Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
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(49): 6341-6349, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27270437

ABSTRACT

Myeloid translocation genes (MTGs), originally identified as chromosomal translocations in acute myelogenous leukemia, are transcriptional corepressors that regulate hematopoietic stem cell programs. Analysis of The Cancer Genome Atlas (TCGA) database revealed that MTGs were mutated in epithelial malignancy and suggested that loss of function might promote tumorigenesis. Genetic deletion of MTGR1 and MTG16 in the mouse has revealed unexpected and unique roles within the intestinal epithelium. Mtgr1-/- mice have progressive depletion of all intestinal secretory cells, and Mtg16-/- mice have a decrease in goblet cells. Furthermore, both Mtgr1-/- and Mtg16-/- mice have increased intestinal epithelial cell proliferation. We thus hypothesized that loss of MTGR1 or MTG16 would modify Apc1638/+-dependent intestinal tumorigenesis. Mtgr1-/- mice, but not Mtg16-/- mice, had a 10-fold increase in tumor multiplicity. This was associated with more advanced dysplasia, including progression to invasive adenocarcinoma, and augmented intratumoral proliferation. Analysis of chromatin immunoprecipitation sequencing data sets for MTGR1 and MTG16 targets indicated that MTGR1 can regulate Wnt and Notch signaling. In support of this, immunohistochemistry and gene expression analysis revealed that both Wnt and Notch signaling pathways were hyperactive in Mtgr1-/- tumors. Furthermore, in human colorectal cancer (CRC) samples MTGR1 was downregulated at both the transcript and protein level. Overall our data indicates that MTGR1 has a context-dependent effect on intestinal tumorigenesis.


Subject(s)
Colorectal Neoplasms/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Animals , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Translocation, Genetic , Tumor Suppressor Proteins/metabolism
4.
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
5.
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
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.
Mucosal Immunol ; 7(6): 1340-53, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24670427

ABSTRACT

Expression of claudin-2, a tight junction protein, is highly upregulated during inflammatory bowel disease (IBD) and, due to its association with epithelial permeability, has been postulated to promote inflammation. Notably, claudin-2 has also been implicated in the regulation of intestinal epithelial proliferation. However, precise role of claudin-2 in regulating colonic homeostasis remains unclear. Here, we demonstrate, using Villin-Claudin-2 transgenic mice, that increased colonic claudin-2 expression augments mucosal permeability as well as colon and crypt length. Most notably, despite leaky colon, Cl-2TG mice were significantly protected against experimental colitis. Importantly, claudin-2 expression increased colonocyte proliferation and provided protection against colitis-induced colonocyte death in a PI-3Kinase/Bcl-2-dependent manner. However, Cl-2TG mice also demonstrated marked suppression of colitis-induced increases in immune activation and associated signaling, suggesting immune tolerance. Accordingly, colons from naive Cl-2TG mice harbored significantly increased numbers of regulatory (CD4(+)Foxp3(+)) T cells than WT littermates. Furthermore, macrophages isolated from Cl-2TG mouse colon exhibited immune anergy. Importantly, these immunosuppressive changes were associated with increased synthesis of the immunoregulatory cytokine TGF-ß by colonic epithelial cells in Cl-2TG mice compared with WT littermates. Taken together, our findings reveal a critical albeit complex role of claudin-2 in intestinal homeostasis by regulating epithelial permeability, inflammation and proliferation and suggest novel therapeutic opportunities.


Subject(s)
Claudins/immunology , Colitis/immunology , Gene Expression Regulation , Immunity, Innate , Immunity, Mucosal , Intestinal Mucosa/immunology , Animals , Caco-2 Cells , Claudins/genetics , Colitis/genetics , Colitis/pathology , Humans , Intestinal Mucosa/injuries , Intestinal Mucosa/pathology , Mice , Mice, Transgenic , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
9.
Oncogene ; 30(29): 3234-47, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21383692

