Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Pathol Int ; 67(3): 131-140, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28088838

ABSTRACT

A total of 313 cases of differentiated-type early gastric adenocarcinomas, including 113 cases of small-sized carcinoma (5< × ≤10 mm) and 121 cases of microcarcinoma (0< × ≤5 mm), were examined immunohistochemically to clarify the phenotypic expressions. They were classified into four categories (gastric phenotype (G-type), intestinal phenotype, gastrointestinal phenotype, and null phenotype) by a two-step process: the phenotype based on an immunoprofile of mucin core proteins (MUCs) with CDX2 (w/.CDX2-assessment); and the phenotype of MUCs only (w/o.CDX2-assessment). CDX2 expression was observed in 89.1% (279/313); it was highly expressed in 87.6% (106/121) of microcarcinomas. MUC2 expression increased as tumor size increased (P < 0.05). Compared with w/o.CDX2-assessment, w/.CDX2-assessment showed significantly fewer G-type carcinomas (P < 0.05). Each phenotype marker was less expressed in the submucosal part than in the mucosal part. In conclusion, CDX2 was a sensitive marker for assessing intestinal phenotype. A large portion of the early differentiated-type adenocarcinomas expressed CDX2 from the very early stage of carcinogenesis, and the proportion of G-type was unexpectedly low. Lower expression of each phenotype marker was considered the cause of phenotype alteration during submucosal invasion.


Subject(s)
Adenocarcinoma/classification , Adenocarcinoma/pathology , Biomarkers, Tumor/analysis , Stomach Neoplasms/classification , Stomach Neoplasms/pathology , Adult , Aged , Aged, 80 and over , CDX2 Transcription Factor/analysis , CDX2 Transcription Factor/biosynthesis , Female , Gastric Mucins/analysis , Gastric Mucins/biosynthesis , Humans , Immunohistochemistry , Male , Middle Aged , Phenotype
2.
Cell Rep ; 16(2): 432-443, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27346351

ABSTRACT

Clostridium difficile infection (CDI) is the most common cause of hospital-acquired infection in the United States. Host susceptibility and the severity of infection are influenced by disruption of the microbiota and the immune response. However, how the microbiota regulate immune responses to mediate CDI outcome remains unclear. Here, we have investigated the role of the microbiota-linked cytokine IL-25 during infection. Intestinal IL-25 was suppressed during CDI in humans and mice. Restoration of IL-25 reduced CDI-associated mortality and tissue pathology even though equivalent levels of C. difficile bacteria and toxin remained in the gut. IL-25 protection was mediated by gut eosinophils, as demonstrated by an increase in intestinal eosinophils and a loss of IL-25 protection upon eosinophil depletion. These findings support a mechanism whereby the induction of IL-25-mediated eosinophilia can reduce host mortality during active CDI. This work may provide targets for future development of microbial or immune-based therapies.


Subject(s)
Clostridioides difficile/immunology , Enterocolitis, Pseudomembranous/metabolism , Eosinophils/immunology , Interleukins/physiology , Animals , Enterocolitis, Pseudomembranous/immunology , Enterocolitis, Pseudomembranous/microbiology , Gastric Mucins/biosynthesis , Gastrointestinal Microbiome , Humans , Interleukin-4/metabolism , Interleukins/pharmacology , Leukocyte Count , Male , Mice, Inbred C57BL , Mucous Membrane/immunology , Mucous Membrane/pathology , Protective Factors
3.
Microbiol Spectr ; 3(3)2015 Jun.
Article in English | MEDLINE | ID: mdl-26185079

ABSTRACT

Host bacterial associations have a profound impact on health and disease. The human gastrointestinal (GI) tract is inhabited by trillions of commensal bacteria that aid in the digestion of food and vitamin production and play crucial roles in human physiology. Disruption of these relationships and the structure of the bacterial communities that inhabit the gut can contribute to dysbiosis, leading to disease. This fundamental relationship between the host and microbiota relies on chemical signaling and nutrient availability and exchange. GI pathogens compete with the endogenous microbiota for a colonization niche (1, 2). The ability to monitor nutrients and combine this information with the host physiological state is important for the pathogen to precisely program the expression of its virulence repertoire. A major nutrient source is carbon, and although the impact of carbon nutrition on the colonization of the gut by the microbiota has been extensively studied, the extent to which carbon sources affect the regulation of virulence factors by invading pathogens has not been fully defined. The GI pathogen enterohemorrhagic E. coli (EHEC) gages sugar sources as an important cue to regulate expression of its virulence genes. EHEC senses whether it is in a gluconeogenic versus a glycolytic environment, as well as fluctuations of fucose levels to fine tune regulation of its virulence repertoire.


