Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Anat Rec (Hoboken) ; 301(12): 2128-2144, 2018 12.
Article in English | MEDLINE | ID: mdl-30382612

ABSTRACT

The fundic glands of the stomach contain two types of mucous cells: surface mucous cells (SMCs) located at the surface of the stomach and the pits, and mucous neck cells (MNCs) situated in the neck of the glands. They produce mucins, highly glycosylated proteins. Very little is known about the glycan composition of these mucins and of gastric secretion in general. We used several lectins combined with deglycosylation pretreatments to analyze the glycan composition of SMCs and MNCs. The results showed the presence of terminal sialic acid and subterminal Gal and GalNAc, which is consistent with previous knowledge about glycosylation in mucins. Our results also support previous reports that showed a different expression of mucins in the SMCs, depending on their superficial or deep location in the pit. Some lectins labeled only the perinuclear region of the SMCs, but not the apical region, where the secretory granules are stored. This suggests that the lectins are labeling sugar residues that are accessible to lectins during the first steps of glycan synthesis, which occurs in the endoplasmic reticulum and Golgi apparatus. Our results indicate that SMCs and MNCs produce a mucus secretion with a different glycoconjugate composition. The secretion is more varied in SMCs. As our results coincide with what we know about glycosylation of mucins, we can conclude that most of the glycans detected belong to mucins, and the differences in glycosylation observed in each cell type may be due, mainly, to the different secreted mucins. Anat Rec, 301:2128-2144, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Gastric Fundus/cytology , Gastric Fundus/metabolism , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Glycoconjugates/metabolism , Mucus/metabolism , Animals , Gastric Fundus/chemistry , Gastric Mucosa/chemistry , Glycoconjugates/analysis , Male , Mucins/analysis , Mucins/metabolism , Mucus/chemistry , Rats , Rats, Sprague-Dawley
2.
World J Gastroenterol ; 21(26): 8208-14, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26185396

ABSTRACT

Recently, a new disease entity termed gastric adenocarcinoma of fundic gland type (GA-FG) was proposed. We treated five cases of GA-FG with endoscopic submucosal dissection. All tumors were small and located in the upper third of the stomach. Four tumors were macroscopically identified as 0-IIa and one was identified as 0-IIb. Narrow-band imaging with magnifying endoscopy showed an irregular microvascular pattern in 2 cases and a regular microvascular pattern in the remainder. All tumors arose from the deep layer of the lamina propria mucosae and showed submucosal invasion. Lymphatic invasion was seen only in one case, while no venous invasion was recognized. All tumors were positive for pepsinogen-I  and MUC6 by immunohistochemistry. None showed p53 overexpression, and the labeling index of Ki-67 was low in all cases. All cases have been free from recurrence or metastasis. Herein, we discussed the clinicopathological features of GA-FG in comparison with past reports.


Subject(s)
Adenocarcinoma/surgery , Gastrectomy/methods , Gastric Fundus/surgery , Gastroscopy/methods , Stomach Neoplasms/surgery , Adenocarcinoma/chemistry , Adenocarcinoma/pathology , Aged , Biomarkers, Tumor/analysis , Biopsy , Female , Gastric Fundus/chemistry , Gastric Fundus/pathology , Humans , Immunohistochemistry , Male , Narrow Band Imaging , Neoplasm Staging , Stomach Neoplasms/chemistry , Stomach Neoplasms/pathology , Treatment Outcome
3.
World J Gastroenterol ; 20(47): 18044-7, 2014 Dec 21.
Article in English | MEDLINE | ID: mdl-25548505

ABSTRACT

Fundic gland polyps (FGPs) are currently the most common type of gastric polyps and are usually benign. However, although rare, gastric adenocarcinoma of FGP has been recently proposed as a new variant of gastric adenocarcinoma. Here we report the first case of a 49-year-old woman with focal signet ring cell carcinoma that arose from an FGP of the stomach. The tumor was completely excised by endoscopic snare polypectomy. FGPs should therefore be evaluated for malignant changes although they occur rarely, if the FGP has an erosive or irregular surface.


Subject(s)
Carcinoma, Signet Ring Cell/pathology , Gastric Fundus/pathology , Polyps/pathology , Stomach Diseases/pathology , Stomach Neoplasms/pathology , Biomarkers, Tumor/analysis , Biopsy , Carcinoma, Signet Ring Cell/chemistry , Carcinoma, Signet Ring Cell/surgery , Female , Gastrectomy/methods , Gastric Fundus/chemistry , Gastric Fundus/surgery , Gastroscopy/methods , Humans , Immunohistochemistry , Middle Aged , Polyps/chemistry , Polyps/surgery , Stomach Diseases/metabolism , Stomach Diseases/surgery , Stomach Neoplasms/chemistry , Stomach Neoplasms/surgery
4.
Food Funct ; 3(10): 1025-34, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22790379