ABSTRACT

Claudin-2 is a unique member of the claudin family of transmembrane proteins, as its expression is restricted to the leaky epithelium in vivo and correlates with epithelial leakiness in vitro. However, recent evidence suggests potential functions of claudin-2 that are relevant to neoplastic transformation and growth. In accordance, here we report, on the basis of analysis of mRNA and protein expression using a total of 309 patient samples that claudin-2 expression is significantly increased in colorectal cancer and correlates with cancer progression. We also report similar increases in claudin-2 expression in inflammatory bowel disease-associated colorectal cancer. Most importantly, we demonstrate that the increased claudin-2 expression in colorectal cancer is causally associated with tumor growth as forced claudin-2 expression in colon cancer cells that do not express claudin-2 resulted in significant increases in cell proliferation, anchorage-independent growth and tumor growth in vivo. We further show that the colonic microenvironment regulates claudin-2 expression in a manner dependent on signaling through the EGF receptor (EGFR), a key regulator of colon tumorigenesis. In addition, claudin-2 expression is specifically decreased in the colon of waved-2 mice, naturally deficient in EGFR activation. Furthermore, genetic silencing of claudin-2 expression in Caco-2, a colon cancer cell line, prevents the EGF-induced increase in cell proliferation. Taken together, these results uncover a novel role for claudin-2 in promoting colon cancer, potentially via EGFR transactivation.


Subject(s)
Colonic Neoplasms/genetics , ErbB Receptors/genetics , Membrane Proteins/metabolism , Transcriptional Activation , Animals , Caco-2 Cells , Cell Division/genetics , Claudins , Colonic Neoplasms/etiology , Colonic Neoplasms/pathology , Epidermal Growth Factor/pharmacology , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Membrane Proteins/genetics , Mice , Mice, Nude , Protein Kinases/metabolism , RNA, Messenger/genetics , Up-Regulation
10.
Aliment Pharmacol Ther ; 21(10): 1211-6, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15882241

ABSTRACT

BACKGROUND: Polypharmacy has not been defined for Crohn's disease. AIMS: To determine the prevalence of polypharmacy, factors associated with polypharmacy, and consequences of polypharmacy in a Crohn's disease population. METHODS: A review of 291 Crohn's disease patients was performed. Polypharmacy was defined as either minor (two to four medications) or major (> or = 5 medications). Clinical status was evaluated with the Harvey-Bradshaw index of disease activity (HBI) and the short inflammatory bowel disease questionnaire (SIBDQ). RESULTS: Major polypharmacy was identified in 50% of patients. Crohn's disease patients on less than two medications at the intake visit had an HBI of 3.6 compared with 5.4 and 6.0 in the minor and major polypharmacy groups (P < 0.05). Similarly, patients on less than two medications had an SIBDQ of 60.3 compared with 55.7 and 53.4 in the minor and major polypharmacy groups (P = 0.11). Predictors of polypharmacy included age > 40 years (OR 1.9), duration of disease > 10 years (OR 2.0), and female sex (OR 2.5). CONCLUSIONS: Polypharmacy is common in Crohn's disease and correlates with increased disease activity and decreased quality of life. Increasing age, increasing duration of disease, and female sex are associated with major polypharmacy. These findings emphasize the need for improved treatment algorithms to optimize Crohn's disease patient management.


Subject(s)
Crohn Disease/drug therapy , Polypharmacy , Adult , Age Factors , Aminosalicylic Acids/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Female , Humans , Male , Middle Aged , Quality of Life , Retrospective Studies , Risk Factors , Severity of Illness Index , Sex Factors
11.
Dis Esophagus ; 15(1): 16-21, 2002.
Article in English | MEDLINE | ID: mdl-12060038

