Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Microbiol Spectr ; 12(6): e0001524, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38682907

ABSTRACT

Gastric cancer is the fifth most diagnosed cancer in the world. Infection by the bacteria Helicobacter pylori (HP) is associated with approximately 75% of gastric cancer cases. HP infection induces chronic gastric inflammation, damaging the stomach and fostering carcinogenesis. Most mechanistic studies on gastric cancer initiation are performed in mice and utilize either mouse-adapted strains of HP or the natural mouse pathogen Helicobacter felis (HF). Here, we identified the differences in gastric inflammation, atrophy, and metaplasia associated with HP and HF infection in mice. PMSS1 HP strain or the CS1 HF strain were co-cultured with mouse peritoneal macrophages to assess their immunostimulatory effects. HP and HF induced similar cytokine production from cultured mouse peritoneal macrophages revealing that both bacteria exhibit similar immunostimulatory effects in vitro. Next, C57BL/6J mice were infected with HP or HF and were assessed 2 months post-infection. HP-infected mice caused modest inflammation within both the gastric corpus and antrum, and did not induce significant atrophy within the gastric corpus. In contrast, HF induced significant inflammation throughout the gastric corpus and antrum. Moreover, HF infection was associated with significant atrophy of the chief and parietal cell compartments and induced the expression of pyloric metaplasia (PM) markers. HP is poorly immunogenic compared to HF. HF induces dramatic CD4+ T cell activation, which is associated with increased gastric cancer risk in humans. Thus, HP studies in mice are better suited for studies on colonization, while HF is more strongly suited for studies on the effects of gastric inflammation on tumorigenesis. . IMPORTANCE: Mouse infection models with Helicobacter species are widely used to study Helicobacter pathogenesis and gastric cancer initiation. However, Helicobacter pylori is not a natural mouse pathogen, and mouse-adapted H. pylori strains are poorly immunogenic. In contrast, Helicobacter felis is a natural mouse pathogen that induces robust gastric inflammation and is often used in mice to investigate gastric cancer initiation. Although both bacterial strains are widely used, their disease pathogenesis in mice differs dramatically. However, few studies have directly compared the pathogenesis of these bacterial species in mice, and the contrasting features of these two models are not clearly defined. This study directly compares the gastric inflammation, atrophy, and metaplasia development triggered by the widely used PMSS1 H. pylori and CS1 H. felis strains in mice. It serves as a useful resource for researchers to select the experimental model best suited for their studies.


Subject(s)
Gastric Mucosa , Helicobacter Infections , Helicobacter felis , Helicobacter pylori , Metaplasia , Mice, Inbred C57BL , Animals , Helicobacter pylori/immunology , Helicobacter pylori/pathogenicity , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter Infections/immunology , Mice , Helicobacter felis/pathogenicity , Metaplasia/microbiology , Metaplasia/pathology , Gastric Mucosa/pathology , Gastric Mucosa/microbiology , Gastric Mucosa/immunology , Gastritis/microbiology , Gastritis/pathology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Inflammation/microbiology , Inflammation/pathology , Female , Cytokines/metabolism , Disease Models, Animal , Stomach/pathology , Stomach/microbiology
2.
Int J Oncol ; 58(3): 388-396, 2021 03.
Article in English | MEDLINE | ID: mdl-33469673

ABSTRACT

Helicobacter pylori (H. pylori) infection is a major risk factor for the development of gastric cancer. The authors previously demonstrated that in mice deficient in myeloid differentiation primary response 88 (Myd88­/­), infection with Helicobacter felis (H. felis) a close relative of H. pylori, subsequently rapidly progressed to neoplasia. The present study examined circulating tumor cells (CTCs) by measuring the expression of cytokeratins, epithelial­to­mesenchymal transition (EMT)­related markers and cancer stem cell (CSC) markers in bone marrow and peripheral blood from Myd88­/­ and wild­type (WT) mice. Cytokeratins CK8/18 were detected as early as 4 months post­infection in Myd88­/­ mice. By contrast, cytokeratins were not detected in WT mice even after 7 months post­infection. The expression of Mucin­1 (MUC1) was observed in both bone marrow and peripheral blood at different time points, suggesting its role in gastric cancer metastasis. Snail, Twist and ZEB were expressed at different levels in bone marrow and peripheral blood. The expression of these EMT­related markers suggests the manifestation of cancer metastasis in the early stages of disease development. LGR5, CD44 and CD133 were the most prominent CSC markers detected. The detection of CSC and EMT markers along with cytokeratins does reinforce their use as biomarkers for gastric cancer metastasis. This early detection of markers suggests that CTCs leave primary site even before cancer is well established. Thus, cytokeratins, EMT, and CSCs could be used as biomarkers to detect aggressive forms of gastric cancers. This information may prove to be of significance in stratifying patients for treatment prior to the onset of severe disease­related characteristics.


