25 research outputs found

    Cell-Cycle Inhibition by Helicobacter pylori L-Asparaginase

    Get PDF
    Helicobacter pylori (H. pylori) is a major human pathogen causing chronic gastritis, peptic ulcer, gastric cancer, and mucosa-associated lymphoid tissue lymphoma. One of the mechanisms whereby it induces damage depends on its interference with proliferation of host tissues. We here describe the discovery of a novel bacterial factor able to inhibit the cell-cycle of exposed cells, both of gastric and non-gastric origin. An integrated approach was adopted to isolate and characterise the molecule from the bacterial culture filtrate produced in a protein-free medium: size-exclusion chromatography, non-reducing gel electrophoresis, mass spectrometry, mutant analysis, recombinant protein expression and enzymatic assays. L-asparaginase was identified as the factor responsible for cell-cycle inhibition of fibroblasts and gastric cell lines. Its effect on cell-cycle was confirmed by inhibitors, a knockout strain and the action of recombinant L-asparaginase on cell lines. Interference with cell-cycle in vitro depended on cell genotype and was related to the expression levels of the concurrent enzyme asparagine synthetase. Bacterial subcellular distribution of L-asparaginase was also analysed along with its immunogenicity. H. pylori L-asparaginase is a novel antigen that functions as a cell-cycle inhibitor of fibroblasts and gastric cell lines. We give evidence supporting a role in the pathogenesis of H. pylori-related diseases and discuss its potential diagnostic application

    Helicobacter pylori L-asparaginase: A Novel Bacterial Antigen that May Contribute to Infection Detection

    No full text
    Helicobacter pylori is responsible for gastric inflammation and for an increased risk of cancer development in humans. Several bacterial antigens contribute to stimulate the immune system, but their relative role has not yet been defined. H. pylori (strain CCUG) type II L-asparaginase (L-ASNase) induces an immune response in mice. To verify if an immune response could also be detected in humans, sera positive (n=11) or negative (n=11), respectively, to H. pylori according to a commercial test were assayed for their reactivity towards purified H. pylori L-ASNase. Among positive samples, 8/11 (72%) were positive to L-ASNase. We conclude that H. pylori L-ASNase is immunogenic in humans and contributes to the generation of the antibody response induced by the bacterium

    Helicobacter pylori L-asparaginase: A promising chemotherapeutic agent

    No full text
    Bacterial L -asparaginases are amidohydrolases that catalyse the conversion of L -asparagine to L -aspartate and ammonia and are used as anti-cancer drugs. The current members of this class of drugs have several toxic side effects mainly due to their associated glutaminase activity. In the present study, we report the molecular cloning, biochemical characterisation and in vitro cytotoxicity of a novel L -asparaginase from the pathogenic strain Helicobacter pylori CCUG 17874. The recombinant enzyme showed a strong preference for L -asparagine over L -glutamine and, in contrast to most L -asparaginases, it exhibited a sigmoidal behaviour towards L -glutamine. The enzyme preserved full activity after 2 h incubation at 45 C. In vitro cytotoxicity assays revealed that different cell lines displayed a variable sensitivity towards the enzyme, AGS and MKN28 gastric epithelial cells being the most affected. These findings may be relevant both for the interpretation of the mechanisms underlying H. pylori associated diseases and for biomedical applications
    corecore