ABSTRACT

The molecular events underlying progression of Barrett's esophagus to adenocarcinoma remain an area of active investigation. Neovascularization and angiogenesis have been studied in esophageal adenocarcinomas by counting of microvessels after staining with vascular markers, and by immunohistochemistry for vascular endothelial growth factor. Angiogenesis appears to be increased early in the neoplastic process, but has poor prognostic value. We have demonstrated that expression levels of two important genes that regulate cell growth, namely inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2, are frequently upregulated in Barrett's esophagus and associated adenocarcinomas. COX-2 expression may be related to reflux of bile salts, which induce COX-2 expression in Barrett's tissues and esophageal adenocarcinoma cells in vitro. COX-2 inhibition induces apoptosis and blocks proliferation in COX-2-expressing esophageal adenocarcinoma cells in vitro, and blocks angiogenesis in both in vivo and in vitro models. Although controversial, recent evidence suggest that iNOS-derived NO can inhibit angiogenesis in some model systems. In conclusion, both iNOS and COX-2 appear to be involved in Barrett's-associated neoplastic progression, but COX-2 inhibition is more promising as a chemopreventive strategy. COX-2 inhibition may exert beneficial effects by decreasing angiogenesis and epithelial proliferation, and by facilitating apoptosis of epithelial cells that have undergone DNA damage.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/pathology , Biomarkers, Tumor/analysis , Cell Transformation, Neoplastic/pathology , Esophageal Neoplasms/pathology , Neovascularization, Pathologic/physiopathology , Adenocarcinoma/genetics , Angiogenesis Inducing Agents/metabolism , Barrett Esophagus/genetics , Esophageal Neoplasms/genetics , Esophagus/blood supply , Humans , Nitric Oxide Synthase/metabolism , Precancerous Conditions/pathology , Prognosis , Prostaglandin-Endoperoxide Synthases/metabolism , Sensitivity and Specificity
12.
Proc Natl Acad Sci U S A ; 98(24): 13844-9, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11717441

ABSTRACT

The antimicrobial effect of nitric oxide (NO) is an essential part of innate immunity. The vigorous host response to the human gastric pathogen Helicobacter pylori fails to eradicate the organism, despite up-regulation of inducible NO synthase (iNOS) in the gastric mucosa. Here we report that wild-type strains of H. pylori inhibit NO production by activated macrophages at physiologic concentrations of l-arginine, the common substrate for iNOS and arginase. Inactivation of the gene rocF, encoding constitutively expressed arginase in H. pylori, restored high-output NO production by macrophages. By using HPLC analysis, we show that l-arginine is effectively consumed in the culture medium by wild-type but not arginase-deficient H. pylori. The substantially higher levels of NO generated by macrophages cocultured with rocF-deficient H. pylori resulted in efficient killing of the bacteria, whereas wild-type H. pylori exhibited no loss of survival under these conditions. Killing of the arginase-deficient H. pylori was NO-dependent, because peritoneal macrophages from iNOS(-/-) mice failed to affect the survival of the rocF mutant. Thus, bacterial arginase allows H. pylori to evade the immune response by down-regulating eukaryotic NO production.


Subject(s)
Arginase/metabolism , Bacterial Proteins , Helicobacter pylori/enzymology , Nitric Oxide/biosynthesis , Animals , Arginase/genetics , Arginase/physiology , Arginine/metabolism , Cell Line , Eukaryotic Cells/metabolism , Gene Expression , Helicobacter pylori/growth & development , Helicobacter pylori/immunology , Helicobacter pylori/physiology , Interferon-gamma/pharmacology , Macrophage Activation , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Knockout , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitrogen Dioxide/metabolism , RNA, Messenger
13.
Oncogene ; 20(3): 329-35, 2001 Jan 18.
Article in English | MEDLINE | ID: mdl-11313962

ABSTRACT

A significant portion of gastric cancers exhibit defective DNA mismatch repair, manifested as microsatellite instability (MSI). High-frequency MSI (MSI-H) is associated with hypermethylation of the human mut-L homologue 1 (hMLH1) mismatch repair gene promoter and diminished hMLH1 expression in advanced gastric cancers. However, the relationship between MSI and hMLH1 hypermethylation has not been studied in early gastric neoplasms. We therefore investigated hMLH1 hypermethylation, hMLH1 expression and MSI in a group of early gastric cancers and gastric adenomas. Sixty-four early gastric neoplasms were evaluated, comprising 28 adenomas, 18 mucosal carcinomas, and 18 carcinomas with superficial submucosal invasion but clear margins. MSI was evaluated using multiplex fluorescent PCR to amplify loci D2S123, D5S346, D17S250, BAT 25 and BAT 26. Methylation-specific PCR was performed to determine the methylation status of hMLH1. In two hypermethylated MSI-H cancers, hMLH1 protein expression was also evaluated by immunohistochemistry. Six of sixty-four early gastric lesions were MSI-H, comprising 1 adenoma, 4 mucosal carcinomas, and 1 carcinoma with superficial submucosal invasion. Two lesions (one adenoma and one mucosal carcinoma) demonstrated low-frequency MSI (MSI-L). The remaining 56 neoplasms were MSI-stable (MSI-S). Six of six MSI-H, one of two MSI-L, and none of thirty MSI-S lesions showed hMLH1 hypermethylation (P<0.001). Diminished hMLH1 protein expression was demonstrated by immunohistochemistry in two of two MSI-H hypermethylated lesions. hMLH1 promoter hypermethylation is significantly associated with MSI and diminished hMLH1 expression in early gastric neoplasms. MSI and hypermethylation-associated inactivation of hMLH1 are more prevalent in early gastric cancers than in gastric adenomas. Thus, hypermethylation-associated inactivation of the hMLH1 gene can occur early in gastric carcinogenesis.