ABSTRACT

There is emerging interest in the impact of food structure on lipid digestion and its relationship to human nutrition. The objective of this study was to investigate the influence of heteroaggregation of lipid droplets on their potential biological fate using a simulated gastrointestinal tract (GIT). At neutral pH, a highly viscous "mixed emulsion" was formed by mixing anionic ß-lactogobulin (ß-Lg) coated lipid droplets with cationic lactoferrin (LF) coated lipid droplets due to electrostatic attraction. We compared the behavior of ß-Lg-emulsions, LF-emulsions and mixed emulsions under in vitro oral, gastric, and small intestinal conditions. In the oral stage, the ß-Lg emulsion and mixed emulsion were stable but the LF emulsion aggregated, which was attributed to electrostatic interactions with mucin. In the gastric stage, extensive droplet aggregation occurred in all three emulsions, which was attributed to proteolysis of adsorbed proteins by pepsin, as well as the influence of high acidity and ionic strength on electrostatic interactions. Despite the differences in the initial compositions and microstructures of the three emulsions, we did not observe an appreciable difference in the rate or extent of their lipid digestion in the small intestine. Qualitatively similar results were obtained using a simple GIT model (small intestine only) and the full GIT model (oral, gastric, and small intestine). The knowledge gained from this study will be useful for the creation of functional foods to improve health and well-being.


Subject(s)
Digestion , Gastrointestinal Tract/metabolism , Lipid Metabolism , Static Electricity , Adsorption , Emulsions/chemistry , Gastric Fundus/chemistry , Humans , Hydrogen-Ion Concentration , Lactoferrin/metabolism , Lipids/chemistry , Models, Biological , Osmolar Concentration , Particle Size , Proteins/metabolism , Rheology , Saliva, Artificial/chemistry
5.
Am J Surg Pathol ; 36(7): 1030-5, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22472957

ABSTRACT

Gastric adenocarcinoma with chief cell differentiation (GA-CCD) has been reported as a new, rare variant of gastric adenocarcinoma. Only 12 cases in Japanese patients have been described to date, but they demonstrate distinct clinicopathologic features. To further characterize these lesions, we have collected 10 additional cases. Patients ranged in age from 44 to 79 years (mean, 64.2 y) with a relatively equal sex distribution (6 women and 4 men). Stratified by race, 4 patients were Hispanic, 2 were White, 2 were African American, 1 was Asian (Chinese), and the race was unknown for 1 patient. All patients presented with gastroesophageal reflux that prompted an endoscopic examination. The majority of GA-CCDs were identified in the fundus (7 of 10, 70%) and the remaining in the cardia (n=3). Grossly, they were solitary and polypoid, ranging in size from 0.2 to 0.8 cm (mean, 0.4 cm). Histologically, all cases were centered in the deep mucosa, with focal involvement of surface foveolar epithelium in 3 (30%) cases but not the submucosa. The tumors consisted of clustered glands and irregular branching cords of oxyntic epithelium. Thin wisps of radiating smooth muscle separated the epithelium, but desmoplasia was distinctly absent in all cases. The oxyntic mucosa was 1 to 2 cells thick and composed of a mixture of mucous neck, parietal, and chief cells. In 7 of 10 (70%) cases, chief cells were the predominant cell type, whereas the remaining 3 cases consisted primarily of mucous neck cells. The nuclei were mildly enlarged with slight nuclear pleomorphism, but no mitotic figures were identified. In addition, necrosis, lymphovascular invasion, and perineural invasion were absent. Immunohistochemically, GA-CCDs were diffusely positive for MUC6 (10 of 10, 100%) and negative for MUC5AC (0%) and MUC2 (0%). Ki-67 immunolabeling demonstrated variable expression, with the highest areas ranging from 0.2% to 10%. Clinical follow-up was available for 9 of 10 (90%) patients and ranged from 6 to 39 months. One patient had persistence of lesion at 6 months because of incomplete removal, whereas the other 8 were disease free. In summary, GA-CCDs are solitary, mucosal lesions of the gastric cardia/fundus that arise in patients from multiple ethnic backgrounds. Considering that patients within this study and those reported previously have had neither true recurrence nor progression of disease, these lesions are best regarded as benign. Consequently, the term GA-CCD is contradictory and we prefer the descriptive term "oxyntic gland polyp/adenoma" until further studies can clarify the pathogenesis of these lesions and their natural history.