Subject(s)
Enterohemorrhagic Escherichia coli/pathogenicity , Gastric Mucins/metabolism , Gastrointestinal Microbiome/genetics , Gastrointestinal Tract/microbiology , Polysaccharides/metabolism , Carbon/metabolism , Energy Metabolism/physiology , Escherichia coli Proteins/genetics , Gastric Mucins/biosynthesis , Gene Expression Regulation, Bacterial , Humans , Symbiosis , Virulence Factors/genetics
4.
Biomed Res ; 33(1): 45-51, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22361886

ABSTRACT

Compared with the aggressive factors, little attention has been paid to the mucosal defensive factors in ulcer therapy, and the role of the H2-receptor antagonists in gastric mucosal protection has not been well characterized. In the present study, the effects of different types of H2-receptor antagonists (famotidine and roxatidine) on rat gastric mucus cells were investigated using both biochemical and histological methods. Each drug (famotidine, 3 mg/kg; roxatidine, 100 mg/kg) was orally administered to rats by gavage once daily for 7 days. The biosynthesis and tissue content of mucin were compared in the gastric mucosa treated with each drug. Using anti-mucin monoclonal antibodies, the mucin content and immunohistochemical localization were also compared. Both the biosynthesis and the accumulation of gastric mucin were significantly decreased in the famotidine-treated rats, but not in the roxatidine. Both the content and the immunoreactivity of surface mucus cell-derived mucin were reduced by famotidine, while they were maintained in roxatidine-treated rat stomachs. There was no difference between the groups in the content and immunoreactivity of mucous neck cell-derived mucin. H2-receptor antagonists should be classified in relation to gastric surface mucus cell function, raising the possibility of more effective ulcer therapy.


Subject(s)
Anti-Ulcer Agents/pharmacology , Famotidine/pharmacology , Gastric Mucosa/drug effects , Histamine H2 Antagonists/pharmacology , Piperidines/pharmacology , Animals , Gastric Mucins/biosynthesis , Gastric Mucosa/metabolism , Male , Mucus/drug effects , Rats , Rats, Wistar
5.
Inflammopharmacology ; 19(2): 89-97, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21279549

ABSTRACT

Loss of mucus coat integrity and the impairment in its mucin component as well as the disturbance in nitric oxide (NO) generation are well-recognized features of gastric disease associated with H. pylori infection. As ghrelin plays a major role in the regulation of nitric oxide synthase system, we investigated the influence of this hormone on H. pylori LPS-induced interference with gastric mucin synthesis. The results revealed that the LPS-induced impairment in mucin synthesis and accompanied induction in inducible nitric oxide synthase (iNOS) expression, were associated with the suppression in Akt kinase activity and the impairment in constitutive nitric oxide synthase (cNOS) phosphorylation. The LPS effect on Akt inactivation was manifested in the kinase protein S-nitrosylation and a decrease in its phosphorylation at Ser(473). Further, we show that the countering effect of ghrelin, on the LPS-induced impairment in mucin synthesis was reflected in the suppression of iNOS and the increase in Akt activation, associated with the loss in S-nitrosylation and the increase in phosphorylation, as well as cNOS activation through phosphorylation. Our findings demonstrate that up-regulation in iNOS with H. pylori infection and subsequent Akt kinase inactivation through S-nitrosylation exerts the detrimental effect on the processes dependent on Akt activation, including that of cNOS activation and mucin synthesis. We also show that ghrelin protection against H. pylori-induced impairment in mucin synthesis is intimately linked to the events of Akt activation and reflected in a decrease in the kinase S-nitrosylation and the increase in its phosphorylation.


Subject(s)
Cysteine/analogs & derivatives , Gastric Mucins/biosynthesis , Ghrelin/pharmacology , Lipopolysaccharides/pharmacology , Protein Processing, Post-Translational/physiology , Proto-Oncogene Proteins c-akt/metabolism , S-Nitrosothiols/metabolism , Animals , Ascorbic Acid/pharmacology , Cells, Cultured , Cysteine/metabolism , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gastric Mucosa/cytology , NF-kappa B/antagonists & inhibitors , Naphthoquinones/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rats , Rats, Inbred Strains , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
6.
Glycoconj J ; 27(1): 125-32, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19562481

ABSTRACT

Endo-alpha-N-acetylgalactosaminidase catalyzes the release of Galbeta1-3GalNAc from the core 1-type O-glycan (Galbeta1-3GalNAcalpha1-Ser/Thr) of mucin glycoproteins and synthetic p-nitrophenyl (pNP) alpha-linked substrates. Here, we report the enzymatic syntheses of core 1 disaccharide-containing glycopeptides using the transglycosylation activity of endo-alpha-N-acetylgalactosaminidase (EngBF) from Bifidobacterium longum. The enzyme directly transferred Galbeta1-3GalNAc to serine or threonine residues of bioactive peptides such as PAMP-12, bradykinin, peptide-T and MUC1a when Galbeta1-3GalNAcalpha1-pNP was used as a donor substrate. The enzyme was also found to catalyze the reverse-hydrolysis reaction. EngBF synthesized the core 1 disaccharide-containing oligosaccharides when the enzyme was incubated with either glucose or lactose and Galbeta1-3GalNAc prepared from porcine gastric mucin using bifidobacterial cells expressing endo-alpha-N-acetylgalactosaminidase. Synthesized oligosaccharides are promising prebiotics for bifidobacteria.