Subject(s)
Biomarkers, Tumor/analysis , Bone Marrow/pathology , Helicobacter Infections/pathology , Neoplastic Stem Cells/pathology , Stomach Neoplasms/diagnosis , Animals , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Helicobacter Infections/microbiology , Helicobacter felis/pathogenicity , Humans , Male , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Neoplastic Stem Cells/metabolism , Stomach Neoplasms/blood , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
3.
J Clin Invest ; 130(10): 5370-5379, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32663196

ABSTRACT

Chronic inflammation is deeply involved in various human disorders, such as cancer, neurodegenerative disorders, and metabolic disorders. Induction of epigenetic alterations, especially aberrant DNA methylation, is one of the major mechanisms, but how it is induced is still unclear. Here, we found that expression of TET genes, methylation erasers, was downregulated in inflamed mouse and human tissues, and that this was caused by upregulation of TET-targeting miRNAs such as MIR20A, MIR26B, and MIR29C, likely due to activation of NF-κB signaling downstream of IL-1ß and TNF-α. However, TET knockdown induced only mild aberrant methylation. Nitric oxide (NO), produced by NOS2, enhanced enzymatic activity of DNA methyltransferases (DNMTs), methylation writers, and NO exposure induced minimal aberrant methylation. In contrast, a combination of TET knockdown and NO exposure synergistically induced aberrant methylation, involving genomic regions not methylated by either alone. The results showed that a vicious combination of TET repression, due to NF-κB activation, and DNMT activation, due to NO production, is responsible for aberrant methylation induction in human tissues.


Subject(s)
DNA Methylation , DNA Modification Methylases/metabolism , Dioxygenases/metabolism , Animals , Dioxygenases/genetics , Disease Models, Animal , Down-Regulation , Epigenesis, Genetic , Gastritis/genetics , Gastritis/metabolism , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Helicobacter felis/pathogenicity , Helicobacter pylori , Humans , Inflammation/genetics , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Up-Regulation
4.
PLoS One ; 11(3): e0150411, 2016.
Article in English | MEDLINE | ID: mdl-26966907

ABSTRACT

Gastric mucosa-associated lymphoid tissue (MALT) lymphomas develop from a chronic Helicobacter infection. Phospholipase C gamma 2 (PLCG2) is important for B-cell survival and proliferation. We used BALB/c mice with a gain-of-function mutation in the Plcg2 gene (Ali5) to analyze its role in the development of gastric MALT lymphoma. Heterozygous BALB/c Plcg2Ali5/+ and wildtype (WT) mice were infected with Helicobacter felis (H. felis) and observed up to 16 months for development of gastric MALT lymphomas. In contrast to our initial hypothesis, Plcg2Ali5/+ mice developed MALT lymphomas less frequently than their WT littermates after long-term infection of 16 months. Infected Plcg2Ali5/+ mice showed downregulation of proinflammatory cytokines and decreased H. felis-specific IgG1 and IgG2a antibody responses. These results suggested a blunted immune response of Plcg2Ali5/+ mice towards H. felis infection. Intriguingly, Plcg2Ali5/+ mice harboured higher numbers of CD73 expressing regulatory T cells (Tregs), possibly responsible for impaired immune response towards Helicobacter infection. We suggest that Plcg2Ali5/+ mice may be protected from developing gastric MALT lymphomas as a result of elevated Treg numbers, reduced response to H. felis and decrease of proinflammatory cytokines.


Subject(s)
Helicobacter Infections/genetics , Helicobacter felis/pathogenicity , Lymphoma, B-Cell, Marginal Zone/virology , Phospholipase C gamma/genetics , Stomach Neoplasms/virology , Animals , Cytokines/metabolism , Gene Expression Regulation , Helicobacter Infections/immunology , Helicobacter Infections/veterinary , Immunoglobulin G/metabolism , Lymphoma, B-Cell, Marginal Zone/genetics , Lymphoma, B-Cell, Marginal Zone/immunology , Mice , Mice, Inbred BALB C , Mutation , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , T-Lymphocytes, Regulatory/metabolism
5.
Curr Protoc Pharmacol ; 69: 14.34.1-14.34.35, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26344212

ABSTRACT

Gastric adenocarcinoma is the fifth most common cancer and third most common cause of cancer-related death in the world. The majority of these cancers develop in genetically susceptible individuals who are chronically infected with the Gram-negative bacterium Helicobacter pylori. Often these individuals have also been exposed to certain environmental factors that increase susceptibility, such as dietary components. Murine models of Helicobacter-induced gastric cancer are valuable tools for investigating the mechanisms responsible for the stepwise pathological changes of chronic atrophic gastritis, intestinal metaplasia, dysplasia and gastric adenocarcinoma. Helicobacter felis colonization greatly accelerates the development of gastric neoplasia in mice, and causes pathologies similar to those observed with Helicobacter pylori-associated gastric carcinogenesis in humans. These mouse models are therefore useful for investigating genetic and environmental factors that may be involved in the pathogenesis and treatment of gastric cancer. Detailed in these protocols are procedures for inducing Helicobacter-associated carcinogenesis in mice as well as the histological analysis and interpretation of gastric pathology in these animals.


Subject(s)
Adenocarcinoma/microbiology , Disease Models, Animal , Helicobacter Infections/microbiology , Helicobacter felis/pathogenicity , Helicobacter pylori/pathogenicity , Paraneoplastic Syndromes/microbiology , Stomach Neoplasms/microbiology , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Anti-Bacterial Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/immunology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Gastric Mucosa/drug effects , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Helicobacter Infections/drug therapy , Helicobacter Infections/immunology , Helicobacter Infections/pathology , Helicobacter felis/drug effects , Helicobacter felis/immunology , Helicobacter pylori/drug effects , Helicobacter pylori/immunology , Humans , Mice , Paraneoplastic Syndromes/drug therapy , Paraneoplastic Syndromes/immunology , Paraneoplastic Syndromes/pathology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology
6.
Helicobacter ; 20(4): 284-90, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25510739