Subject(s)
Adenocarcinoma/classification , Adenoma/classification , Cardia/pathology , Cell Proliferation , Chief Cells, Gastric/pathology , Gastric Fundus/pathology , Parietal Cells, Gastric/pathology , Polyps/classification , Stomach Neoplasms/classification , Terminology as Topic , Adenocarcinoma/chemistry , Adenocarcinoma/ethnology , Adenocarcinoma/pathology , Adenoma/chemistry , Adenoma/ethnology , Adenoma/pathology , Adult , Aged , Baltimore , Biomarkers, Tumor/analysis , Cardia/chemistry , Chief Cells, Gastric/chemistry , Female , Gastric Fundus/chemistry , Humans , Immunohistochemistry , Male , Middle Aged , Nebraska , Parietal Cells, Gastric/chemistry , Polyps/chemistry , Polyps/ethnology , Polyps/pathology , Stomach Neoplasms/chemistry , Stomach Neoplasms/ethnology , Stomach Neoplasms/pathology
6.
Zhongguo Dang Dai Er Ke Za Zhi ; 12(7): 563-8, 2010 Jul.
Article in Chinese | MEDLINE | ID: mdl-20637157

ABSTRACT

OBJECTIVE: To study the relationship between Ghrelin and growth hormone secretagogue receptor (GHSR) expression and the catch-up growth in rats with intrauterine growth restriction (IUGR). METHODS: The rat model of IUGR was established by food restriction during pregnancy. The small for gestational age (SGA) and appropriate for gestational age (AGA) rat pups from the pregnant rats were used as the experimental group. The AGA rat pups from the pregnant rats without food restriction served as the control group. The samples from the stomach fundus and hypothalamus were taken postnatal days 0, 20 and 40. Ghrelin mRNA and GHSR mRNA expression were determined by real-time fluorescence quantitative PCR (real-time FQ-PCR). Ghrelin protein and GHSR protein expression were examined by immunohistochemistry (IHC). RESULTS: At postnatal day 0, both Gherlin mRNA and protein levels in the stomach fundus were significantly higher, while GHSR mRNA expression in the hypothalamus were significantly lower in SGA rats from food restriction group than those in AGA rats from restriction and control groups. At postnatal day 20, the ghrelin protein expression in the stomach of fundus, and GHSR mRNA and protein expression in the hypothalamus in SGA catch-up rats were significantly higher than those in SGA non-catch-up growth rats and AGA rats from the control group. At postnatal day 40, there were no significant differences among SGA catch-up growth rats, SGA non-catch-up growth rats and normal AGA rats. CONCLUSIONS: Ghrelin-GHSR might be involved in the physiological regulation and pathological process in IUGR rats. It is also possibly involved in the regulation of catch-up growth in the early life of SGA rats.


Subject(s)
Fetal Growth Retardation/physiopathology , Ghrelin/genetics , Receptors, Ghrelin/genetics , Animals , Female , Gastric Fundus/chemistry , Ghrelin/analysis , Ghrelin/physiology , Growth , Hypothalamus/chemistry , Immunohistochemistry , Pregnancy , Rats , Receptors, Ghrelin/analysis
7.
Am J Physiol Gastrointest Liver Physiol ; 296(6): G1370-81, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19372102

ABSTRACT

Populations of interstitial cells of Cajal (ICC) are altered in several gastrointestinal neuromuscular disorders. ICC are identified typically by ultrastructure and expression of Kit (CD117), a protein that is also expressed on mast cells. No other molecular marker currently exists to independently identify ICC. The expression of ANO1 (DOG1, TMEM16A), a Ca(2+)-activated Cl(-) channel, in gastrointestinal stromal tumors suggests it may be useful as an ICC marker. The aims of this study were therefore to determine the distribution of Ano1 immunoreactivity compared with Kit and to establish whether Ano1 is a reliable marker for human and mouse ICC. Expression of Ano1 in human and mouse stomach, small intestine, and colon was investigated by immunofluorescence labeling using antibodies to Ano1 alone and in combination with antibodies to Kit. Colocalization of immunoreactivity was demonstrated by epifluorescence and confocal microscopy. In the muscularis propria, Ano1 immunoreactivity was restricted to cells with the morphology and distribution of ICC. All Ano1-positive cells in the muscularis propria were also Kit positive. Kit-expressing mast cells were not Ano1 positive. Some non-ICC in the mucosa and submucosa of human tissues were Ano1 positive but Kit negative. A few (3.2%) Ano1-positive cells in the human gastric muscularis propria were labeled weakly for Kit. Ano1 labels all classes of ICC and represents a highly specific marker for studying the distribution of ICC in mouse and human tissues with an advantage over Kit since it does not label mast cells.