Subject(s)
Bifidobacterium/enzymology , Gastric Mucins/biosynthesis , Glycopeptides/biosynthesis , Oligosaccharides/biosynthesis , alpha-N-Acetylgalactosaminidase/metabolism , Amino Acid Sequence , Animals , Bifidobacterium/cytology , Galactosamine/metabolism , Glycopeptides/chemistry , Glycosylation , Hydrolysis , Molecular Sequence Data , Serine/metabolism , Sus scrofa , Threonine/metabolism
7.
J Med Dent Sci ; 56(1): 25-35, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19697516

ABSTRACT

The intramucosal lesion of gastric signet ring cell carcinoma (SIG) is known to form a layered structure (LS) that simulates mucin expression in ordinary gastric mucosa. In this study, we suspected the influence of background mucosa on the formation of LS and performed histopathological analysis. We examined 35 cases of intramucosal SIG with a maximum diameter of 30 mm or less. The LS patterns were classified into those with a layer of MUC6-positive cells (complete pattern, CP) and those lacking this layer (incomplete pattern, ICP). The relationship between LS patterns and the characteristics of the background mucosa, the expression of MUC2 (intestinal-type mucin antigen), MUC5AC (foveolar-type mucin antigen), and Ki-67 (the marker of cell proliferation activity) was examined by histochemistry and immunohistochemistry. Intestinal metaplasia in the background mucosa and MUC2 expression were frequently observed in cases with ICP. Ki-67-positive cells were much more and they were distributed more widely in the lesion of cases with ICP alone than in the other cases. Mucin expression and LS formation of gastric SIG are strongly influenced by its background mucosa. The cases completely lacking MUC6 expression may have higher malignant potential.


Subject(s)
Carcinoma, Signet Ring Cell/pathology , Gastric Mucins/biosynthesis , Gastric Mucosa/metabolism , Stomach Neoplasms/pathology , Carcinoma, Signet Ring Cell/metabolism , Cell Differentiation , Cell Proliferation , Female , Humans , Immunohistochemistry , Intestinal Mucosa/metabolism , Ki-67 Antigen/biosynthesis , Male , Middle Aged , Mucin-2/biosynthesis , Stomach Neoplasms/metabolism
8.
J Gastroenterol Hepatol ; 24(3): 488-92, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19175830

ABSTRACT

BACKGROUND AND AIM: In Japan, peptic ulcer disease (PUD) is treated clinically with a combination of a mucosal protectant and acid suppressants, but there is scant information regarding the effects of these drugs on normal gastric mucus cells. In the present study, the effects of co-administration of methylmethionine sulfonium chloride (MMSC) and famotidine on rat gastric mucus cells were investigated using both biochemical and histological methods. METHODS: Rats were divided into four groups: controls were given carboxymethylcellulose orally once daily for 7 days and the second, third and fourth groups were treated similarly with famotidine (famotidine group), MMSC (MMSC group) or famotidine plus MMSC (combination group). After killing the rats on the 8th day, the stomachs were removed and the biosynthesis and amount of mucin in different areas of the gastric mucosa were compared among groups. Using anti-mucin monoclonal antibodies, the mucin content and immunoreactivity were also compared. RESULTS: Both the biosynthesis and accumulation of mucin were significantly decreased in the famotidine group, but increased in the MMSC and combination groups. The amount and immunoreactivity of surface mucus cell-derived mucin were both reduced in the famotidine group, and increased in the MMSC and combination groups. There was no difference among the groups in the content and immunoreactivity of gland mucus cell-derived mucin. CONCLUSION: Famotidine-induced suppression of gastric surface mucus cell function is prevented by combined treatment with MMSC, raising the possibility of a more effective cure of PUD.


Subject(s)
Anti-Ulcer Agents/pharmacology , Famotidine/pharmacology , Gastric Mucins/biosynthesis , Gastric Mucosa/drug effects , Histamine H2 Antagonists/pharmacology , Vitamin U/pharmacology , Administration, Oral , Animals , Anti-Ulcer Agents/administration & dosage , Anti-Ulcer Agents/toxicity , Drug Administration Schedule , Drug Therapy, Combination , Famotidine/administration & dosage , Famotidine/toxicity , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Histamine H2 Antagonists/administration & dosage , Histamine H2 Antagonists/toxicity , Male , Rats , Rats, Wistar , Vitamin U/administration & dosage
9.
J Physiol Pharmacol ; 58(1): 117-30, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17440231

ABSTRACT

Activation of cytosolic phospholipase A(2) (cPLA(2)) by bacterial LPS for the rapid release of arachidonic acid from membrane phospholipids is considered a key step in the generation of platelet-activating factor (PAF), recognized as the most proximal mediator of inflammatory events triggered by bacterial infection. In this study, we report on the role of leptin in modulation of the detrimental consequences of H. pylori LPS-induced cPLA(2) activation that result in the disturbances in gastric mucin synthesis. Employing gastric mucosal cells labeled with [(3)H] arachidonic acid, we show that H. pylori LPS-induced cPLA(2) activation, associated with up-regulation in apoptosis and PAF generation, and the impairment in gastric mucin synthesis, was subject to a dose-dependent suppression by leptin, as well as the inhibition by MAFP, a specific inhibitor of cPLA(2). A potentiation in the countering capacity of leptin on the LPS-induced up-regulation in apoptosis, arachidonic acid release and PAF generation was attained in the presence of ERK inhibitor, PD98059, while PI3K inhibitor, wortmannin had no effect. On the other hand, the prevention by leptin of the LPS detrimental effect on mucin synthesis was subject to suppression by wortmannin, an inhibitor of PI3K as well as the inhibitor of ERK, PD98059. Moreover, potentiation in the effect of leptin on the LPS-induced decrease in mucin synthesis was attained with cPLA(2) inhibitor, MAFP as well as PAF receptor antagonist, BN52020. The results of our findings point to H. pylori LPS-induced ERK-dependent cPLA(2) activation as a critical factor influencing the level of PAF generation, and hence the extent of pathological consequences of H. pylori infection on the synthesis of gastric mucin. Furthermore, we show that leptin counters the pathological consequences of H. pylori-induced cPLA(2) activation on gastric mucin synthesis through the involvement in signaling events controlled by MAPK/ERK and PI3K pathways.


