Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Oncogene ; 36(27): 3807-3819, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28263971

ABSTRACT

Epidermal growth factor receptor (EGFR) signaling is a known mediator of colorectal carcinogenesis. Studies have focused on the role of EGFR signaling in epithelial cells, although the exact nature of the role of EGFR in colorectal carcinogenesis remains a topic of debate. Here, we present evidence that EGFR signaling in myeloid cells, specifically macrophages, is critical for colon tumorigenesis in the azoxymethane-dextran sodium sulfate (AOM-DSS) model of colitis-associated carcinogenesis (CAC). In a human tissue microarray, colonic macrophages demonstrated robust EGFR activation in the pre-cancerous stages of colitis and dysplasia. Utilizing the AOM-DSS model, mice with a myeloid-specific deletion of Egfr had significantly decreased tumor multiplicity and burden, protection from high-grade dysplasia and significantly reduced colitis. Intriguingly, mice with gastrointestinal epithelial cell-specific Egfr deletion demonstrated no differences in tumorigenesis in the AOM-DSS model. The alterations in tumorigenesis in myeloid-specific Egfr knockout mice were accompanied by decreased macrophage, neutrophil and T-cell infiltration. Pro-tumorigenic M2 macrophage activation was diminished in myeloid-specific Egfr-deficient mice, as marked by decreased Arg1 and Il10 mRNA expression and decreased interleukin (IL)-4, IL10 and IL-13 protein levels. Surprisingly, diminished M1 macrophage activation was also detectable, as marked by significantly reduced Nos2 and Il1b mRNA levels and decreased interferon (IFN)-γ, tumor necrosis factor (TNF)-α and IL-1ß protein levels. The alterations in M1 and M2 macrophage activation were confirmed in bone marrow-derived macrophages from mice with the myeloid-specific Egfr knockout. The combined effect of restrained M1 and M2 macrophage activation resulted in decreased production of pro-angiogenic factors, CXCL1 and vascular endothelial growth factor (VEGF), and reduced CD31+ blood vessels, which likely contributed to protection from tumorigenesis. These data reveal that EGFR signaling in macrophages, but not in colonic epithelial cells, has a significant role in CAC. EGFR signaling in macrophages may prove to be an effective biomarker of CAC or target for chemoprevention in patients with inflammatory bowel disease.


Subject(s)
Carcinogenesis/metabolism , Colitis/pathology , Colonic Neoplasms/metabolism , ErbB Receptors/physiology , Macrophage Activation , Precancerous Conditions/metabolism , Animals , Carcinogenesis/immunology , Colon/immunology , Colon/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/immunology , Dextran Sulfate , Humans , Immunity, Innate , Macrophages/physiology , Male , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Precancerous Conditions/immunology , Signal Transduction
2.
Mucosal Immunol ; 10(2): 493-507, 2017 03.
Article in English | MEDLINE | ID: mdl-27435104

ABSTRACT

Calcium signaling in phagocytes is essential for cellular activation, migration, and the potential resolution of infection or inflammation. The generation of reactive oxygen species (ROS) via activation of NADPH (nicotinamide adenine dinucleotide phosphate)-oxidase activity in macrophages has been linked to altered intracellular calcium concentrations. Because of its role as an oxidative stress sensor in phagocytes, we investigated the function of the cation channel transient receptor potential melastatin 2 (TRPM2) in macrophages during oxidative stress responses induced by Helicobacter pylori infection. We show that Trpm2-/- mice, when chronically infected with H. pylori, exhibit increased gastric inflammation and decreased bacterial colonization compared with wild-type (WT) mice. The absence of TRPM2 triggers greater macrophage production of inflammatory mediators and promotes classically activated macrophage M1 polarization in response to H. pylori. TRPM2-deficient macrophages upon H. pylori stimulation are unable to control intracellular calcium levels, which results in calcium overloading. Furthermore, increased intracellular calcium in TRPM2-/- macrophages enhanced mitogen-activated protein kinase and NADPH-oxidase activities, compared with WT macrophages. Our data suggest that augmented production of ROS and inflammatory cytokines with TRPM2 deletion regulates oxidative stress in macrophages and consequently decreases H. pylori gastric colonization while increasing inflammation in the gastric mucosa.