Subject(s)
Chloride Channels/analysis , Gastrointestinal Tract/chemistry , Gastrointestinal Tract/cytology , Membrane Proteins/analysis , Neoplasm Proteins/analysis , Adult , Animals , Anoctamin-1 , Antigens, Surface/analysis , Colon/chemistry , Colon/cytology , Female , Gastric Fundus/chemistry , Gastric Fundus/cytology , Gastric Mucosa/chemistry , Gastric Mucosa/cytology , Humans , Intestinal Mucosa/chemistry , Intestinal Mucosa/cytology , Intestine, Small/chemistry , Intestine, Small/cytology , Jejunum/chemistry , Jejunum/cytology , Male , Mast Cells/chemistry , Mast Cells/cytology , Mice , Mice, Inbred BALB C , Middle Aged , Proto-Oncogene Proteins c-kit/analysis , Pyloric Antrum/chemistry , Pyloric Antrum/cytology , Stomach/chemistry , Stomach/cytology , Tryptases/analysis
8.
Eur J Clin Invest ; 38(6): 397-403, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18422979

ABSTRACT

BACKGROUND: Conflicting data concerning the involvement of ghrelin in the pathophysiology of alcohol dependence have been reported. The aim of this study is to investigate how chronic alcohol ingestion influences plasma ghrelin levels and whether potential changes observed in plasma relate to modifications in ghrelin production in the stomach where this peptide is primarily synthesized. MATERIALS AND METHODS: Fifty-one consecutive alcoholics admitted for alcohol withdrawal were prospectively enrolled and compared to a control group of 32 healthy volunteers matched for age, sex, height and weight. All subjects underwent fasting plasma ghrelin determination. Twenty-seven randomly selected alcoholics and 17 controls underwent gastroscopy for fundic and duodenal biopsies. Tissues were fixed for histology or frozen in liquid nitrogen for ghrelin protein and mRNA determinations by a radioimmunoassay and quantitative polymerase chain reaction, respectively. Alcohol consumption was normalized to body weight (BW) or body mass index (BMI) given the influence of BW and volume distribution on alcohol levels. RESULTS: Plasma and fundic ghrelin protein levels were significantly decreased in alcoholics. Fundic but not plasma ghrelin protein levels inversely correlated with alcohol consumption normalized to BW or BMI. Ghrelin mRNA levels in fundic biopsies were similar in alcoholics and controls. No significant differences in duodenal ghrelin protein and mRNA levels were found between both groups. CONCLUSIONS: Alcoholism was associated with decreased plasma ghrelin levels partly due to reduced ghrelin production in the stomach. Alcohol affected ghrelin production on the post-transcriptional level in the fundus, whereas duodenal ghrelin secretion did not respond in a similar manner to alcohol intake.


Subject(s)
Alcoholism/metabolism , Gastric Fundus/chemistry , Ghrelin/analysis , Adult , Aged , Alcoholism/blood , Appetite Regulation , Body Mass Index , Body Weight , Case-Control Studies , Chronic Disease , Duodenum , Female , Ghrelin/blood , Ghrelin/genetics , Humans , Intestinal Mucosa/chemistry , Male , Middle Aged , RNA, Messenger/analysis
9.
Mod Pathol ; 20(9): 974-89, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17643099

ABSTRACT

Pathogenic mechanisms associated with Helicobacter pylori infection enhance susceptibility of the gastric epithelium to carcinogenic conversion. We have characterized the gene expression profiles of gastric biopsies from 69 French Caucasian patients, of which 43 (62%) were infected with H. pylori. The bacterium was detected in 27 of the 42 antral biopsies examined and in 16 of the 27 fundic biopsies. Infected biopsies were selected for the presence of chronic active gastritis, in absence of metaplasia and dysplasia of the gastric mucosa. Infected antral and fundic biopsies exhibited distinct transcriptional responses. Altered responses were linked with: (1) the extent of polymorphonuclear leukocyte infiltration, (2) bacterial density, and (3) the presence of the virulence factors vacA, babA2, and cagA. Robust modulation of transcripts associated with Toll-like receptors, signal transduction, the immune response, apoptosis, and the cell cycle was consistent with expected responses to Gram-negative bacterial infection. Altered expression of interferon-regulated genes (IFITM1, IRF4, STAT6), indicative of major histocompatibility complex (MHC) II-mediated and Th1-specific responses, as well as altered expression of GATA6, have previously been described in precancerous states. Upregulation of genes abundantly expressed in cancer tissues (UBD, CXCL13, LY96, MAPK8, MMP7, RANKL, CCL18) or in stem cells (IFITM1 and WFDC2) may reveal a molecular switch towards a premalignant state in infected tissues. Tissue microarray analysis of a large number of biopsies, which were either positive or negative for the cag-A virulence factor, when compared to each other and to noninfected controls, confirmed observed gene alterations at the protein level, for eight key transcripts. This study provides 'proof-of-principle' data for identifying molecular mechanisms driving H. pylori-associated carcinogenesis before morphological evidence of changes along the neoplastic progression pathway.