Subject(s)
Gastric Mucins/biosynthesis , Gastric Mucosa/drug effects , Leptin/metabolism , Lipopolysaccharides/pharmacology , Phospholipases A/metabolism , Signal Transduction/drug effects , Androstadienes/pharmacology , Animals , Apoptosis/drug effects , Arachidonic Acid/metabolism , Arachidonic Acids/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Gastric Mucosa/enzymology , Ginkgolides , Lactones/pharmacology , Leptin/pharmacology , Organophosphonates/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phospholipases A/antagonists & inhibitors , Phospholipases A2 , Platelet Activating Factor/biosynthesis , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Wortmannin
10.
Clin Cancer Res ; 12(21): 6469-79, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17085661

ABSTRACT

PURPOSE: Gastric and intestinal phenotypic cell markers are expressed in gastric carcinomas, irrespective of their histologic type. In the present study, we determined the clinicopathologic significance of phenotypic marker expression in early-stage gastric differentiated-type tumors and the association between marker expression and genetic alterations. EXPERIMENTAL DESIGN: Phenotypic marker expression was determined by examining the expressions of human gastric mucin (HGM), MUC6, MUC2, and CD10 in 63 gastric adenomas, 133 early differentiated-type carcinomas, and 24 follow-up cases with gastric adenoma. Tumors were classified into gastric, gastric and intestinal mixed, or intestinal phenotypes according to the immunopositivity of the above markers. The presence of mutations in APC, K-ras, and p53 and the microsatellite instability status were also determined in all tumors. RESULTS: The expressions of HGM and MUC6, representing gastric or gastric and intestinal mixed phenotypes, were significantly associated with high-grade atypia in the 63 gastric adenomas. Among the 133 early differentiated-type carcinomas, HGM expression was significantly associated with mixed-type (with an undifferentiated-type component) tumors and lymph node metastasis. MUC2 expression was inversely associated with submucosal invasion. A multivariate analysis revealed that gastric adenomas were significantly associated with the intestinal phenotype and were inversely associated with p53 mutation compared with early differentiated-type carcinomas. Among all 196 tumors, APC mutation was significantly associated with CD10 expression and the intestinal phenotype and was inversely associated with the expressions of HGM and MUC6. The microsatellite instability status was significantly associated with MUC6 expression. Malignant transformation from gastric adenoma to carcinoma was shown in 5 of the 24 follow-up cases of gastric adenoma. The malignant transformation was significantly associated with the gastric and intestinal mixed phenotype and was inversely associated with APC mutation. No malignant transformation was found in intestinal phenotype gastric adenomas with APC mutation. CONCLUSIONS: Our present findings show that phenotypic marker expression is associated with tumor aggressiveness during the early stage of gastric differentiated-type tumors. Differences in the biological behavior of tumors with different phenotypes may result from differences in the genetic backgrounds during the incipient phase of gastric tumorigenesis.


Subject(s)
Adenoma/genetics , Adenoma/metabolism , Biomarkers, Tumor/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Adenoma/pathology , DNA Mutational Analysis , Gastric Mucins/biosynthesis , Genes, APC , Genes, p53 , Genes, ras , Humans , Immunohistochemistry , Microsatellite Instability , Microsatellite Repeats , Mucin-2 , Mucin-6 , Mucins/biosynthesis , Mutation , Neprilysin/biosynthesis , Phenotype , Polymerase Chain Reaction , Stomach Neoplasms/pathology
11.
J Cancer Res Clin Oncol ; 132(6): 363-75, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16447040