ABSTRACT

BACKGROUND: The Helicobacter heilmannii sensu lato (H. heilmannii s.l.) group consists of long, spiral-shaped bacteria naturally colonizing the stomach of animals. Moreover, bacteria belonging to this group have been observed in 0.2-6% of human gastric biopsy specimens, and associations have been made with the development of chronic gastritis, peptic ulceration, and gastric MALT lymphoma in humans. MATERIALS AND METHODS: To gain insight into the prevalence of H. heilmannii s.l. infections in patients suffering from gastric disease in China, H. heilmannii s.l. species-specific PCRs were performed on DNA extracts from rapid urease test (RUT)-positive gastric biopsies from 1517 patients followed by nucleotide sequencing. At the same time, Helicobacter pylori cultivation and specific PCR was performed to assess H. pylori infection in these patients. RESULTS: In total, H. heilmannii s.l. infection was detected in 11.87% (178/1499) of H. pylori-positive patients. The prevalence of H. suis, H. felis, H. bizzozeronii, H. heilmannii sensu stricto (s.s.), and H. salomonis in the patients was 6.94%, 2.20%, 0.13%, 0.07%, and 2.54%, respectively. Results revealed that all patients with H. heilmannii s.l. infection were co-infected with H. pylori, and some patients were co-infected with more than two different Helicobacter species. CONCLUSIONS: Helicobacter heilmannii s.l. infections are fairly common in Chinese patients. This should be kept in mind when diagnosing the cause of gastric pathologies in patients. Helicobacter suis was shown to be by far the most prevalent H. heilmannii s.l.species.


Subject(s)
Helicobacter Infections/epidemiology , Helicobacter felis/isolation & purification , Helicobacter heilmannii/isolation & purification , Helicobacter pylori/isolation & purification , Stomach Diseases/microbiology , Adult , Base Sequence , China/epidemiology , Coinfection/microbiology , DNA, Bacterial/genetics , Female , Gastric Mucosa/microbiology , Gastritis/microbiology , Helicobacter Infections/microbiology , Helicobacter felis/genetics , Helicobacter felis/pathogenicity , Helicobacter heilmannii/genetics , Helicobacter heilmannii/pathogenicity , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Humans , Male , Molecular Typing , Sequence Analysis, DNA
7.
Gastroenterology ; 147(2): 396-406.e7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24751878

ABSTRACT

BACKGROUND & AIMS: Bone morphogenetic protein (BMP)4 is a mesenchymal peptide that regulates cells of the gastric epithelium. We investigated whether BMP signaling pathways affect gastric inflammation after bacterial infection of mice. METHODS: We studied transgenic mice that express either the BMP inhibitor noggin or the ß- galactosidase gene under the control of a BMP-responsive element and BMP4(ßgal/+) mice. Gastric inflammation was induced by infection of mice with either Helicobacter pylori or Helicobacter felis. Eight to 12 weeks after inoculation, gastric tissue samples were collected and immunohistochemical, quantitative, reverse-transcription polymerase chain reaction and immunoblot analyses were performed. We used enzyme-linked immunosorbent assays to measure cytokine levels in supernatants from cultures of mouse splenocytes and dendritic cells, as well as from human gastric epithelial cells (AGS cell line). We also measured the effects of BMP-2, BMP-4, BMP-7, and the BMP inhibitor LDN-193189 on the expression of interleukin (IL)8 messenger RNA by AGS cells and primary cultures of canine parietal and mucus cells. The effect of BMP-4 on NFkB activation in parietal and AGS cells was examined by immunoblot and luciferase assays. RESULTS: Transgenic expression of noggin in mice increased H pylori- or H felis-induced inflammation and epithelial cell proliferation, accelerated the development of dysplasia, and increased expression of the signal transducer and activator of transcription 3 and activation-induced cytidine deaminase. BMP-4 was expressed in mesenchymal cells that expressed α-smooth muscle actin and activated BMP signaling pathways in the gastric epithelium. Neither BMP-4 expression nor BMP signaling were detected in immune cells of C57BL/6, BRE-ß-galactosidase, or BMP-4(ßgal/+) mice. Incubation of dendritic cells or splenocytes with BMP-4 did not affect lipopolysaccharide-stimulated production of cytokines. BMP-4, BMP-2, and BMP-7 inhibited basal and tumor necrosis factor α-stimulated expression of IL8 in canine gastric epithelial cells. LDN-193189 prevented BMP4-mediated inhibition of basal and tumor necrosis factor α-stimulated expression of IL8 in AGS cells. BMP-4 had no effect on TNFα-stimulated phosphorylation and degradation of IκBα, or on TNFα induction of a NFκß reporter gene. CONCLUSIONS: BMP signaling reduces inflammation and inhibits dysplastic changes in the gastric mucosa after infection of mice with H pylori or H felis.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Gastric Mucosa/metabolism , Gastritis/prevention & control , Signal Transduction , Animals , Binding Sites , Bone Morphogenetic Protein 4/deficiency , Bone Morphogenetic Protein 4/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Dogs , Female , Gastritis/genetics , Gastritis/immunology , Gastritis/metabolism , Gastritis/microbiology , Gastritis/pathology , Gene Expression Regulation , Genes, Reporter , Helicobacter felis/pathogenicity , Helicobacter pylori/pathogenicity , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic , RNA, Messenger/metabolism , Stomach/immunology , Stomach/microbiology , Stomach/pathology , Time Factors , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
8.
PLoS One ; 8(11): e79615, 2013.
Article in English | MEDLINE | ID: mdl-24260263