Subject(s)
Gastric Fundus/microbiology , Gastric Mucosa/microbiology , Gene Expression Profiling/methods , Helicobacter Infections/genetics , Helicobacter pylori/isolation & purification , Pyloric Antrum/microbiology , Stomach Neoplasms/microbiology , Transcription, Genetic , Adult , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Case-Control Studies , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , France , Gastric Fundus/chemistry , Gastric Fundus/pathology , Gastric Mucosa/chemistry , Gastric Mucosa/pathology , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Neoplastic , Genotype , Helicobacter Infections/complications , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Humans , Immunity, Mucosal/genetics , Inflammation/genetics , Neutrophil Infiltration , Oligonucleotide Array Sequence Analysis , Peptide Hydrolases/genetics , Phenotype , Pyloric Antrum/chemistry , Pyloric Antrum/pathology , RNA, Bacterial/analysis , RNA, Messenger/analysis , Receptors, Cell Surface/genetics , Signal Transduction/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tissue Array Analysis
10.
Dig Dis Sci ; 52(5): 1345-54, 2007 May.
Article in English | MEDLINE | ID: mdl-17373588

ABSTRACT

Our objective was to systematically review the existing literature regarding the use of cytokeratin (CK) stain in differentiating Barrett's esophagus (BE) from tissues of the gastric cardia, corpus, or antrum, with or without intestinal metaplasia (IM). Pubmed was searched for full publications in English (1983-2005) addressing the use of CK for differentiation of BE from contiguous tissues. Information was collected on the study sample, blinding, the methods used for CK staining, and for defining and applying the gold standard tests. Test characteristics were obtained or calculated. Sixteen studies (containing 46 comparisons) met the inclusion and exclusion criteria. Immunostaining for CK 7 and 20 was generally highly specific in distinguishing long-segment BE from antrum IM, fundus IM, or noncardiac gastric IM; 27 comparisons showed statistically significant differences. However, only 8 of 15 comparisons (6 of 12 studies) reported significant differences in CK staining patterns between BE and gastric cardia IM with a high sensitivity (89%-100%) and specificity (83%-100%) for long-segment BE and lower estimates for short-segment BE, while the other seven comparisons showed no significant differences and a very low sensitivity. Examination by a blinded pathologist was reported in five of six positive studies and in only one of six of the negative studies. In addition, variation in the patient populations, use of surgical resection versus endoscopic biopsies, and biopsy sampling technique in endoscopic studies may have accounted for these differences. Finally, two studies did not find significant differences in CK staining patterns between BE and normal cardiac mucosa. In conclusions, CK immunostaining has not performed well in differentiating BE, especially short-segment BE, from cardia IM. There seems to be a spectrum bias where the accuracy varies with different tested populations. CK immunostaining distinguished well between BE and IM in noncardiac segments of the stomach; however, these comparisons are not clinically relevant.


Subject(s)
Barrett Esophagus/diagnosis , Immunohistochemistry , Keratins/analysis , Stomach Diseases/diagnosis , Stomach/chemistry , Barrett Esophagus/metabolism , Biomarkers/analysis , Cardia/chemistry , Cardia/pathology , Diagnosis, Differential , Gastric Fundus/chemistry , Gastric Fundus/pathology , Humans , Immunohistochemistry/methods , Keratin-20/analysis , Keratin-7/analysis , Metaplasia , Predictive Value of Tests , Pyloric Antrum/chemistry , Pyloric Antrum/pathology , Sensitivity and Specificity , Stomach/pathology , Stomach Diseases/metabolism , Stomach Diseases/pathology
11.
Pathol Int ; 55(4): 170-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15826243

ABSTRACT

Intestinal metaplasia (IM) in the human stomach has previously been classified into a gastric and intestinal mixed (GI-IM) and a solely intestinal phenotype (I-IM). The phenotypes of mucous and endocrine cells were evaluated in 3034 glandular ducts associated with chronic gastritis. In the pyloric region, the relative expression of gastric endocrine cell markers, such as gastrin and somatostatin, decreased gradually from glandular ducts with only gastric mucous cell phenotype (G type) to GI-IM toward I-IM, while that of the intestinal endocrine cell markers, glicentin, gastric inhibitory polypeptide (GIP), and glucagon-like peptide-1 (GLP-1) was inversely correlated. In the fundic region, gastrin-positive cells emerged in the pseudo-pyloric and GI-IM glands, whereas I-IM glands did not possess any gastrin-positive cells, suggesting the presence of a distinct pathway of intestinalization. Double staining revealed coexistence of gastrin- and GLP-1-positive cells in the same gland and occasionally in the same cell in GI-IM glands. These results suggest that the phenotypes of endocrine cells are in line with those for mucous counterparts and support the concept that all of the different types of mucous and endocrine cells in normal and IM glands might be derived from a single progenitor cell in each gland.