Subject(s)
Gastritis/genetics , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Macrophages/immunology , TRPM Cation Channels/metabolism , Animals , Calcium Signaling , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Gastritis/immunology , Helicobacter Infections/genetics , Inflammation Mediators/metabolism , Macrophage Activation/genetics , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NADP/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , TRPM Cation Channels/genetics
3.
Oncogene ; 35(48): 6262-6269, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27157617

ABSTRACT

Helicobacter pylori (H. pylori) is the strongest identified risk factor for gastric cancer, the third most common cause of cancer-related death worldwide. An H. pylori constituent that augments cancer risk is the strain-specific cag pathogenicity island, which encodes a type IV secretion system (T4SS) that translocates a pro-inflammatory and oncogenic protein, CagA, into epithelial cells. However, the majority of persons colonized with CagA+ H. pylori strains do not develop cancer, suggesting that other microbial effectors also have a role in carcinogenesis. Toll-like receptor 9 (TLR9) is an endosome bound, innate immune receptor that detects and responds to hypo-methylated CpG DNA motifs that are most commonly found in microbial genomes. High-expression tlr9 polymorphisms have been linked to the development of premalignant lesions in the stomach. We now demonstrate that levels of H. pylori-mediated TLR9 activation and expression are directly related to gastric cancer risk in human populations. Mechanistically, we show for the first time that the H. pylori cancer-associated cag T4SS is required for TLR9 activation and that H. pylori DNA is actively translocated by the cag T4SS to engage this host receptor. Activation of TLR9 occurs through a contact-dependent mechanism between pathogen and host, and involves transfer of microbial DNA that is both protected as well as exposed during transport. These results indicate that TLR9 activation via the cag island may modify the risk for malignancy within the context of H. pylori infection and provide an important framework for future studies investigating the microbial-epithelial interface in gastric carcinogenesis.


Subject(s)
Bacterial Proteins/metabolism , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/physiology , Toll-Like Receptor 9/metabolism , Type IV Secretion Systems , Bacterial Proteins/genetics , Biological Transport , Carcinogenesis , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Helicobacter Infections/complications , Humans , Mutation , Stomach Neoplasms/etiology
4.
Oncogene ; 35(42): 5480-5488, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27041578

ABSTRACT

Chronic inflammation contributes to the development of various forms of cancer. The polyamine catabolic enzyme spermine oxidase (SMOX) is induced in chronic inflammatory conditions, including Helicobacter pylori-associated gastritis, where its production of hydrogen peroxide contributes to DNA damage and subsequent tumorigenesis. MicroRNA expression levels are also altered in inflammatory conditions; specifically, the tumor suppressor miR-124 becomes silenced by DNA methylation. We sought to determine if this repression of miR-124 is associated with elevated SMOX activity and concluded that miR-124 is indeed a negative regulator of SMOX. In gastric adenocarcinoma cells harboring highly methylated and silenced mir-124 gene loci, 5-azacytidine treatment allowed miR-124 re-expression and decreased SMOX expression. Overexpression of an exogenous miR-124-3p mimic repressed SMOX mRNA and protein expression as well as H2O2 production by >50% within 24 h. Reporter assays indicated that direct interaction of miR-124 with the 3'-untranslated region of SMOX mRNA contributes to this negative regulation. Importantly, overexpression of miR-124 before infection with H. pylori prevented the induction of SMOX believed to contribute to inflammation-associated tumorigenesis. Compelling human in vivo data from H. pylori-positive gastritis tissues indicated that the mir-124 gene loci are more heavily methylated in a Colombian population characterized by elevated SMOX expression and a high risk for gastric cancer. Furthermore, the degree of mir-124 methylation significantly correlated with SMOX expression throughout the population. These results indicate a protective role for miR-124 through the inhibition of SMOX-mediated DNA damage in the etiology of H. pylori-associated gastric cancer.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Gene Silencing , MicroRNAs/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , 3' Untranslated Regions , Biopsy , DNA Methylation , Down-Regulation , Gastritis/etiology , Gastritis/pathology , Helicobacter Infections/complications , Helicobacter pylori , Humans , Stomach Neoplasms/etiology , Stomach Neoplasms/pathology , Polyamine Oxidase
5.
Mucosal Immunol ; 8(5): 1047-59, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25603827