Subject(s)
DNA Methylation , Microsatellite Repeats , Neoplasm Proteins/genetics , Stomach Neoplasms/genetics , Adaptor Proteins, Signal Transducing , Adenoma/genetics , Adenoma/pathology , Base Pair Mismatch , Carcinoma/genetics , Carcinoma/pathology , Carrier Proteins , Case-Control Studies , Gastric Mucosa/pathology , Humans , MutL Protein Homolog 1 , Neoplasm Proteins/metabolism , Nuclear Proteins , Polymerase Chain Reaction/methods , Promoter Regions, Genetic , Stomach Neoplasms/pathology
14.
Cancer Res ; 61(6): 2399-403, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11289104

ABSTRACT

Cyclooxygenase (COX)-2, the inducible form of the rate-limiting enzyme for prostaglandin synthesis, is up-regulated in gastrointestinal cancers and is a key mediator of epithelial cell growth. Helicobacter pylori is causally linked to gastric cancer. In H. pylori gastritis, COX-2 expression localizes to the subepithelial region, with variable levels in the epithelium. In contrast, in gastric cancer, COX-2 strongly predominates in the epithelium, suggesting that the transition to consistent epithelial COX-2 overexpression may be a critical molecular event in gastric carcinogenesis. Because aberrant promoter methylation inhibits expression of a variety of genes in gastrointestinal cancers, we sought to determine whether methylation of the COX-2 promoter could regulate the response to H. pylori in gastric epithelial cells. We assessed COX-2 expression and promoter methylation status in six gastric epithelial cell lines. In all four of the cell lines that exhibited basal expression of COX-2 and a significant increase in expression in response to H. pylori, the COX-2 promoter was unmethylated, whereas in the two cell lines that did not express COX-2, the COX-2 promoter was methylated. Treatment of COX-2-methylated cells with the demethylating agent 5-azacytidine had a modest effect on COX-2 expression, but when 5-azacytidine-treated cells were subsequently stimulated with H. pylori, there was a significant, 5-10-fold enhancement of both COX-2 mRNA and protein expression and release of the COX-2 product, prostaglandin E2. In contrast, in COX-2-expressing cell lines that were unmethylated at the COX-2 promoter, 5-azacytidine had no effect on H. pylori-stimulated COX-2 expression. These findings suggest that loss of COX-2 methylation may facilitate COX-2 expression and promote gastric carcinogenesis associated with H. pylori infection.


Subject(s)
Adenocarcinoma/microbiology , DNA Methylation , Helicobacter Infections/enzymology , Helicobacter pylori , Isoenzymes/biosynthesis , Promoter Regions, Genetic , Prostaglandin-Endoperoxide Synthases/biosynthesis , Stomach Neoplasms/microbiology , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Cyclooxygenase 2 , Epithelial Cells/enzymology , Epithelial Cells/microbiology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Isoenzymes/genetics , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/genetics , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Tumor Cells, Cultured
15.
Free Radic Biol Med ; 29(9): 881-8, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11063913