ABSTRACT

Gastric and intestinal phenotypic cell markers are widely expressed in gastric carcinomas, irrespective of their histological type. In the present study, the relations between the phenotypic marker expression of the tumour, histological findings, expression of cell adhesion molecules, and the chromosomal changes in gastric differentiated-type carcinomas were examined. The phenotypic marker expression of the tumour was determined by the combination of the expression of the human gastric mucin (HGM), MUC6, MUC2 and CD10, and was evaluated in comparison with the expression of cell adhesion molecules, such as E-cadherin and beta-catenin, and chromosomal changes by comparative genomic hybridization (CGH) in 34 gastric differentiated-type carcinomas. Tumours were classified into the gastric- (G-), gastric and intestinal mixed- (GI-), intestinal- (I-), or unclassified- (UC-) phenotype according to the immunopositivity of staining for HGM, MUC6, MUC2, and CD10. G-phenotype tumours were significantly associated with a higher incidence of differentiated-type tumours mixed with undifferentiated-type component, compared with GI- and I-phenotype tumours (88.9 vs 33.3%, P=0.0498 and 88.9 vs 42.9%, P=0.0397; respectively). HGM-positive tumours were significantly associated with a higher incidence of tumours with abnormal expression of E-cadherin, compared with HGM-negative tumours (66.7 vs 21.1%, P=0.0135). GI-phenotype tumours were significantly associated with a higher incidence of tumours with abnormal expression of E-cadherin, compared with I-phenotype tumours (77.8 vs 21.4%, P=0.0131). HGM-negative tumours were significantly associated with higher frequencies of the gains of 19q13.2 and 19q13.3, compared with HGM-positive tumours (57.9 vs 20.0%, P=0.0382 and 63.2 vs 13.3%, P=0.0051; respectively). MUC6-positive tumours were significantly associated with higher frequencies of the gains of 20q13.2, compared with MUC6-negative tumours (71.4 vs 30.0%, P=0.0349). MUC2-positive tumours were significantly associated with the gain of 19p13.3, compared with MUC2-negative tumours (41.2 vs 5.9%, P=0.0391). I-phenotype tumours were significantly associated with higher frequencies of gains of 5p15.2 and 13q33-34, compared with G-phenotype tumours (66.7 vs 0%, P=0.0481, each) and also associated with higher frequencies of gain of 7p21, compared with GI-phenotype tumours (66.7 vs 0%, P=0.0481). Our present results show that gastric differentiated-type carcinomas have different characteristics according to the phenotypic marker expression of the tumour in terms of histological findings, E-cadherin expression and pattern of chromosomal changes.


Subject(s)
Biomarkers, Tumor/biosynthesis , Cadherins/biosynthesis , Carcinoma/genetics , Chromosome Aberrations , Gene Expression Regulation, Neoplastic , Stomach Neoplasms/genetics , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinoma/metabolism , Carcinoma/pathology , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Female , Gastric Mucins/biosynthesis , Gastric Mucins/genetics , Humans , Immunohistochemistry , Male , Middle Aged , Mucin-2 , Mucin-6 , Mucins/biosynthesis , Mucins/genetics , Neprilysin/biosynthesis , Neprilysin/genetics , Nucleic Acid Hybridization , Phenotype , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , beta Catenin/biosynthesis
12.
Dis Esophagus ; 18(6): 383-7, 2005.
Article in English | MEDLINE | ID: mdl-16336609

ABSTRACT

Intestinal metaplasia is a prerequisite criterion for the diagnosis of Barrett's metaplasia and the sole columnar esophageal lining associated with malignancy. It is recognized by the presence of goblet cells, but columnar non-goblet elements, producing gastric or intestinal proteins, are the prevalent cell population. The cellular heterogeneity of Barrett's metaplasia is well documented but the relationship between the distinct cell subtypes and neoplasia is unclear. Our aim was to clarify the relationship between the different metaplastic populations and malignancy in order to investigate putative markers for risk stratification of Barrett's patients. We studied 46 columnar-lined esophageal segments, 15 with associated adenocarcinoma. The presence of the gastric, MUC5AC and MUC6, and the intestinal, MUC2, proteins was evaluated in metaplastic (columnar and goblet) and neoplastic cells. In neoplasia MUC5AC and MUC6 were detected in 100% and 86.6% of the cases, respectively. In metaplasia there were no differences in MUC5AC and MUC6 immunoreactivity, between cases with and without associated neoplasia, except for goblet elements producing MUC6 that were exclusive of metaplasia adjacent to adenocarcinoma (P < 0.05). MUC2 was present in 86.6% of the neoplasia. In metaplasia it was restricted to Barrett's cases and was more frequent in areas with intestinal metaplasia. Columnar-lined esophagus without intestinal metaplasia did not express MUC2. Our study suggests a relationship between the metaplastic population with gastric phenotype and malignancy, and points to the involvement of columnar as well as goblet elements in tumorigenesis. The association between goblet cells aberrantly producing MUC6 and the presence of neoplasia suggests they may be useful for risk stratification.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/pathology , Esophageal Neoplasms/pathology , Gastric Mucosa/pathology , Biomarkers, Tumor/biosynthesis , Cell Transformation, Neoplastic/pathology , Diagnosis, Differential , Gastric Mucins/biosynthesis , Goblet Cells/pathology , Humans , Immunohistochemistry , Intestines/pathology , Metaplasia , Mucin-6 , Mucins/biosynthesis
13.
Scand J Gastroenterol ; 40(8): 921-28, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16173126

ABSTRACT

OBJECTIVE: Endothelin-1 (ET-1), a key mediator of inflammatory processes associated with bacterial infection, is a 21-amino acid peptide produced from a biologically inactive big ET-1 by the action of endothelin-converting enzyme-1 (ECE-1) that acts through G protein-coupled ET(A) and ET(B) receptors. Here we report on the role of ET-1 in the mediation of the detrimental influence of Helicobacter pylori on the synthesis of gastric mucin. MATERIAL AND METHODS: Rat gastric mucosal cells were exposed to H. pylori key virulence factor, lipopolysaccharide (LPS). RESULTS: The LPS inhibitory effect on gastric mucin synthesis was accompanied by a marked increase in ET-1 generation and enhancement in ECE-1 activity. Inhibition of ECE-1 with phosphoramidon not only led to the impedance of LPS-induced ET-1 generation, but also countered the detrimental effect of LPS on mucin synthesis. Moreover, the LPS inhibitory effect on mucin synthesis was blocked by ET(A) receptor antagonist BQ610, but not by ET(B) receptor antagonist BQ788. Furthermore, the LPS-induced suppression in gastric mucin synthesis was countered in a concentration-dependent fashion by PD153035 (81.7%), a specific inhibitor of epidermal growth factor receptor (EGFR) kinase as well as PP2 (69.8%), a selective inhibitor of tyrosine kinase Src responsible for ligand-independent EGFR transactivation. CONCLUSIONS: Our findings are the first to show that the detrimental effect of H. pylori on gastric mucin synthesis is intimately linked to the events associated with ECE-1 up-regulation, enhancement in ET-1 production, and G protein-coupled ET(A) receptor activation that triggers the EGFR transactivation.