ABSTRACT

Lymphoid tissue inducer (LTi) cells are activated by accessory cell IL-23, and promote lymphoid tissue genesis and antibacterial peptide production by the mucosal epithelium. We investigated the role of LTi cells in the gastric mucosa in the context of microbial infection. Mice deficient in IRAK-M, a negative regulator of TLR signaling, were investigated for increased LTi cell activity, and antibody mediated LTi cell depletion was used to analyze LTi cell dependent antimicrobial activity. H. pylori infected IRAK-M deficient mice developed increased gastric IL-17 and lymphoid follicles compared to wild type mice. LTi cells were present in naive and infected mice, with increased numbers in IRAK-M deficient mice by two weeks. Helicobacter and Candida infection of LTi cell depleted rag1(-/-) mice demonstrated LTi-dependent increases in calprotectin but not RegIII proteins. However, pathogen and commensal microbiota populations remained unchanged in the presence or absence of LTi cell function. These data demonstrate LTi cells are present in the stomach and promote lymphoid follicle formation in response to infection, but are limited by IRAK-M expression. Additionally, LTi cell mediated antimicrobial peptide production at the gastric epithelium is less efficacious at protecting against microbial pathogens than has been reported for other tissues.


Subject(s)
Candida/immunology , Candidiasis/immunology , Gastric Mucosa/immunology , Helicobacter Infections/immunology , Helicobacter/immunology , Interleukin-1 Receptor-Associated Kinases/immunology , Lymphoid Tissue/immunology , Animals , Candidiasis/genetics , Candidiasis/pathology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Helicobacter Infections/genetics , Helicobacter Infections/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-23/genetics , Interleukin-23/immunology , Lymphoid Tissue/pathology , Mice , Mice, Knockout
6.
Oncogene ; 34(14): 1865-71, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-24837365

ABSTRACT

Helicobacter pylori is the strongest risk factor for the development of gastric cancer. Although the specific mechanisms by which this pathogen induces carcinogenesis have not been fully elucidated, high-expression interleukin (IL)-1ß alleles are associated with increased gastric cancer risk among H. pylori-infected persons. In addition, loss of matrix metalloproteinase 7 (MMP7) increases mucosal inflammation in mouse models of epithelial injury, and we have shown that gastric inflammation is increased in H. pylori-infected MMP7(-/-) C57BL/6 mice. In this report, we define mechanisms that underpin such responses and extend these results into a genetic model of MMP7 deficiency and gastric cancer. Wild-type (WT) or MMP7(-/-) C57BL/6 mice were challenged with broth alone as an uninfected control or the H. pylori strain PMSS1. All H. pylori-challenged mice were successfully colonized. As expected, H. pylori-infected MMP7(-/-) C57BL/6 mice exhibited a significant increase in gastric inflammation compared with uninfected or infected WT C57BL/6 animals. Loss of MMP7 resulted in M1 macrophage polarization within H. pylori-infected stomachs, as assessed by Luminex technology and immunohistochemistry, and macrophages isolated from infected MMP7-deficient mice expressed significantly higher levels of the M1 macrophage marker IL-1ß compared with macrophages isolated from WT mice. To extend these findings into a model of gastric cancer, hypergastrinemic WT INS-GAS or MMP7(-/-) INS-GAS mice were challenged with H. pylori strain PMSS1. Consistent with findings in the C57BL/6 model, H. pylori-infected MMP7-deficient INS-GAS mice exhibited a significant increase in gastric inflammation compared with either uninfected or infected WT INS-GAS mice. In addition, the incidence of gastric hyperplasia and dysplasia was significantly increased in H. pylori-infected MMP7(-/-) INS-GAS mice compared with infected WT INS-GAS mice, and loss of MMP7 promoted M1 macrophage polarization. These results suggest that MMP7 exerts a restrictive role on H. pylori-induced gastric injury and the development of premalignant lesions by suppressing M1 macrophage polarization.


