cell Death/drug effects , Cells, Cultured , Epithelial Cells/cytology , Escherichia coli , Humans , Mice, Inbred C57BL , Transcription Factor CHOP
4.
Gastroenterology ; 143(4): 951-62.e8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22710190

ABSTRACT

BACKGROUND & AIMS: CD8(+) T cells that produce interleukin (IL)-17 (Tc17 cells) promote inflammation and have been identified in tumors. We investigated their role in the pathogenesis of gastric cancer. METHODS: We used flow cytometry analyses to determine levels and phenotype of Tc17 cells in blood and tumor samples from 103 patients with gastric cancer. We performed multivariate analysis to identify factors associated with overall survival using the Cox proportional hazards model. CD8(+) T cells and monocytes were isolated and cocultured in an assay for induction of Tc17 cells. Tumor cells and myeloid-derived suppressor cells (MDSCs) were isolated and used in assays of Tc17 cell function. RESULTS: Tc17 cells with distinct cytokine and functional profiles were found in gastric tumor samples from patients. The percentage of Tc17 cells increased with tumor progression and was associated with overall survival time. Tumor-activated monocytes secreted IL-6, IL-1ß, and IL-23, which promoted development of Tc17 cell populations. Supernatants from cultured Tc17 cells induced production of the chemokine CXCL12 by tumor cells; this promoted CXCR4-dependent migration of MDSCs and impaired functions of anti-tumor CD8(+) cytotoxic T cells via a cell contact-dependent mechanism. CONCLUSIONS: Percentages of Tc17 cells in gastric tumors are associated with survival times of patients. These cells promote chemotaxis of MDSCs, which might promote tumor progression.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Interleukin-17/biosynthesis , Receptors, CXCR4/metabolism , Stomach Neoplasms/immunology , Stomach Neoplasms/metabolism , Aged , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Movement , Chemokine CXCL12/metabolism , Chemotaxis , Coculture Techniques , Dendritic Cells/metabolism , Female , Flow Cytometry , Granulocytes/metabolism , Humans , Immune Tolerance , Interleukin-1beta/metabolism , Interleukin-23/metabolism , Interleukin-6/metabolism , Macrophages/metabolism , Male , Middle Aged , Monocytes/metabolism , Multivariate Analysis , Myeloid Progenitor Cells/metabolism , Phenotype , Prognosis , Proportional Hazards Models , Stomach Neoplasms/pathology
5.
Clin Immunol ; 138(1): 77-84, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21075691

ABSTRACT

Enterohemorrhagic E. coli (EHEC) causes severe diseases in humans and animals via the production of Shiga toxins, and injection of effectors into epithelia using type III secretion system (TTSS). E. coli secreted protein A (EspA) forms the filamentous conduits of TTSS, which extends into the translocation pore embedded in host cell membranes and aids in the transportation of bacterial effectors. In addition, EspA is closely associated with initial bacterial adhesion and the formation of biofilms. EspA in its various forms elicits protective immune responses, although the epitope responsible has not to be identified. Here we report the presence of a linear, immunogenic, conserved and partially protective epitope E07 (100Lys-120Val) on EspA, which is recognized by the novel monoclonal antibody 1H10. This antibody blocks EHEC-induced actin polymerization and confers protection in mice. These findings provide a better understanding of EspA-induced immune responses and could lead to epitope-based vaccines and antibody-based therapies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Enterohemorrhagic Escherichia coli/immunology , Epitopes/immunology , Escherichia coli Proteins/immunology , Actins/metabolism , Amino Acid Sequence/genetics , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Antigen-Antibody Reactions/immunology , Conserved Sequence/genetics , Conserved Sequence/immunology , Enterohemorrhagic Escherichia coli/drug effects , Enterohemorrhagic Escherichia coli/genetics , Epitope Mapping , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Escherichia coli O157/drug effects , Escherichia coli O157/genetics , Escherichia coli O157/immunology , Escherichia coli Proteins/genetics , Feces/microbiology , Female , HeLa Cells , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/immunology , Polymerization/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccination
6.
J Microbiol ; 48(2): 223-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20437155