Subject(s)
Endocrine Glands/pathology , Gastric Mucosa/pathology , Intestines/pathology , Stomach Neoplasms/pathology , Aged , Chromogranin A , Chromogranins/analysis , Endocrine Glands/chemistry , Female , Gastric Fundus/chemistry , Gastric Fundus/pathology , Gastric Inhibitory Polypeptide/analysis , Gastric Mucosa/chemistry , Gastrins/analysis , Glicentin , Glucagon/analysis , Glucagon-Like Peptide 1 , Glucagon-Like Peptides , Humans , Immunohistochemistry , Intestines/chemistry , Male , Metaplasia , Middle Aged , Peptide Fragments/analysis , Protein Precursors/analysis , Pyloric Antrum/chemistry , Pyloric Antrum/pathology , Somatostatin/analysis , Stomach Neoplasms/metabolism
12.
J Smooth Muscle Res ; 41(1): 35-47, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15855738

ABSTRACT

The contractile response of the stomach fundus to endothelin-1 (ET-1) was examined in streptozotocin (STZ)-induced diabetic rats. In STZ-diabetic rats (versus age-matched control rats) (a) ET-1 caused a longer-lasting contraction of stomach fundus strips, and (b) in the dose-response curve, the ET-1-induced contraction was significantly greater for a given concentration (3 x 10(-7) to 10(-7) M). Although repeated application of ET-1 led to desensitization, the desensitization was less pronounced in STZ-diabetic rats than in the controls. The density of the binding sites for [(125)I]-ET-1 was increased in the diabetic stomach fundus (versus the controls), but Kd values were similar between the two groups. The ET(B) receptor mRNA expression level was significantly increased in the diabetic stomach fundus. These results suggest that the diabetes-related enhancement of the ET-1-induced contraction of the stomach fundus may be due to an increase in the ET(B) receptor population.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Endothelin-1/pharmacology , Gastric Fundus/physiopathology , Muscle Contraction/drug effects , Animals , Gastric Fundus/chemistry , In Vitro Techniques , Male , Muscle, Smooth/drug effects , Muscle, Smooth/physiopathology , Rats , Rats, Wistar , Receptor, Endothelin A/analysis , Receptor, Endothelin B/analysis , Streptozocin
13.
Pathol Res Pract ; 200(11-12): 817-21, 2005.
Article in English | MEDLINE | ID: mdl-15792126

ABSTRACT

A case of an unusual variant of fundic gland polyp (FGP) composed of chief cell hyperplasia with structural and nuclear atypia in an 87-year-old woman is presented. Gastrointestinal endoscopy revealed a sessile polyp in the cardia/ corpus transition zone and a polypoid lesion in the fundus. Histologically, the polyp in the cardia/corpus showed a typical appearance of FGP, while that in fundus demonstrated a tumorous lesion composed of irregular branched tubules with nuclear stratification. Despite the structural distortion and nuclear atypia, mitotic figures were absent and MIB-1 positive cells were less than 3%. Immunohistochemically, the cytoplasms of the tubules were negative for gastric mucin and Muc-5AC glycoprotein, but mostly positive for pepsinogen-I, indicating that the proliferated glands consisted mainly of chief cells, not mucous cells. Parietal cells were occasionally found in the glands. At the periphery of the lesion, microcysts composed of parietal cells, chief cells, and mucous cells had developed. Altogether, the polyp in the fundus was diagnosed as an unusual variant of FGP with chief cell hyperplasia. This FGP should be differentiated from tubular adenocarcinoma. Proliferation of chief cells with occasional parietal cells is critical for the differential diagnosis.


Subject(s)
Gastric Fundus/pathology , Polyps/pathology , Stomach Neoplasms/pathology , Adenocarcinoma/diagnosis , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Cell Count , Cell Nucleus/pathology , Cell Proliferation , Diagnosis, Differential , Female , Gastric Fundus/chemistry , Gastric Mucosa/chemistry , Gastric Mucosa/pathology , Humans , Hyperplasia , Ki-67 Antigen/analysis , Pepsinogen A/analysis , Polyps/chemistry , Stomach Neoplasms/chemistry
14.
J Anat ; 205(3): 239-46, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15379929

ABSTRACT

Ghrelin is a newly identified gastric peptide hormone that has various important functions, including growth-hormone release and appetite stimulation. Ghrelin-immunoreactive cells (ghrelin cells) are characterized by X-type endocrine cells in the rat stomach. In the present study, we analysed ghrelin cells in fundi of stomach from ICR mice and Syrian hamsters immunohistochemically, immunoelectron microscopically and morphometrically, and compared the results with those from Wistar rats. Immunohistochemistry revealed that ghrelin cells were sparsely distributed in the proper gastric glands in all species. The number of ghrelin cells per unit area in hamsters was significantly lower than that in rats. Immunoelectron microscopy detected ghrelin immunolabelling in granules in the X-type endocrine cells. However, the diameter of granules in the hamsters was significantly smaller than that in the mice and rats. Gastric ghrelin contents were determined by radioimmunoassay, and levels in the hamsters were significantly lower than those in mice and rats. The results from mice were identical to those from rats. In conclusion, gastric ghrelin cells in mice and hamsters are characterized by X-type endocrine cells, as has been observed in rats. However, the data indicated that gastric ghrelin production was lower in hamster than in mouse or rat.