Subject(s)
Gastritis/microbiology , Helicobacter Infections/pathology , Macrophages/immunology , Matrix Metalloproteinase 7/genetics , Stomach , Animals , Disease Models, Animal , Gastric Mucosa/cytology , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastritis/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Helicobacter pylori/pathogenicity , Hyperplasia/genetics , Inflammation/immunology , Inflammation/microbiology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Precancerous Conditions/immunology , Precancerous Conditions/microbiology , Stomach/immunology , Stomach/microbiology , Stomach/pathology , Stomach Neoplasms/immunology , Stomach Neoplasms/microbiology
7.
Oncogene ; 34(26): 3429-40, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25174398

ABSTRACT

Helicobacter pylori infection causes gastric cancer, the third leading cause of cancer death worldwide. More than half of the world's population is infected, making universal eradication impractical. Clinical trials suggest that antibiotic treatment only reduces gastric cancer risk in patients with non-atrophic gastritis (NAG), and is ineffective once preneoplastic lesions of multifocal atrophic gastritis (MAG) and intestinal metaplasia (IM) have occurred. Therefore, additional strategies for risk stratification and chemoprevention of gastric cancer are needed. We have implicated polyamines, generated by the rate-limiting enzyme ornithine decarboxylase (ODC), in gastric carcinogenesis. During H. pylori infection, the enzyme spermine oxidase (SMOX) is induced, which generates hydrogen peroxide from the catabolism of the polyamine spermine. Herein, we assessed the role of SMOX in the increased gastric cancer risk in Colombia associated with the Andean mountain region when compared with the low-risk region on the Pacific coast. When cocultured with gastric epithelial cells, clinical strains of H. pylori from the high-risk region induced more SMOX expression and oxidative DNA damage, and less apoptosis than low-risk strains. These findings were not attributable to differences in the cytotoxin-associated gene A oncoprotein. Gastric tissues from subjects from the high-risk region exhibited greater levels of SMOX and oxidative DNA damage by immunohistochemistry and flow cytometry, and this occurred in NAG, MAG and IM. In Mongolian gerbils, a prototype colonizing strain from the high-risk region induced more SMOX, DNA damage, dysplasia and adenocarcinoma than a colonizing strain from the low-risk region. Treatment of gerbils with either α-difluoromethylornithine, an inhibitor of ODC, or MDL 72527 (N(1),N(4)-Di(buta-2,3-dien-1-yl)butane-1,4-diamine dihydrochloride), an inhibitor of SMOX, reduced gastric dysplasia and carcinoma, as well as apoptosis-resistant cells with DNA damage. These data indicate that aberrant activation of polyamine-driven oxidative stress is a marker of gastric cancer risk and a target for chemoprevention.


Subject(s)
Adenocarcinoma , Helicobacter Infections/complications , Helicobacter pylori/physiology , Oxidoreductases Acting on CH-NH Group Donors/physiology , Stomach Neoplasms , Adenocarcinoma/epidemiology , Adenocarcinoma/genetics , Adenocarcinoma/microbiology , Adult , Animals , Cells, Cultured , Colombia/epidemiology , DNA Damage/genetics , Enzyme Induction , Gerbillinae , Helicobacter Infections/genetics , Humans , Hydrogen Peroxide/metabolism , Male , Middle Aged , Oxidative Stress/genetics , Risk Factors , Stomach Neoplasms/epidemiology , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology , Polyamine Oxidase
8.
Clin Microbiol Infect ; 16(4): 369-78, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19456839

ABSTRACT

The C-terminus of the Helicobacter pylori CagA protein is polymorphic, bearing different EPIYA sequences (EPIYA-A, B, C or D), and one or more CagA multimerization (CM) motifs. The number of EPIYA-C motifs is associated with precancerous lesions and gastric cancer (GC). The relationship between EPIYA, CM motifs and gastric lesions was examined in H. pylori-infected Colombian patients from areas of high and low risk for GC. Genomic DNA was extracted from H. pylori strains cultured from gastric biopsies from 80 adults with dyspeptic symptoms. Sixty-seven (83.8%) of 80 strains were cagA positive. The 3' region of cagA was sequenced, and EPIYA and CM motifs were identified. CagA proteins contained one (64.2%), two (34.3%) or three EPIYA-C motifs (1.5%), all with Western type CagA-specific sequences. Strains with one EPIYA-C motif were associated with less severe gastric lesions (non-atrophic and multifocal atrophic gastritis), whereas strains with multiple EPIYA-C motifs were associated with more severe lesions (intestinal metaplasia and dysplasia) (p <0.001). In 54 strains, the CM motifs were identical to those common in Western strains. Thirteen strains from the low-risk area contained two different CM motifs: one of Western type located within the EPIYA-C segment and another following the EPIYA-C segment and resembling the CM motif found in East Asian strains. These strains induced significantly shorter projections in AGS cells and an attenuated reduction in levels of CagA upon immunodepletion of SHP-2 than strains possessing Western/Western motifs. This novel finding may partially explain the difference in GC incidence in these populations.