Subject(s)
Endothelin-1/metabolism , ErbB Receptors/metabolism , Gastric Mucins/biosynthesis , Helicobacter pylori , Lipopolysaccharides/pharmacology , Transcriptional Activation/physiology , Up-Regulation/physiology , Animals , Aspartic Acid Endopeptidases/drug effects , Aspartic Acid Endopeptidases/metabolism , Cells, Cultured , Endothelin-1/genetics , Endothelin-Converting Enzymes , Enzyme Inhibitors/pharmacology , ErbB Receptors/drug effects , Gastric Mucins/drug effects , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , In Vitro Techniques , Metalloendopeptidases/drug effects , Metalloendopeptidases/metabolism , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley
14.
Eur J Cell Biol ; 83(7): 347-58, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15503858

ABSTRACT

The expression of mucin genes was evaluated in rat intestinal cell lines in order to establish an in vitro model for investigating the regulation of intestinal mucin expression in this species. Two rat intestinal cancer cell lines (DHE, LGA) and three nontumoral rat intestinal cell lines (IEC6, IEC17, IEC18) were screened. The mRNA expression of rMuc1, rMuc2, rMuc3, rMuc4, and rMuc5AC mucin genes was studied by semiquantitative RT-PCR, real-time RT-PCR and Northern-blot analysis. Results were correlated with immunohistochemical expression of rat gastric and intestinal mucin proteins, and secretion of glycoconjugates was examined by enzyme-linked lectin assay. We showed that mRNA of rMucl and rMuc2 were constitutively expressed in all IEC cell populations but periodic acid Schiff staining of these cells did not reveal the presence of glycoproteins. DHE cells expressed rMuc1-5AC mRNA and LGA expressed the same mucins but the level of rMuc4 was much lower. Mucin mRNA expression also differed in relation with the length of cultivation. Immunocytochemical studies revealed the presence of gastric and intestinal mucins in the two tumoral cell lines. Functional experiments showed that bethanechol, A23187 and PMA stimulated release of glycoconjugates in DHE but not in LGA cells. Treatment of DHE cells with dexamethasone (10(-7) mol/l) enhanced rMuc2 mRNA but decreased rMuc1 and rMuc5AC mRNA. Real-time RT-PCR showed that the expression of rMuc1 and rMuc5AC genes was reduced by more than tenfold after 24 h. The increased expression of rMuc2 gene was confirmed by Northern blot analysis. In conclusion, DHE cells provide a valuable cellular model for research on rat mucin secretion and expression.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cell Line, Tumor , Dexamethasone/pharmacology , Gastric Mucins/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Animals , Bethanechol/pharmacology , Blotting, Northern , Calcimycin/pharmacology , Carcinogens/pharmacology , Cell Line, Tumor/metabolism , Cell Line, Tumor/pathology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Immunohistochemistry , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Ionophores/pharmacology , Muscarinic Agonists/pharmacology , RNA, Messenger/biosynthesis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Tetradecanoylphorbol Acetate/pharmacology
15.
Inflammopharmacology ; 12(2): 177-88, 2004.
Article in English | MEDLINE | ID: mdl-15265318

ABSTRACT

Peroxisome-proliferator-activated receptor gamma (PPARgamma) is recognized for its role in regulation of genes associated with inflammation, and its activation of phosphatidylinositol 3-kinase (PI3K) has emerged recently as an important regulator of mucosal responses to bacterial infection. In this study, we report that PPARgamma activation leading to the impedance of Helicobacter pylori lipopolysaccharide (LPS) inhibitory effect on salivary mucin synthesis requires epidermal growth factor receptor (EGFR) participation. Using gastric mucosal cells in culture, we show that activation of PPARgamma with a specific agonist, ciglitazone, prevents the LPS-induced reduction in mucin synthesis, and the effect is reflected in a marked decrease in apoptosis, caspase-3 activity and NO generation. The impedance by ciglitazone of the LPS-induced reduction in mucin synthesis was blunted (up to 65.8%) in a concentration-dependent fashion by a specific inhibitor of EGFR kinase, PD153035, as well as the PPARgamma antagonist BADGE, and wortmannin, an inhibitor of PI3K. Moreover, the inhibitory effect of ciglitazone on the LPS-induced reduction in mucin synthesis and upregulation in apoptosis, caspase-3 activity and NO generation was countered by PP2, a selective inhibitor of tyrosine kinase Src responsible for ligand-independent EGFR transactivation. These findings indicate that PPARgamma activation leading to the suppression of H. pylori LPS inhibition of gastric mucin synthesis involves Src kinase-dependent EGFR transactivation.