ABSTRACT

A multivalent fusion vaccine is a promising option for protection against Helicobacter pylori infection. In this study, UreB414 was identified as an antigenic fragment of urease B subunit (UreB) and it induced an antibody inhibiting urease activity. Immunization with UreB414 partially protected mice from H. pylori infection. Furthermore, a trivalent fusion vaccine was constructed by genetically linking heat shock protein A (HspA), H. pylori adhesin A (HpaA), and UreB414, resulting in recombinant HspA-HpaA-UreB414 (rHHU). Its protective effect against H. pylori infection was tested in BALB/c mice. Oral administration of rHHU significantly protected mice from H. pylori infection, which was associated with H. pylori-specific antibody production and Th1/Th2-type immune responses. The results show that a trivalent fusion vaccine efficiently combats H. pylori infection, and that an antigenic fragment of the protein can be used instead of the whole protein to construct a multivalent vaccine.


Subject(s)
Adhesins, Bacterial/immunology , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Heat-Shock Proteins/immunology , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Urease/immunology , Adhesins, Bacterial/genetics , Administration, Oral , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Female , Heat-Shock Proteins/genetics , Helicobacter Infections/immunology , Helicobacter pylori/genetics , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Stomach/microbiology , Stomach/pathology , Urease/genetics , Virulence Factors/genetics , Virulence Factors/immunology
7.
J Infect Dis ; 200(6): 916-25, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19650740

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are small, noncoding RNAs that regulate gene expression at posttranscriptional level. H. pylori is a major human pathogenic bacterium in gastric mucosa. To date, the role of miRNAs in response to H. pylori infection has not been explored. METHODS: The expression profile of cellular miRNAs during H. pylori infection was analyzed by using microarray and quantitative reverse-transcriptase polymerase chain reaction. The potential target of miR-155 was identified by luciferase assay and Western blot. Promoter analysis and inhibitor experiment were used to investigate the pathway involved in the induction of miR-155. Examination of miR-155 function was performed by overexpression and inhibition of miR-155. RESULTS: H. pylori was able to increase the miR-155 expression in gastric epithelial cell lines and gastric mucosal tissues, and nuclear factor-kappaB (NF-kappaB) and activator protein-1 (AP-1) pathway were required for the induction of miR-155. miR-155 may down-regulate IkappaB kinase epsilon, Sma- and Mad-related protein 2 (SMAD2), and Fas-associated death domain protein. Furthermore, the overexpression of miR-155 negatively regulated the release of interleukin-8 and growth-related oncogene-alpha. CONCLUSIONS: This study provides the first description of increased expression of miR-155 in H. pylori infection, and miR-155 may function as novel negative regulator that help to fine-tune the inflammation response of H. pylori infection.


Subject(s)
Helicobacter Infections/microbiology , Helicobacter pylori , Inflammation/metabolism , MicroRNAs/metabolism , Adult , Chemokine CXCL1/metabolism , Fas-Associated Death Domain Protein/metabolism , Female , Gastric Mucosa/metabolism , Gene Expression Profiling , Gene Expression Regulation , Helicobacter Infections/metabolism , Humans , I-kappa B Kinase/metabolism , Interleukin-8/metabolism , Male , Middle Aged , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Smad2 Protein/metabolism
8.
Curr Microbiol ; 57(4): 281-6, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18587616

ABSTRACT

Helicobacter pylori, an etiological agent of gastroduodenal diseases, undergoes drastic morphological transition from spiral shape to coccoid form under oxidative stress. However, the knowledge of the specific expression profile in response to oxidative stress is relatively limited. Here, we report global proteomic analysis of H. pylori coccoids under oxidative stress. Two-dimensional gel electrophoresis analysis of H. pylori featuring coccoid revealed that 10 unique protein spots exhibit different expression profiles with comparison of that under normal microaerophilic condition. In total, seven proteins including superoxide dismutase, alkyl hydroperoxide reductase, urease G, and so forth were confirmed using matrix-assisted laser desorption/ionization time-of-flight/mass spectroscopy and then validated by reverse transcription-polymerase chain reaction, indicating that they play key roles in the physiological adaptation mechanisms of H. pylori to oxygen challenge. These data provide preliminary insights into H. pylori on coccoid generation under oxidative stress.