Subject(s)
Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Helicobacter Infections/complications , Helicobacter pylori/genetics , Precancerous Conditions/microbiology , Stomach Neoplasms/microbiology , Adult , Amino Acid Sequence , Antigens, Bacterial/chemistry , Antigens, Bacterial/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Colombia , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Helicobacter pylori/isolation & purification , Helicobacter pylori/metabolism , Humans , Male , Middle Aged , Molecular Sequence Data , Polymorphism, Genetic , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
9.
Infect Immun ; 76(9): 3992-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18519558

ABSTRACT

Chronic infection with the gastric pathogen Helicobacter pylori significantly increases the risk of developing atrophic gastritis, peptic ulcer disease, and gastric adenocarcinoma. H. pylori strains that possess the cag pathogenicity island, which translocates CagA into the host cells, augment these risks. The aim of this study was to determine the molecular mechanisms through which H. pylori upregulates the expression of plasminogen activator inhibitor 1 (PAI-1), a member of the urokinase activator system that is involved in tumor metastasis and angiogenesis. Levels of PAI-1 mRNA and protein were examined in tissues from H. pylori-infected patients and in vitro using AGS gastric epithelial cells. In vitro, cells were infected with toxigenic cag-positive or nontoxigenic cag-negative strains of H. pylori or isogenic mutants. The amount of PAI-1 secretion was measured by enzyme-linked immunosorbent assay, and mRNA levels were determined using real-time PCR. The regulation of PAI-1 was examined using the extracellular signal-regulated kinase 1/2 (ERK1/2) inhibitor and small interfering RNA. Analysis of human biopsy samples revealed an increase in both PAI-1 mRNA and protein levels in patients with H. pylori gastritis compared to those of uninfected controls. Infection of AGS cells with H. pylori significantly increased PAI-1 mRNA expression and the secretion of PAI-1 protein. Moreover, PAI-1 mRNA and protein production was more pronounced when AGS cells were infected by H. pylori strains carrying a functional cag secretion system than when cells were infected by strains lacking this system. PAI-1 secretion was also reduced when cells were infected with either cagE-negative or cagA-negative mutants. The ectopic overexpression of CagA significantly increased the levels of PAI-1 mRNA and protein, whereas blockade of the ERK1/2 pathway inhibited H. pylori-mediated PAI-1 upregulation. These findings suggest that the upregulation of PAI-1 in H. pylori-infected gastric epithelial cells may contribute to the carcinogenic process.


Subject(s)
Epithelial Cells/microbiology , Gastric Mucosa/microbiology , Helicobacter pylori/physiology , Plasminogen Activator Inhibitor 1/biosynthesis , Up-Regulation , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Cell Line , Enzyme-Linked Immunosorbent Assay , Female , Gastric Mucosa/pathology , Gene Deletion , Gene Expression Profiling , Humans , Male , Middle Aged
10.
Dig Liver Dis ; 40(7): 490-6, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18396115

ABSTRACT

This report describes the modalities of chronic gastritis induced by Helicobacter pylori infection in different populations. The full gamut of lesions representing the precancerous cascade is very prevalent in populations of low socioeconomic background experiencing very high gastric cancer risk, as seen in the Latin American Andes Mountains. In populations of high socioeconomic standards and high cancer risk, such as Japan and Korea, the precancerous cascade predominates and "early" cancers are also diagnosed frequently. Some reports describe frequent corpus atrophy, not prominent in the former group. The so-called African enigma is seen in populations of low socioeconomic standards, usually living at low altitudes, with high prevalence of infection but low frequency of cancer and precancerous lesions. In populations in transition from high to low cancer risk, duodenal ulcer and antral non-atrophic gastritis are frequently seen. In affluent societies at low risk of cancer, such as Western Europe, Australia and North America, mild non-atrophic gastritis associated with low virulence Helicobacter pylori genotypes predominate. The varied phenotypes of gastritis may reflect secular changes in the ecology of our species.