ABSTRACT

AIM: It has been reported that bone marrow-derived cells (BMDC) can be original cells of mouse gastric cancers induced by Helicobacter felis (H. felis) infection. However, it is unknown whether BMDCs are also the original cells of mouse gastrointestinal cancers induced by gastric carcinogens N-nitroso-N-methylurea (NMU) and H. felis infection. METHODS: C57BL/6 recipient mice were initially irradiated with 10Gy X-ray, reconstituted with bone marrow cells from the C57BL/6-Tg (CAG-EGFP) donor mice to label BMDCs with green fluorescence protein (GFP). After 4 weeks of recovery, the bone marrow-transplanted mice were given NMU in drinking water (240 ppm) and subsequently infected with H. felis by gavage. Eighty weeks later, all mice were euthanized for pathological examination. The BMDCs expressing GFP were detected in tissues using direct GFP fluorescence confocal microscopy analysis and immunohistochemistry staining (IHC) assays. RESULTS: Neoplastic lesions were induced by NMU treatment and/or H. felis infection at the antrum of the glandular stomach and small intestine. In the direct GFP fluorescence confocal assay, GFP(+) epithelial cell cluster or glands were not observed in these gastrointestinal tumors, however, most GFP(+) BMDCs sporadically located in the tumor stromal tissues. Some of these GFP(+) stromal BMDCs co-expressed the hematopoietic marker CD45 or myofibroblasts markers αSMA and SRF. In the indirect GFP IHC assay, similar results were observed among 11 gastric intraepithelial neoplasia lesions and 2 small intestine tumors. CONCLUSION: These results demonstrated that BMDCs might not be the source of gastrointestinal tumor cells induced by NMU and/or H. felis infection.


Subject(s)
Bone Marrow Cells/pathology , Carcinoma/chemically induced , Carcinoma/pathology , Gastrointestinal Neoplasms/chemically induced , Gastrointestinal Neoplasms/pathology , Animals , Carcinoma/microbiology , Cells, Cultured , Flow Cytometry , Gastrointestinal Neoplasms/microbiology , Helicobacter felis/pathogenicity , Immunohistochemistry , Mice , Mice, Inbred C57BL , Nitrosourea Compounds/toxicity
9.
PLoS One ; 7(11): e50194, 2012.
Article in English | MEDLINE | ID: mdl-23185574

ABSTRACT

There is increasing evidence from clinical and population studies for a role of H. pylori infection in the aetiology of iron deficiency. Rodent models of Helicobacter infection are helpful for investigating any causal links and mechanisms of iron deficiency in the host. The aim of this study was to investigate the effects of gastric Helicobacter infection on iron deficiency and host iron metabolism/transport gene expression in hypergastrinemic INS-GAS mice. INS-GAS mice were infected with Helicobacter felis for 3, 6 and 9 months. At post mortem, blood was taken for assessment of iron status and gastric mucosa for pathology, immunohistology and analysis of gene expression. Chronic Helicobacter infection of INS- GAS mice resulted in decreased serum iron, transferrin saturation and hypoferritinemia and increased Total iron binding capacity (TIBC). Decreased serum iron concentrations were associated with a concomitant reduction in the number of parietal cells, strengthening the association between hypochlorhydria and gastric Helicobacter-induced iron deficiency. Infection with H. felis for nine months was associated with decreased gastric expression of iron metabolism regulators hepcidin, Bmp4 and Bmp6 but increased expression of Ferroportin 1, the iron efflux protein, iron absorption genes such as Divalent metal transporter 1, Transferrin receptor 1 and also Lcn2 a siderophore-binding protein. The INS-GAS mouse is therefore a useful model for studying Helicobacter-induced iron deficiency. Furthermore, the marked changes in expression of gastric iron transporters following Helicobacter infection may be relevant to the more rapid development of carcinogenesis in the Helicobacter infected INS-GAS model.


Subject(s)
Anemia, Iron-Deficiency/metabolism , Helicobacter Infections/metabolism , Helicobacter felis/metabolism , Iron/metabolism , Parietal Cells, Gastric/metabolism , Acute-Phase Proteins/genetics , Acute-Phase Proteins/metabolism , Anemia, Iron-Deficiency/complications , Anemia, Iron-Deficiency/microbiology , Anemia, Iron-Deficiency/pathology , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Gastrins/genetics , Gene Expression Regulation , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter felis/pathogenicity , Hepcidins , Insulin/genetics , Lipocalin-2 , Lipocalins/genetics , Lipocalins/metabolism , Male , Mice , Mice, Transgenic , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Parietal Cells, Gastric/microbiology , Parietal Cells, Gastric/pathology , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Signal Transduction
10.
Gastroenterology ; 142(4): 824-833.e7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22248660