Subject(s)
Bacterial Proteins/metabolism , Heat-Shock Response , Helicobacter pylori/physiology , Helicobacter pylori/ultrastructure , Oxidative Stress , Proteomics , Bacterial Proteins/genetics , Electrophoresis, Gel, Two-Dimensional , Gene Expression Regulation, Bacterial , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Humans , Microscopy, Electron, Transmission , Proteome , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
Helicobacter ; 13(3): 191-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18466394

ABSTRACT

BACKGROUND: Development of Helicobacter pylori vaccine would be a new effective strategy for prevention and treatment of H. pylori infection. Recombinant H. pylori vaccine comprising a single subunit antigen can only induce immune response with limited protection efficiency. In this study, the protective effect of H. pylori multicomponent vaccines consisting of three recombinant subunit antigens was investigated using the Mongolian gerbil model. MATERIALS AND METHODS: Mongolian gerbils were immunized with different formulations of three recombinant H. pylori antigens (UreB, HspA, and HpaA) with two different adjuvants (Al(OH)3, LT(R72DITH)) by intragastric (i.g.) or intramuscular (i.m.) routes. The protective effects of multicomponent vaccines were assessed after H. pylori challenge in different studies. The specific IgG antibodies in serum were monitored by ELISA, and the mRNA expressions of IL-4 and IFN-gamma in spleen tissue were detected by reverse transcribed polymerase chain reaction (RT-PCR). RESULTS: The protective effect against H. pylori challenge in gerbils immunized with three recombinant antigens and LT(R72DITH) or Al(OH)3 was significantly higher than that in single- or double-antigen vaccine-immunized and control gerbils. Furthermore, the protective effect of the triple-antigen vaccine combined with the LT(R72DITH) adjuvant (average 86.3%) was significantly greater than that of vaccine combined with the Al(OH)3 adjuvant (average 53.4%). After the first immunization, the anti-UreB/HspA/HpaA serum IgG level in gerbils immunized with triple-antigen vaccine combined with Al(OH)3 was higher than that in gerbils immunized with the vaccine combined with LT(R72DITH). Splenic interferon (IFN)-gamma and interleukin (IL)-4 transcript levels were significantly increased in LT(R72DITH) vaccine-immunized gerbils as compared to the Al(OH)3 vaccine group. Moreover, splenic IL-4 mRNA levels were higher than IFN-gamma in gerbils immunized with triple-antigen vaccine with either LT(R72DITH) or Al(OH)3. CONCLUSIONS: This study indicated that the recombinant multicomponent vaccine provided effective protection against H. pylori infection as compared to the single-antigen vaccine. This protective immunity would be closely associated with a predominant Th2-type response.


Subject(s)
Bacterial Vaccines/administration & dosage , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Animals , Antigens, Bacterial/immunology , Bacterial Vaccines/therapeutic use , Female , Gastritis/microbiology , Gerbillinae , Humans , Th2 Cells/immunology
10.
Wei Sheng Wu Xue Bao ; 47(3): 461-4, 2007 Jun.
Article in Chinese | MEDLINE | ID: mdl-17672306

ABSTRACT

To study adaptive protein variation of H. pylori after colonization in Mongolian gerbils, Firstly, Clinical isolated strain M0 of H. pylori were inoculated into Mongolian gerbils and acclimated through serial passages in vivo for procuring an adaptive colonization H. pylori strain. Then, two-dimensional electrophoresis (2-DE) and mass spectrometry (MS) was taken to separate and identify the global proteins significantly changed between H. pylori strain M0 and the adaptive strain. Through serial passages the infectious rate increased from about 2/10 to 9/10 and a adaptive colonization strain M13 has been obtained. Comparative proteomic technology display that the proteinogram of H. pylori have changed after colonization in gerbils. Out of 5 differential protein spots cut out of gels for MALDI-TOF-MS identification, 4 spots were successfully identified, among which, Icd, RfaD and HP0318 were significantly higher in M13 compared with M0, while only HypB were found in M13. So far, HP0318 is a conserved hypothetical protein. These proteins may be important factors of H. pylori for adaptive colonization.


Subject(s)
Helicobacter Infections/microbiology , Helicobacter pylori/chemistry , Helicobacter pylori/growth & development , Proteomics , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Electrophoresis, Gel, Two-Dimensional , Gerbillinae , Helicobacter pylori/metabolism , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...