Subject(s)
Gastric Fundus/chemistry , Gastric Fundus/cytology , Peptide Hormones/analysis , Animals , Cricetinae , Ghrelin , Male , Mesocricetus , Mice , Mice, Inbred ICR , Microscopy, Immunoelectron , Radioimmunoassay/methods , Rats , Rats, Wistar , Species Specificity
15.
Neurogastroenterol Motil ; 15(6): 655-62, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14651601

ABSTRACT

The major functions of the stomach are under the control of the enteric nervous system (ENS), but the neuronal circuits involved in this control are largely unknown in humans. Enteric neurones can be characterized by their neuromediator or marker content, i.e. by neurochemical coding. The purpose of this study was to characterize the presence and co-localization of neurotransmitters in myenteric neurones of the human gastric fundus. Choline acetyltransferase (ChAT), neurone-specific enolase (NSE), vasoactive intestinal polypeptide (VIP), nitric oxide synthase (NOS), substance P (SP) were detected by immunohistochemical methods in whole mounts of gastric fundus myenteric plexus (seven patients). Antibodies against ChAT and NOS labelled the majority of myenteric neurones identified by NSE (57.2 +/- 5.6% and 40.8 +/- 4.5%, respectively; mean +/- SD). The proportions of VIP- and SP-immunoreactive neurones were significantly smaller, constituting 19.6 +/- 6.9% and 16.0 +/- 3.7%, respectively. Co-localization studies revealed five major populations representing over 75% of the myenteric neurones: ChAT/-, 30.1 +/- 6.1%; NOS/-, 24.2 +/- 4.4%; ChAT/SP/-, 8.3 +/- 3.1%; NOS/VIP/-, 7.2 +/- 6.0%; ChAT/VIP/-, 4.9 +/- 2.6. Some similarities are apparent in the neurochemical coding of myenteric neurones in the stomach and intestine of humans, and between the stomach of humans and animals, but striking differences exist. The precise functional role of the neurochemically identified classes of neurones remains to be determined.


Subject(s)
Gastric Fundus/chemistry , Gastric Fundus/metabolism , Myenteric Plexus/chemistry , Myenteric Plexus/metabolism , Aged , Analysis of Variance , Female , Humans , In Vitro Techniques , Male , Neurons/chemistry , Neurons/metabolism
16.
Am J Gastroenterol ; 97(10): 2514-23, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12385432

ABSTRACT

OBJECTIVE: The normal histology at the gastroesophageal junction, and in particular the nature of cardiac mucosa, remains in dispute. Likewise, the relationship of intestinal metaplasia at the gastroesophageal junction (CIM) to Barrett's and intestinal metaplasia of the stomach (GIM) is unclear. The aim of this study was to assess the immunostaining characteristics of cardiac mucosa and CIM and compare their staining pattern with that of other foregut mucosal types. We hypothesized that the immunostaining patterns of these foregut tissues would provide insight into the nature and etiology of cardiac mucosa and CIM. METHODS: Paraffin-embedded biopsy specimens from 50 patients with normal antral or fundic mucosa, cardiac mucosa, squamous mucosa, CIM, GIM, or Barrett's were obtained and immunostained with a panel of monoclonal antibodies including those for cytokeratins 7 and 20 (CK7/CK20) and DAS-1. RESULTS: Biopsies from normal gastric antral and fundic mucosa and squamous esophageal mucosa all showed a non-Barrett's type CK7/CK20 immunostaining pattern, whereas in 85% of patients, cardiac mucosa had a Barrett's type CK7/CK20 pattern (p < 0.001). A Barrett's type CK7/ CK20 staining pattern was seen in 100% of Barrett's, 78% of CIM, and 0% of GIM patients. Likewise, DAS-1 staining was similar in patients with CIM and Barrett's and significantly different in patients with GIM. CONCLUSIONS: Cytokeratin immunostaining of cardiac mucosa demonstrates significant differences from recognized normal gastric and esophageal mucosa but a similarity to Barrett's. This suggests that cardiac mucosa, like Barrett's, may be acquired. Likewise, immunostaining similarities between CIM and Barrett's biopsies point to the possibility of a reflux etiology for CIM in some patients.