ABSTRACT

BACKGROUND & AIMS: Previous studies have suggested that dietary folic acid (FA) can protect against certain types of cancers. However, the findings have varied, and the mechanisms by which FA exerts chemopreventive effects remain to be clarified. We examined the effects of FA supplementation on DNA methylation, gene expression, and gastric dysplasia in a transgenic mouse model that is etiologically and histologically well matched with human gastric cancers. METHODS: Hypergastrinemic mice infected with Helicobacter felis were studied at multiple stages of gastric dysplasia and early cancer with FA supplementation initiated both at weaning and later in life. Global DNA methylation was assessed by a methylation sensitive cytosine incorporation assay, bisulfite pyrosequencing of B1 repetitive elements, and immunohistochemistry with anti-5-methylcytosine. We also profiled gene expression in the same tissues. RESULTS: We found a decrease in global DNA methylation and tissue folate and an increase in serum homocysteine with progression of gastric dysplasia. FA supplementation prevented this loss of global DNA methylation and markedly reduced gastric dysplasia and mucosal inflammation. FA protected against the loss of global DNA methylation both in the dysplastic gastric epithelial cells and in gastric stromal myofibroblasts. In addition, FA supplementation had an anti-inflammatory effect, as indicated by expression profiling and immunohistochemistry for lymphocyte markers. CONCLUSIONS: We conclude that FA supplementation is chemopreventive in this model of Helicobacter-associated gastric cancer. The beneficial effect of FA is likely due to its ability to prevent global loss of methylation and suppress inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Anticarcinogenic Agents/pharmacology , DNA Methylation/drug effects , Folic Acid/pharmacology , Gastritis/prevention & control , Helicobacter Infections/prevention & control , Helicobacter felis/pathogenicity , Stomach Neoplasms/prevention & control , Stomach/drug effects , Animals , Disease Models, Animal , Gastric Mucosa/metabolism , Gastrins/genetics , Gastrins/metabolism , Gastritis/blood , Gastritis/genetics , Gastritis/microbiology , Gastritis/pathology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Helicobacter Infections/blood , Helicobacter Infections/complications , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Homocysteine/blood , Immunohistochemistry , Lymphocytes/drug effects , Lymphocytes/microbiology , Lymphocytes/pathology , Male , Mice , Mice, Transgenic , Myofibroblasts/drug effects , Myofibroblasts/microbiology , Myofibroblasts/pathology , Stomach/microbiology , Stomach/pathology , Stomach Neoplasms/blood , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Stromal Cells/drug effects , Stromal Cells/microbiology , Stromal Cells/pathology , Up-Regulation
11.
Gut Microbes ; 2(6): 358-60, 2011.
Article in English | MEDLINE | ID: mdl-22198616

ABSTRACT

Development of the pathologies associated with Helicobacter pylori infection, most seriously gastric adenocarcinoma, are a consequence of chronic inflammation, which both host and pathogen go to some lengths to minimize. Recently, we presented evidence that H. pylori can suppress the development of inflammation in its immediate microenvironment in the gastric mucosa of 129/Sv mice. We have now extended this study by showing that H. felis, a gastric colonizing Helicobacter closely related to H. pylori, does not possess the same ability to suppress Helicobacter-induced gastritis in mice. Differences between these bacterial species may provide clues as to the mechanism behind the inflammation-regulating ability of H. pylori. Moreover, our demonstration that H. pylori but not H. felis can locally suppress inflammation in vivo may explain why H. felis infection induces superior levels of gastritis as compared with H. pylori infection of mice.


Subject(s)
Gastritis/microbiology , Helicobacter Infections/pathology , Helicobacter felis/growth & development , Helicobacter pylori/pathogenicity , Animals , DNA, Bacterial/genetics , Female , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis/pathology , Helicobacter Infections/microbiology , Helicobacter felis/genetics , Helicobacter felis/pathogenicity , Helicobacter pylori/genetics , Helicobacter pylori/growth & development , Inflammation/microbiology , Male , Mice , Mice, 129 Strain , Sex Factors , Species Specificity , Statistics, Nonparametric
12.
J Histochem Cytochem ; 59(9): 826-41, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21852692

ABSTRACT

Human Helicobacter pylori infection leads to multiple pathological consequences, including gastritis and adenocarcinoma. Although this association has led to the classification of H. pylori as a type 1 carcinogen, it is not clear if additional nonhelicobacter gastric microbiota play a role in these diseases. In this study, we utilized either specific pathogen-free C57BL/6 mice (B6.SPF) or mice colonized with altered Schaedler flora (B6.ASF) to evaluate the role of nonhelicobacter gastric microbiota in disease development after Helicobacter felis infection. Despite similar histological changes, H. felis persisted in B6.ASF stomachs, while H. felis could no longer be detected in the majority of B6.SPF mice. The B6.SPF mice also acquired multiple Lactobacillus spp. in their stomachs after H. felis infection. Our data indicate that potential mechanisms responsible for the ineffective H. felis clearance in the B6.ASF model include the absence of new gastric microbiota to compete for the gastric niche, the lack of expression of new gastric mucins, and a reduced ratio of H. felis-specific IgG2c:IgG1 serum antibodies. These data suggest that although H. felis is sufficient to initiate gastric inflammation and atrophy, bacterial eradication and the systemic immune response to infection are significantly influenced by pre-existing and acquired gastric microbiota.


Subject(s)
Helicobacter Infections/immunology , Helicobacter Infections/pathology , Helicobacter felis/immunology , Helicobacter felis/pathogenicity , Metagenome/physiology , Stomach Diseases/microbiology , Animals , Disease Progression , Female , Helicobacter felis/isolation & purification , Metagenome/immunology , Mice , Mice, Inbred C57BL , Stomach Diseases/immunology , Stomach Diseases/pathology
13.
Gastroenterology ; 140(3): 879-91, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21111741