ABSTRACT

Microvascular endothelial cells play a key role in inflammation by undergoing activation and recruiting circulating immune cells into tissues and foci of inflammation, an early and rate-limiting step in the inflammatory process. We have previously [Binion et al., Gastroenterology112:1898-1907, 1997] shown that human intestinal microvascular endothelial cells (HIMEC) isolated from surgically resected inflammatory bowel disease (IBD) patient tissue demonstrate significantly increased leukocyte binding in vitro compared to normal HIMEC. Our studies [Binion et al., Am. J. Physiol.275 (Gastrointest. Liver Physiol. 38):G592-G603, 1998] have also demonstrated that nitric oxide (NO) production by inducible nitric oxide synthase (iNOS) normally plays a key role in downregulating HIMEC activation and leukocyte adhesion. Using primary cultures of HIMEC derived from normal and IBD patient tissues, we sought to determine whether alterations in iNOS-derived NO production underlies leukocyte hyperadhesion in IBD. Both nonselective (N(G)-monomethyl-L-arginine) and specific (N-Iminoethyl-L-lysine) inhibitors of iNOS significantly increased leukocyte binding by normal HIMEC activated with cytokines and lipopolysaccharide (LPS), but had no effect on leukocyte adhesion by similarly activated IBD HIMEC. When compared to normal HIMEC, IBD endothelial cells had significantly decreased levels of iNOS mRNA, protein, and NO production following activation. Addition of exogenous NO by co-culture with normal HIMEC or by pharmacologic delivery with the long-acting NO donor detaNONOate restored a normal leukocyte binding pattern in the IBD HIMEC. These data suggest that loss of iNOS expression is a feature of chronically inflamed microvascular endothelial cells, which leads to enhanced leukocyte binding, potentially contributing to chronic, destructive inflammation in IBD.


Subject(s)
Endothelium, Vascular/enzymology , Endothelium, Vascular/pathology , Inflammatory Bowel Diseases/enzymology , Inflammatory Bowel Diseases/pathology , Intestines/blood supply , Leukocytes/pathology , Nitric Oxide Synthase/deficiency , Cell Adhesion/physiology , Cells, Cultured , Free Radicals/metabolism , Humans , Inflammatory Bowel Diseases/genetics , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Infect Immun ; 68(11): 6265-72, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11035734

ABSTRACT

The gastric inflammatory and immune response in Helicobacter pylori infection may be due to the effect of different H. pylori products on innate immune mechanisms. The aim of this study was to determine whether bacterial components could modulate cytokine production in vitro and thus contribute to Th1 polarization of the gastric immune response observed in vivo. The effect of H. pylori recombinant urease, bacterial lysate, intact bacteria, and bacterial DNA on proliferation and cytokine production by peripheral blood mononuclear cells (PBMCs) from H. pylori-negative donors was examined as a model for innate cytokine responses. Each of the different H. pylori preparations induced gamma interferon (IFN-gamma) and interleukin-12p40 (IL-12p40), but not IL-2 or IL-5, production, and all but H. pylori DNA stimulated release of IL-10. Addition of anti-IL-12 antibody to cultures partially inhibited IFN-gamma production. In addition, each bacterial product inhibited mitogen-stimulated IL-2 production by PBMCs and Jurkat T cells. The inhibitory effect of bacterial products on IL-2 production correlated with inhibition of mitogen-stimulated lymphocyte proliferation, although urease inhibited IL-2 production without inhibiting proliferation, suggesting that inhibition of IL-2 production alone is not sufficient to inhibit lymphocyte proliferation. The results of these studies demonstrate that Th1 polarization of the gastric immune response may be due in part to the direct effects of multiple different H. pylori components that enhance IFN-gamma and IL-12 production while inhibiting both IL-2 production and cell proliferation that may be necessary for Th2 responses.


Subject(s)
Cytokines/biosynthesis , Helicobacter pylori/immunology , Animals , Cell Line , Humans , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Leukocytes, Mononuclear/metabolism , Mice
17.
Cancer Res ; 60(17): 4864-8, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10987299