Subject(s)
Gastritis, Atrophic/microbiology , Helicobacter Infections/epidemiology , Helicobacter pylori , Precancerous Conditions/epidemiology , Precancerous Conditions/microbiology , Stomach Neoplasms/epidemiology , Stomach Neoplasms/microbiology , Chronic Disease , Europe/epidemiology , Gastritis, Atrophic/epidemiology , Gastritis, Atrophic/pathology , Helicobacter pylori/pathogenicity , Humans , Italy/epidemiology , Japan/epidemiology , Korea/epidemiology , Latin America/epidemiology , North America/epidemiology , Precancerous Conditions/pathology , Prevalence , Risk Factors , Socioeconomic Factors , Stomach Neoplasms/pathology
11.
Oncogene ; 27(15): 2170-6, 2008 Apr 03.
Article in English | MEDLINE | ID: mdl-17952118

ABSTRACT

The p73 protein is a transcription factor and member of the p53 protein family that expresses as a complex variety of isoforms. DeltaNp73alpha is an N-terminally truncated isoform of p73. We found that DeltaNp73 protein is upregulated in human gastric carcinoma suggesting that DeltaNp73 may play an oncogenic role in these tumors. Although it has been shown that DeltaNp73alpha inhibits apoptosis and counteracts the effect of chemotherapeutic drugs, the underlying mechanism by which this p73 isoform contributes to chemotherapeutic drug response remains to be explored. We found that DeltaNp73alpha upregulates MDR1 mRNA and p-glycoprotein (p-gp), which is involved in chemotherapeutic drug transport. This p-gp upregulation was accompanied by increased p-gp functional activity in gastric cancer cells. Our data suggest that upregulation of MDR1 by DeltaNp73alpha is mediated by interaction with p53 at the MDR1 promoter.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation, Neoplastic , Genes, p53/physiology , Nuclear Proteins/physiology , Tumor Suppressor Proteins/physiology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Carcinoma/genetics , Carcinoma/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , Humans , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
13.
Gut ; 54(11): 1536-40, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15985559

ABSTRACT

BACKGROUND: Helicobacter pylori infection induces progressive inflammatory changes in the gastric mucosa that may lead to gastric cancer. Understanding long term effects resulting from the cure of this infection is needed to design cancer prevention strategies. METHODS: A cohort of 795 adults with preneoplastic gastric lesions was randomised to receive anti-H pylori treatment and/or antioxidants. At the end of six years of intervention, those who did not receive anti-H pylori treatment were offered it. Gastric biopsies were obtained at baseline, and at 3, 6, and 12 years. A histopathology score was utilised to document changes in gastric lesions. Non-linear mixed models were used to estimate the cumulative effect of H pylori clearance on histopathology scores adjusted for follow up time, interventions, and confounders. RESULTS: Ninety seven per cent of subjects were H pylori positive at baseline, and 53% were positive at 12 years. Subjects accumulated 1703 person years free of infection. A multivariate model showed a significant regression in histopathology score as a function of the square of H pylori negative time. Subjects who were H pylori negative had 14.8% more regression and 13.7% less progression than patients who were positive at 12 years (p = 0.001). The rate of healing of gastric lesions occurred more rapidly as years free of infection accumulated, and was more pronounced in less advanced lesions. CONCLUSIONS: Preneoplastic gastric lesions regress at a rate equal to the square of time in patients rendered free of H pylori infection. Our findings suggest that patients with preneoplastic gastric lesions should be treated and cured of their H pylori infection.


Subject(s)
Helicobacter Infections/drug therapy , Helicobacter pylori , Precancerous Conditions/microbiology , Stomach Neoplasms/microbiology , Acute Disease , Adult , Aged , Anti-Bacterial Agents , Antioxidants/therapeutic use , Chronic Disease , Disease Progression , Drug Therapy, Combination/therapeutic use , Female , Follow-Up Studies , Gastritis/microbiology , Gastritis/pathology , Helicobacter Infections/complications , Humans , Male , Middle Aged , Precancerous Conditions/pathology , Recurrence , Stomach Neoplasms/pathology , Stomach Neoplasms/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...