ABSTRACT

BACKGROUND & AIMS: Epigenetic alterations have been correlated with field cancerization in human patients, but evidence from experimental models that specific epigenetic changes can initiate cancer has been lacking. Although hormones have been associated with cancer risk, the mechanisms have not been determined. The peptide hormone gastrin exerts a suppressive effect on antral gastric carcinogenesis. METHODS: N-methyl-N-nitrosourea (MNU)-dependent gastric cancer was investigated in hypergastrinemic (INS-GAS), gastrin-deficient (GAS(-/-)), Tff1-deficient (Tff1(+/-)), and wild-type (WT) mice. Epigenetic alterations of the trefoil factor 1 (TFF1) tumor suppressor gene were evaluated in vitro and in vivo. RESULTS: Human intestinal-type gastric cancers in the antrum exhibited progressive TFF1 repression and promoter hypermethylation. Mice treated with MNU exhibited a field defect characterized by widespread Tff1 repression associated with histone H3 lysine 9 methylation and H3 deacetylation at the Tff1 promoter in epithelial cells. In MNU-induced advanced cancers, DNA methylation at the Tff1 promoter was observed. Tumor induction and Tff1 repression were increased in MNU-treated mice by Helicobacter infection. Hypergastrinemia suppressed MNU-dependent tumor initiation and progression in a manner that correlated with gene silencing and epigenetic alterations of Tff1. In contrast, homozygous gastrin-deficient and heterozygous Tff1-deficient mice showed enhanced MNU-dependent field defects and cancer initiation compared with WT mice. In gastric cancer cells, gastrin stimulation partially reversed the epigenetic silencing in the TFF1 promoter. CONCLUSIONS: Initiation of antral gastric cancer is associated with progressive epigenetic silencing of TFF1, which can be suppressed by the hormone gastrin.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gastrins/metabolism , Gene Expression Regulation, Neoplastic , Gene Silencing , Peptides/genetics , Stomach Neoplasms/prevention & control , Tumor Suppressor Proteins/genetics , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chromatin Assembly and Disassembly , DNA Methylation , Disease Models, Animal , Female , Gastrins/deficiency , Gastrins/genetics , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter felis/pathogenicity , Histones/metabolism , Humans , Male , Methylnitrosourea , Mice , Mice, Knockout , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Peptides/deficiency , Promoter Regions, Genetic , RNA, Messenger/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Time Factors , Transfection , Trefoil Factor-1 , Tumor Suppressor Proteins/metabolism
14.
Gastroenterology ; 138(3): 1022-34.e1-10, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19962981

ABSTRACT

BACKGROUND & AIMS: The nuclear factor kappaB (NF-kappaB)/IkappaB-kinase-beta (IKKbeta) pathway has been shown to represent a key link between inflammation and cancer, inducing pro-inflammatory cytokines in myeloid cells and anti-apoptotic pathways in epithelial cells. However, the role of NF-kappaB pathway in gastric carcinogenesis and injury has not been well-defined. We derived mice with a conditional knockout of IKKbeta in gastric epithelial cells (GECs) and myeloid cells, and examined responses to ionizing radiation (IR) and Helicobacter felis infection. METHODS: Ikkbeta(Deltastom) mice were generated by crossing Foxa3-Cre mice to Ikkbeta(F/F) mice. Cellular stress was induced with IR and H felis in Ikkbeta(Deltastom), Ikkbeta(F/F), and cis-NF-kappaB-enhanced green fluorescent protein (GFP) reporter mice. Gastric histopathology, apoptosis, proliferation, necrosis, reactive oxygen species, and expression of cytokines, chemokines, and anti-apoptotic genes were assessed. The role of myeloid IKKbeta in these models was studied by crosses with LysM-Cre mice. RESULTS: NF-kappaB activity was upregulated in myeloid cells with acute H felis infection, but in GECs by IR or long-term H felis infection during progression to dysplasia. Deletion of IKKbeta in GECs led to increased apoptosis, reactive oxygen species, and cellular necrosis, and resulted in up-regulation of interleukin-1alpha and down-regulation of anti-apoptotic genes. Loss of IKKbeta in GECs resulted in worse inflammation and more rapid progression to gastric preneoplasia, while loss of IKKbeta in myeloid cells inhibited development of gastric atrophy. CONCLUSIONS: The loss of IKKbeta/NF-kappaB signaling in GECs results in increased apoptosis and necrosis in response to cellular stress, and accelerated development of dysplasia by Helicobacter infection.


Subject(s)
Apoptosis , Cell Proliferation , Gastric Mucosa/enzymology , Helicobacter Infections/enzymology , Helicobacter felis/pathogenicity , I-kappa B Kinase/deficiency , Precancerous Conditions/enzymology , Stomach Neoplasms/enzymology , Animals , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/metabolism , Atrophy , Cell Proliferation/radiation effects , Disease Models, Animal , Disease Progression , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastric Mucosa/radiation effects , Genes, Reporter , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Hepatocyte Nuclear Factor 3-gamma/genetics , I-kappa B Kinase/genetics , Inflammation Mediators/metabolism , Integrases/genetics , Interleukin-1alpha/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/enzymology , Myeloid Cells/microbiology , Myeloid Cells/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Necrosis , Oxidative Stress , Precancerous Conditions/genetics , Precancerous Conditions/microbiology , Precancerous Conditions/pathology , Reactive Oxygen Species/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Time Factors
15.
Cancer Cell ; 14(5): 408-19, 2008 Nov 04.
Article in English | MEDLINE | ID: mdl-18977329

ABSTRACT

Polymorphisms of interleukin-1beta (IL-1beta) are associated with an increased risk of solid malignancies. Here, we show that stomach-specific expression of human IL-1beta in transgenic mice leads to spontaneous gastric inflammation and cancer that correlate with early recruitment of myeloid-derived suppressor cells (MDSCs) to the stomach. IL-1beta activates MDSCs in vitro and in vivo through an IL-1RI/NF-kappaB pathway. IL-1beta transgenic mice deficient in T and B lymphocytes develop gastric dysplasia accompanied by a marked increase in MDSCs in the stomach. Antagonism of IL-1 receptor signaling inhibits the development of gastric preneoplasia and suppresses MDSC mobilization. These results demonstrate that pathologic elevation of a single proinflammatory cytokine may be sufficient to induce neoplasia and provide a direct link between IL-1beta, MDSCs, and carcinogenesis.