ABSTRACT

Twelve to 15% of sporadic colorectal cancers display defective DNA mismatch repair (MMR), manifested as microsatellite instability (MSI). In this group of cancers, promoter hypermethylation of the MMR gene hMLH1 is strongly associated with, and believed to be the cause of, MSI. A subset of colorectal neoplastic lesions arising in inflammatory bowel disease (IBD) is also characterized by MSI. We wished to determine whether hMLH1 hypermethylation was associated with diminished hMLH1 protein expression and MSI in IBD neoplasms. We studied 148 patients with IBD neoplasms, defined as carcinoma or dysplasia occurring in patients with ulcerative colitis or Crohn's disease. MSI was evaluated using multiplex fluorescent PCR to amplify loci D2S123, BAT-25, BAT-26, D5S346, and D17S250 in all cases. Lesions were characterized as high-frequency MSI (MSI-H) if they manifested instability at two or more loci, low-frequency MSI (MSI-L) if unstable at only one locus, or MS-stable (MSS) if showing no instability at any loci. Methylation-specific PCR was performed to determine the methylation status of the hMLH1 promoter region. hMLH1 protein expression was also evaluated by immunohistochemistry. Thirteen (9%) of 148 neoplasms arising in IBD were MSI-H, comprising 11 carcinomas and 2 dysplastic lesions. Sixteen additional lesions (11%) were MSI-L, comprising 11 carcinomas and 5 dysplastic lesions. The remaining 118 neoplasms (80%) were MSS. Six (46%) of 13 MSI-H, 1 (6%) of 16 MSI-L, and 4 (15%) of 27 MSS lesions showed hMLH1 hypermethylation (P = 0.013). Diminished hMLH1 protein expression in neoplastic cell nuclei relative to surrounding normal cell nuclei was demonstrated immunohistochemically in four of four (100%) hypermethylated lesions tested. In IBD neoplasia, hMLH1 promoter hypermethylation occurs frequently in the setting of MSI, particularly MSI-H. Furthermore, hMLH1 hypermethylation and MSI are strongly associated with diminished hMLH1 protein expression in IBD neoplasms. These findings suggest that hMLH1 hypermethylation causes defective DNA MMR in at least a subset of IBD neoplasms.


Subject(s)
Colorectal Neoplasms/genetics , DNA Methylation , DNA Repair/genetics , Inflammatory Bowel Diseases/genetics , Microsatellite Repeats/genetics , Neoplasm Proteins/genetics , Adaptor Proteins, Signal Transducing , Carrier Proteins , Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , Humans , Immunohistochemistry , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/metabolism , MutL Protein Homolog 1 , Neoplasm Proteins/biosynthesis , Nuclear Proteins , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics
18.
Oncogene ; 19(32): 3642-6, 2000 Jul 27.
Article in English | MEDLINE | ID: mdl-10951570

ABSTRACT

The adenomatous polyposis coli (APC) tumor suppressor gene is mutationally inactivated in both familial and sporadic forms of colorectal cancers. In addition, hypermethylation of CpG islands in the upstream portion of APC, a potential alternative mechanism of tumor suppressor gene inactivation, has been described in colorectal cancer. Because a subset of both gastric and colorectal cancers display the CpG island methylator phenotype, we hypothesized that epigenetic inactivation of APC was likely to occur in at least some gastric cancers. APC exhibits two forms of transcripts from exons 1A and 1B in the stomach. Therefore, we investigated CpG island methylation in the sequences upstream of exons 1A and 1B, i.e., promoters 1A and 1B, respectively. We evaluated DNAs from 10 gastric cancer cell lines, 40 primary gastric cancers, and 40 matching non-cancerous gastric mucosae. Methylated alleles of promoter 1A were present in 10 (100%) of 10 gastric cancer cell lines, 33 (82.5%) of 40 primary gastric cancers, and 39 (97.5%) of 40 noncancerous gastric mucosae. In contrast, promoter 1B was unmethylated in all of these same samples. APC transcripts from exon 1A were not expressed in nine of the 10 methylated gastric cancer cell lines, whereas APC transcripts were expressed from exon 1B. Thus, expression from a given promoter correlated well with its methylation status. We conclude that in contrast to the colon, methylation of promoter 1A is a normal event in the stomach; moreover, promoter 1B is protected from methylation in the stomach and thus probably does not participate in this form of epigenetic APC inactivation.