Subject(s)
ErbB Receptors/genetics , ErbB Receptors/metabolism , Gastric Mucins/biosynthesis , Helicobacter pylori/growth & development , Helicobacter pylori/metabolism , PPAR gamma/physiology , Transcriptional Activation/physiology , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , ErbB Receptors/physiology , Gastric Mucins/antagonists & inhibitors , Helicobacter pylori/drug effects , Rats , Rats, Sprague-Dawley , Thiazolidinediones/pharmacology , Transcriptional Activation/drug effects
16.
IUBMB Life ; 56(1): 41-6, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14992379

ABSTRACT

Platelet-activating factor (PAF) is now recognized as the most proximal mediator of cellular events triggered by bacterial infection. In this study, we report that a specific PAF antagonist, BN52020, impedes the reduction in mucin synthesis evoked in gastric mucosal cells by H. pylori LPS. The impedance by BN52020 of the LPS inhibitory effect on mucin synthesis was blocked by wortmannin, an inhibitor of phosphatidylinositol 3-kinase (P13K), which also obviated the inhibitory effect of BN52020 on the LPS-induced upregulation in apoptosis, TNF-alpha, and NO generation. A reduction in the impedance by BN52020 of the LPS detrimental effect on mucin synthesis was also attained with cNOS inhibitor, L-NNA, whereas NOS-2 inhibitor, 1400W caused a potentiation in the impedance effect of BN52020. However, while 1400W and BN52020 countered the potentiating effect of wortmannin on the LPS-induced decrease in mucin synthesis, a further exacerbation of the potentiating effect of wortmannin was attained in the presence of cNOS inhibitor, L-NNA. Our findings suggest that PAF, through the interference with PI3K-dependent cNOS activation, plays a critical role in influencing the extent of pathological consequences of H. pylori infection on the synthesis of gastric mucin.


Subject(s)
Gastric Mucins/biosynthesis , Gastric Mucins/drug effects , Helicobacter pylori/pathogenicity , Lipopolysaccharides/toxicity , Platelet Activating Factor/metabolism , Amidines/pharmacology , Androstadienes/pharmacology , Animals , Benzylamines/pharmacology , Enzyme Inhibitors/pharmacology , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastritis/etiology , Gastritis/metabolism , Ginkgolides , Helicobacter Infections/etiology , Helicobacter Infections/metabolism , In Vitro Techniques , Lactones/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Phosphoinositide-3 Kinase Inhibitors , Platelet Activating Factor/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Wortmannin
17.
Am J Pathol ; 164(2): 689-99, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14742272

ABSTRACT

We evaluated the significance of aberrant DNA methyltransferase 1 (DNMT1) protein expression during gastric carcinogenesis. The protein expression of DNMT1, Muc2, human gastric mucin, E-cadherin, and proliferating cell nuclear antigen was examined immunohistochemically in gastric cancers and corresponding noncancerous mucosae from 134 patients. The DNA methylation status of the CpG islands of the p16, human MutL homologue 1 (hMLH1), E-cadherin, and thrombospondin-1 (THBS-1) genes and the methylated in tumor (MINT)-1, -2, -12, and -31 clones was examined by methylation-specific polymerase chain reaction and combined bisulfite restriction enzyme analysis. Epstein-Barr virus (EBV) infection was detected by in situ hybridization. Nuclear immunoreactivity for DNMT1 was not detected in any of the noncancerous epithelia, except in proliferative zones (positive internal control), but was found in 97 (72%) of the gastric cancers. DNMT1 overexpression correlated significantly with poorer tumor differentiation (P < 0.001), but not with the phenotype (gastric type versus intestinal type) of the cancer cells. It also correlated significantly with DNA hypermethylation of the CpG islands of the hMLH1 (P = 0.024) and THBS-1 genes (P = 0.043), and with the CpG island methylator phenotype in the gastric cancers (P = 0.007). Reduced E-cadherin expression correlated significantly with poorer tumor differentiation (P = 0.002), DNA hypermethylation of the E-cadherin gene (P < 0.001) and DNMT1 overexpression (P = 0.014). DNMT1 overexpression was also associated with EBV infection (a potential etiological factor in gastric carcinogenesis) but not with the proliferative activity of the cancer cells as indicated by the proliferating cell nuclear antigen-labeling index. These results suggest that DNMT1 overexpression may not be just a secondary effect of increased cancer cell proliferative activity, but may be associated with EBV infection and other etiological factors during gastric carcinogenesis. Furthermore, DNMT1 may play a significant role in the development of poorly differentiated gastric cancers by inducing frequent DNA hypermethylation of multiple CpG islands.