Subject(s)
Gastritis/etiology , Inflammation/etiology , Interleukin-1beta/metabolism , Mice, Transgenic/immunology , Myeloid Cells/immunology , Stomach Neoplasms/etiology , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow/pathology , Cells, Cultured , DNA-Binding Proteins/physiology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Gastritis/metabolism , Gastritis/pathology , H(+)-K(+)-Exchanging ATPase/physiology , Helicobacter Infections/immunology , Helicobacter Infections/metabolism , Helicobacter Infections/virology , Helicobacter felis/pathogenicity , Inflammation/metabolism , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-1beta/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic/metabolism , Mice, Transgenic/virology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Precancerous Conditions/immunology , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transfection
16.
Helicobacter ; 12(2): 112-23, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17309747

ABSTRACT

BACKGROUND: Surfactant protein D (SP-D), a component of innate immunity, is expressed in the gastric mucosa and is up-regulated in the presence of Helicobacter infection. SP-D binds to Helicobacter in vitro, suggesting the involvement of SP-D in Helicobacter-induced immune responses. The aim of this study was to determine the role of SP-D in gastric epithelial defense in vivo. METHODS: Specific pathogen-free SP-D-deficient mice (SP-D(-/-)) and C57BL/6 wild-type controls were challenged by gavage with different doses of Helicobacter felis, a mouse-adapted Helicobacter strain. Mice were assessed for colonization rates and density of infection. Inflammatory responses were measured by neutrophil counting and T-cell responses by proliferation assays on spleen cells stimulated with H. felis sonicate. The in vitro effect of SP-D on Helicobacter uptake by monocyte-derived dendritic cells was assessed by confocal microscopy and FACS analyses. RESULTS: SP-D(-/-) mice were more susceptible to low-dose infectious challenge than C57BL/6 controls (p = .02). The density of colonization was higher in the SP-D(-/-) infected mice. Neutrophil infiltrates were lower in the SP-D(-/-) mice, particularly in the acid-secreting regions of the stomach. T-cell proliferative responses to Helicobacter antigen were reduced in SP-D(-/-) mice (p = .001) after 12 weeks infection. In vitro uptake of Helicobacter by dendritic cells was significantly enhanced in the presence of SP-D (p = .001). CONCLUSION: In the absence of SP-D, Helicobacter uptake by dendritic cells is impaired. This provides an explanation for the diminished inflammation and immune responses in the SP-D(-/-) mice.


Subject(s)
Helicobacter Infections/metabolism , Helicobacter felis/pathogenicity , Pulmonary Surfactant-Associated Protein D/physiology , Agglutination , Animals , Cells, Cultured , Dendritic Cells/microbiology , Female , Gastritis/microbiology , Gastritis/pathology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter felis/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Pulmonary Surfactant-Associated Protein D/genetics , T-Lymphocytes/pathology
17.
J Clin Microbiol ; 44(5): 1692-6, 2006 May.
Article in English | MEDLINE | ID: mdl-16672395

ABSTRACT

Helicobacter pylori and Helicobacter felis are two of the Helicobacter spp. that infect humans. H. pylori has been linked to significant gastric pathology. Coinfection with Helicobacter spp. may influence infectious burden, pathogenesis, and antibiotic resistance; however, this has not been studied. The aims of this study were to identify the incidence of H. felis and to analyze the effects of coinfection with both organisms on gastric pathology in a well-characterized South African population. Biopsy samples from the gastric corpora and antra of volunteers (n = 90) were subjected to histological examination and PCR for the identification of H. pylori and H. felis. We further investigated the effect of global strain type on the occurrence of precursor lesions by assigning nucleotide sequences derived from PCR amplification of three genes to global groupings (ancestral Africa1, ancestral Africa2, ancestral Europe, ancestral Asia, and mixed). H. pylori was detected in 75 (83.3%), H. felis in 23 (25.6%), and coinfection in 21 (23.3%) of the volunteers by PCR. H. felis was randomly distributed among adults and children but clustered within families, suggesting intrafamilial transmission. Analysis of histopathology scores revealed no differences in atrophy, activity, and helicobacter density between H. felis-positive and H. felis-negative volunteers. H. pylori substrains common to southern Africa showed no differences in inflammation or atrophy scores. The incidences of H. felis and coinfection with H. pylori in the African population are high. H. felis infection, however, does not influence specific gastric pathology in this population.


Subject(s)
Gastric Mucosa/pathology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter felis , Helicobacter pylori , Adolescent , Adult , Bacterial Proteins/genetics , Base Sequence , Child , Child, Preschool , DNA, Bacterial/genetics , Female , Genes, Bacterial , Helicobacter Infections/epidemiology , Helicobacter Infections/transmission , Helicobacter felis/genetics , Helicobacter felis/isolation & purification , Helicobacter felis/pathogenicity , Helicobacter pylori/genetics , Helicobacter pylori/isolation & purification , Helicobacter pylori/pathogenicity , Humans , Male , Membrane Transport Proteins/genetics , Pedigree , South Africa/epidemiology
18.
Microbes Infect ; 8(2): 503-10, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16311055