Subject(s)
DNA Methylation , DNA, Neoplasm/metabolism , Genes, APC , Promoter Regions, Genetic , Stomach Neoplasms/genetics , Adolescent , Base Sequence , Female , Gastric Mucosa/metabolism , Gene Expression , Humans , Molecular Sequence Data , RNA, Messenger , RNA, Neoplasm , Tumor Cells, Cultured
19.
J Natl Cancer Inst ; 92(7): 569-73, 2000 Apr 05.
Article in English | MEDLINE | ID: mdl-10749913

ABSTRACT

BACKGROUND: E (epithelial)-cadherin, the cell adhesion molecule also considered a potential invasion/metastasis suppressor, is mutationally inactivated in nearly half of all undifferentiated-scattered (diffuse-type) gastric carcinomas. In addition, silencing of E-cadherin by CpG methylation within its promoter region has been reported in several gastric carcinoma cell lines. We investigated the methylation status of the E-cadherin promoter region in 53 primary human gastric carcinomas. METHODS: Hypermethylation of the E-cadherin promoter was determined by utilizing methylation-specific polymerase chain reaction (PCR)-single-strand conformation polymorphism (MSP-SSCP) analysis followed by direct sequencing of PCR products. Expression of E-cadherin was studied by western blot analysis. All statistical tests were two-sided. RESULTS: Hypermethylation of the E-cadherin promoter was evident in 27 (51%) of 53 primary gastric carcinomas examined by MSP-SSCP. It occurred more frequently in carcinomas of the undifferentiated-scattered type (in 15 [83%] of 18) than in other histologic subtypes (in 12 [34%] of 35) (P =.0011, Fisher's exact test), and it was present at similar rates in early (in six [60%] of 10) versus advanced (in 21 [49%] of 43) carcinomas (P =.73, Fisher's exact test). Methylation occurring at all cytosine-guanosine sequences (CpGs) near the transcriptional start site was confirmed in six of six tumors examined by bisulfite-DNA sequencing, including two early gastric carcinomas. In addition, loss or diminished expression of E-cadherin was confirmed by western blotting in four of the six tumor tissues demonstrating hypermethylation. CONCLUSIONS: The E-cadherin promoter frequently undergoes hypermethylation in human gastric cancers, particularly those of the undifferentiated-scattered histologic subtype. E-cadherin promoter hypermethylation is associated with decreased expression and may occur early in gastric carcinogenesis.


Subject(s)
Cadherins/genetics , Carcinoma/metabolism , Promoter Regions, Genetic/genetics , Stomach Neoplasms/metabolism , Blotting, Western , Carcinoma/genetics , Cytosine/metabolism , DNA Primers , DNA, Neoplasm/chemistry , Down-Regulation , Gene Expression Regulation, Neoplastic , Guanosine/metabolism , Humans , Methylation , Polymerase Chain Reaction/methods , Polymorphism, Single-Stranded Conformational , Sequence Analysis, DNA , Stomach Neoplasms/genetics
20.
Curr Opin Gastroenterol ; 16(1): 51-5, 2000 Jan.
Article in English | MEDLINE | ID: mdl-17024016

ABSTRACT

Treatment of gastrointestinal infections continues to be complicated by expanding resistance to antibiotics. This has led to both new antibiotic treatments and to research on alternative therapies. In the case of Clostridium difficile colitis, interesting preliminary results have been obtained with both probiotic therapy and passive immunization strategies. The newer macrolides were found to be effective for multidrug-resistant Salmonella typhi and safe in the pediatric population. The efficacy of rifaximin, a nonabsorbed antimicrobial agent, was demonstrated in the treatment of travelers diarrhea in Mexico. The benefit of the proteolytic agent bromelain, which degrades enterocyte receptors for enterotoxigenic Escherichia coli, was shown in an experimental model of piglets challenged with this infection after bromelain prophylaxis. The potential value of zinc supplementation in infant diarrhea was shown in a study in India. The recently approved rotavirus vaccine is being withheld because of multiple reported cases of intussusception. Cryptosporidiosis continues to challenge old and new chemotherapeutic agents. The usefulness of paromomycin in Cryptosporidium infection was shown in both murine and piglet models. The cost effectiveness of prophylaxis for parasitosis in immigrants has been an area of investigation.

SELECTION OF CITATIONS
SEARCH DETAIL
...