Subject(s)
CpG Islands , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methylation , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Animals , Cadherins/biosynthesis , Cell Transformation, Neoplastic/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , Epstein-Barr Virus Infections/metabolism , Female , Gastric Mucins/biosynthesis , Humans , Immunohistochemistry , In Situ Hybridization , Male , Middle Aged , Mucin-2 , Mucins/biosynthesis , Polymerase Chain Reaction , Proliferating Cell Nuclear Antigen/biosynthesis , Stomach Neoplasms/virology
18.
Tumour Biol ; 24(3): 109-15, 2003.
Article in English | MEDLINE | ID: mdl-14610314

ABSTRACT

Colorectal cancer is a major cause of death in Europe and the USA, and much effort is therefore devoted to improve its early detection. In this article, we report the abnormal expression of gastric mucin in aberrant crypt foci (ACF) that appear in the colon mucosae removed from colorectal cancer patients and rats treated with methyl-N'-nitro-N-nitroso-guanidine (MNNG). We performed the immunoperoxidase test using monoclonal antibodies raised against gastric M1 mucin encoded by the MUC5AC gene and against rat gastric mucins (MAb 660), respectively. In both human and rat colon, these anti-gastric mucin MAbs stained specifically goblet cells within ACF. In humans, the M1/MUC5AC mucin was expressed in the upper part of the glands in hyperplastic ACF and in the typical ACF. In addition, the anti-gastric mucin MAbs stained some rare, scattered, histologically normal glands in the human and rat colon mucosae. These glands may be regarded as precursors of ACF. The abnormal expression of the MUC5AC gene constitutes a novel change in addition to genetic modifications already observed in ACF, and supports our previous findings demonstrating the potential of this gastric mucin as an early marker of human and rat colon carcinogenesis.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gastric Mucins/biosynthesis , Gastric Mucosa/metabolism , Precancerous Conditions/metabolism , Animals , Antibodies, Monoclonal/metabolism , Carcinogens , Cell Transformation, Neoplastic , Colon/metabolism , Colon/pathology , Humans , Immunohistochemistry , Methylnitronitrosoguanidine , Rats
19.
Hepatogastroenterology ; 50(53): 1739-42, 2003.
Article in English | MEDLINE | ID: mdl-14571831

ABSTRACT

BACKGROUND/AIMS: Alterations of gastric mucin have been postulated as important pathogenic properties of Helicobacter pylori. In this study, we investigated gastric mucin synthesis in H. pylori-infected gastric mucosa by measuring UDP-galactosyltransferase activity, a key enzyme for the synthesis of mucin, and the amount of intracellular mucin in the gastric mucosa. METHODOLOGY: Gastric biopsy specimens were obtained from thirty-seven patients (20 H. pylori-positive and 17 H. pylori-negative). UDP-galactosyltransferase activity of the biopsy specimens was measured by an assay system we had developed, using a peanut agglutinin lectin. The amount of intracellular mucin in the gastric epithelial cells was analyzed by measuring the cells' periodic acid-Schiff-alcian blue staining-positive substances. RESULTS: UDP-galactosyltransferase activities in the antral mucosa, but not in the body mucosa, of H. pylori-positive patients were significantly lower than those of H. pylori-negative patients (p < 0.05). The amount of intracellular mucin in antral epithelial cells of H. pylori-positive patients was significantly lower than that of H. pylori-negative patients (p < 0.01). CONCLUSIONS: These findings suggest that H. pylori infection decreases gastric mucin synthesis via inhibition of UDP-galactosyltransferase. This effect may impair the gastric mucosal barrier and contribute to the mucosal injury induced by H. pylori infection.


Subject(s)
Galactosyltransferases/metabolism , Gastric Mucins/biosynthesis , Helicobacter Infections/enzymology , Helicobacter pylori , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged
20.
J Gastroenterol ; 38(8): 727-33, 2003.
Article in English | MEDLINE | ID: mdl-14505125

ABSTRACT

BACKGROUND: Epidermal growth factor (EGF) has an anti-ulcer effect, but the mechanisms of this gastric mucosal protection are incompletely understood. We have suggested the importance of mucin as a mucosal protectant. We investigated whether increased mucin biosynthesis might be involved in the gastric mucosal protection conferred by EGF. METHODS: EGF and then ethanol were added to primary monolayer cultures of guinea pig gastric mucous cells, in which factors such as gastric acid and gastrointestinal hormones were excluded. Mucin and prostaglandin E(2) (PGE(2)) were assayed. RESULTS: Cytoprotection induced by EGF was demonstrated. Mucin biosynthesis and PGE(2) release were both significantly increased by EGF. When endogenous PGE(2) synthesis was inhibited by pretreatment with 10(-5) M or 10(-4) M indomethacin (IND), mucin biosynthesis was still significantly increased by EGF. Ethanol-induced cell damage was concentration-dependent in cultures with no other additions (normal PGE(2) and mucin biosynthesis). Damage by ethanol was decreased by EGF pretreatment (increased PGE(2) and mucin biosynthesis). Damage by ethanol was increased by 10(-5) M IND pretreatment (decreased PGE(2); normal mucin biosynthesis) and by 10(-4) M IND pretreatment (decreased PGE(2) and mucin biosynthesis). Ethanol-induced damage was decreased by EGF pretreatment even in the presence of 10(-5) M and 10(-4) M IND (decreased PGE(2); increased mucin biosynthesis). CONCLUSIONS: Increased mucin biosynthesis, induced by EGF independently of PGE(2), protects gastric mucous cells.


Subject(s)
Cytoprotection/drug effects , Dinoprostone/metabolism , Epidermal Growth Factor/pharmacology , Gastric Mucins/biosynthesis , Gastric Mucosa/cytology , Gastric Mucosa/drug effects , Animals , Cell Culture Techniques , Cell Survival/drug effects , Ethanol/pharmacology , Gastric Mucosa/metabolism , Guinea Pigs , Male
SELECTION OF CITATIONS
SEARCH DETAIL