ABSTRACT

Non-pylori helicobacter infections are associated with gastritis, gastric ulcers and MALT lymphomas in man. Approximately 50% of these are caused by helicobacters commonly found in dogs and cats, including Helicobacter felis, Helicobacter bizzozeronii and H. salomonis. In contrast to Helicobacter pylori, the virulence mechanisms of these species are unknown. In this study the virulence of H. felis, H. bizzozeronii and H. salomonis was investigated in Mongolian gerbils. Female SPF gerbils were inoculated intragastrically with H. felis, H. bizzozeronii or H. salomonis and sacrificed 3 weeks later. Fundus and antrum samples were taken for bacterial detection by PCR. A longitudinal strip covering all stomach regions was taken for histology. Gastric colonization, inflammation, apoptosis, loss of parietal cells and cell proliferation were assessed. Controls and H. salomonis inoculated gerbils were negative in PCR. H. felis and H. bizzozeronii inoculated animals were positive. H. felis inoculated animals showed loss of parietal cells extending from the limiting ridge into the fundus. A high cell proliferation rate was noticed in the mucosal area devoid of parietal cells. A dense band of apoptotic cells and large numbers of Helicobacter bacteria were seen at the transition zone between affected and normal parietal cells. In H. bizzozeronii infected gerbils, this was less pronounced. Focal apoptotic loss of gastric epithelial cells was spatially associated with the presence of bacteria especially in H. felis and to a lesser extent in H. bizzozeronii infected gerbils. This loss of cells may lead to intestinal metaplasia.


Subject(s)
Helicobacter Infections/pathology , Helicobacter felis/pathogenicity , Helicobacter/pathogenicity , Parietal Cells, Gastric/pathology , Animals , DNA, Bacterial/analysis , Female , Gerbillinae , Helicobacter/classification , Helicobacter/genetics , Helicobacter Infections/microbiology , Helicobacter Infections/physiopathology , Helicobacter felis/genetics , Helicobacter felis/isolation & purification , Parietal Cells, Gastric/microbiology , Polymerase Chain Reaction , Specific Pathogen-Free Organisms
19.
Helicobacter ; 10(6): 586-91, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16302984

ABSTRACT

BACKGROUND: We previously REPORTED that mice with diabetes and short-term Helicobacter felis infection had an increase in glycated hemoglobin (HbA1c). Here we report the effect of long-term infection. MATERIALS AND METHODS: Six-week-old C57BL/6 mice were injected with streptozotocin to induce diabetes and started on daily insulin. Following streptozotocin injection, animals were paired according to their HbA1c values and randomized to orally receive either H. felis or culture medium alone. Weight and HbA1c were monitored monthly for 6 months. RESULTS: Thirty animals corresponding to 15 pairs were included in the study. H. felis-infected diabetic mice developed significantly more gastritis than uninfected animals. Sixteen mice died during the observation period. As compared to uninfected animals, infected mice died more frequently (40% versus 67%, p = .14) and earlier (160 versus 61 days, p = .20); both variables combined showed that H. felis infection significantly decreased the chances of survival during the study period (p = .045). In addition, infected mice showed a trend for higher increase in their HbA1c (0.97 +/- 2.5% versus - 0.22 +/- 3.0%; p = .21) and lower weight gain (2.0 +/- 3.4 g versus 2.9 +/- 2.0 g; p = .15) than uninfected mice. CONCLUSION: Long-term H. felis infection had a deleterious effect in mice with streptozotocin-induced diabetes resulting in increased mortality. If the same phenomenon occurs in humans this could lead to interventions to improve the long-term outcome of patients with diabetes.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/mortality , Disease Models, Animal , Helicobacter Infections/complications , Helicobacter Infections/pathology , Helicobacter felis/pathogenicity , Animals , Cats , Female , Gastritis/complications , Gastritis/microbiology , Gastritis/mortality , Gastritis/pathology , Glycated Hemoglobin/metabolism , Helicobacter Infections/mortality , Mice , Mice, Inbred C57BL , Random Allocation , Time Factors
20.
J Comp Pathol ; 133(2-3): 83-91, 2005.
Article in English | MEDLINE | ID: mdl-15949811

ABSTRACT

The inflammatory response in the mouse stomach was evaluated as a means of distinguishing different non-pylori Helicobacter (H.) strains in terms of virulence. Mice of four strains (BALB/c, SJL, C57BL/6 and CFW) were infected intragastrically with four bacterial strains (H. felis ATCC 49179 and CCUG 37471, H. bizzozeronii and H. salomonis). The animals were killed for gastric examination at 3, 9 or 16 weeks post-inoculation. H. salomonis could not be detected by the polymerase chain reaction, but the other three organisms were detected in all stomach samples at all timepoints. SJL mice consistently showed particularly severe gastric inflammation regardless of bacterial strain. Lymphocytes and occasionally neutrophils were seen in submucosa and lamina propria mucosae. BALB/c mice showed the least severe inflammatory changes. H. bizzozeronii differed from the two H. felis strains in producing less striking pathological changes in mice. Of the two H. felis strains, ATCC 49179 produced the more severe inflammatory changes in SJL mice.


Subject(s)
Gastritis/pathology , Helicobacter Infections/pathology , Helicobacter felis/pathogenicity , Stomach/pathology , Animals , DNA, Bacterial/analysis , Disease Models, Animal , Gastritis/microbiology , Helicobacter Infections/microbiology , Helicobacter felis/classification , Helicobacter felis/genetics , Male , Mice , Mice, Inbred Strains , Polymerase Chain Reaction , Species Specificity , Specific Pathogen-Free Organisms , Stomach/microbiology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...