Subject(s)
Antibodies/analysis , Barrett Esophagus/metabolism , Cardia/metabolism , Esophagogastric Junction/metabolism , Intermediate Filament Proteins/analysis , Keratins/analysis , Barrett Esophagus/complications , Barrett Esophagus/pathology , Biopsy , Cardia/pathology , Esophagogastric Junction/pathology , Gastric Fundus/chemistry , Gastric Fundus/pathology , Helicobacter Infections/complications , Helicobacter Infections/pathology , Helicobacter pylori , Humans , Immunohistochemistry , Keratin-20 , Keratin-7 , Metaplasia , Mucous Membrane/metabolism , Mucous Membrane/pathology , Pyloric Antrum/chemistry , Pyloric Antrum/pathology
17.
Gut ; 51(5): 628-33, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12377798

ABSTRACT

BACKGROUND: Sonic hedgehog (Shh) is an important endodermal morphogenetic signal during the development of the vertebrate gut. It controls gastrointestinal patterning in general, and gastric gland formation in particular. We have previously shown that Shh regulates gastric gland proliferation in the adult but detailed analysis of its expression along the adult gastrointestinal tract has never been undertaken. We therefore studied Shh expression along the normal human and rodent adult gastrointestinal tract as well as in intestinal metaplasia of the stomach, gastric and intestinal metaplasia of the oesophagus, and gastric heterotopia in Meckel's diverticulum. METHODS: The studies were performed with in situ hybridisation and by immunohistochemistry using an antibody that recognises the Shh precursor form. RESULTS: We found that in the normal gastrointestinal tract, high levels of Shh were expressed in the fundic glands of the stomach. Shh expression was also found in fundic gland metaplasia and heterotopia. However, Shh expression was lost in intestinal metaplasia of the stomach. CONCLUSION: We found a strong correlation between Shh expression and fundic gland differentiation. Our current study therefore provides evidence that in addition to its role in gastric epithelial development, Shh plays a unique role in gastric epithelial differentiation in adults.


Subject(s)
Gastric Fundus/chemistry , Meckel Diverticulum/metabolism , RNA, Messenger/analysis , Trans-Activators/analysis , Adult , Cell Differentiation , Esophagus/metabolism , Esophagus/pathology , Gastric Fundus/cytology , Hedgehog Proteins , Humans , Immunohistochemistry/methods , In Situ Hybridization/methods , Intestinal Mucosa/metabolism , Intestines/pathology , Metaplasia , Trans-Activators/genetics
18.
Am J Physiol Gastrointest Liver Physiol ; 283(6): G1290-7, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12388203

ABSTRACT

Calmodulin (Cal) plays important roles for contractile activity in smooth muscles. Recently, two distinct Ca(2+)-binding protein superfamilies with sequence similarities to Cal have been identified in neuronal cells: neuronal Ca(2+)-binding proteins (NCBPs) and Cal-like Ca(2+)-binding proteins (CaBPs). Some NCBPs and CaBPs play significant roles for Ca(2+)-dependent cellular signaling in the nervous system. In gastrointestinal smooth muscles (GISMs), Cal functions as the regulator of contractile behavior and electrical rhythmicity. However, the molecular identification of NCBPs and CaBPs has not been elucidated in GISMs. Here, we have identified NCBPs and CaBPs expressed in GISMs and determined the expression levels of their transcripts by quantitative RT-PCR. Of 12 NCBPs, the transcripts for neuronal Ca(2+) sensor 1, neural visinin-like proteins 1, 2, and 3, and K(+) channel-interacting proteins 1 and 3 were detected in proximal colon, gastric fundus, gastric antrum, and jejunum. On the other hand, of seven CaBPs including alternatively spliced variants, only CaBP1L transcripts were detected in GISMs.


Subject(s)
Calcium-Binding Proteins/genetics , Digestive System/metabolism , Gene Expression , Muscle, Smooth/metabolism , Receptors, Calcium-Sensing , Repressor Proteins , Alternative Splicing , Animals , Brain Chemistry , Calcium-Binding Proteins/physiology , Calmodulin/physiology , Colon/chemistry , Digestive System/chemistry , Gastric Fundus/chemistry , Jejunum/chemistry , Kv Channel-Interacting Proteins , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Muscle, Smooth/chemistry , Myocardium/chemistry , Nerve Tissue Proteins/genetics , Neurocalcin , Neuronal Calcium-Sensor Proteins , Neurons/chemistry , Neuropeptides/genetics , Pyloric Antrum/chemistry , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
19.
J Lab Clin Med ; 139(2): 101-8, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11919548

ABSTRACT

Reg protein has a trophic effect on gastric mucosal cells and pancreatic islets. Recently, the Reg receptor (Reg-R) has been cloned, and Reg-Reg-R interaction has been reported in the pancreas. The aim of this study was to investigate the localization of Reg-R in rat fundic mucosa. Gene expression of Reg-R was investigated with Northern blot analysis, laser capture microdissection coupled with reverse transcription-polymerase chain reaction, and in situ hybridization in the fundic mucosa, and the types of cells expressing this gene were determined. Reg-R mRNA expression was detected mainly in chief cells and parietal cells of the deep layers and faintly in surface epithelial cells and mucous neck cells of the proliferating zone. Our results suggest that regenerating protein may act not only as a regulator of gastric epithelial cell proliferation but also as a modifier of other multiple physiologic functions.


Subject(s)
Calcium-Binding Proteins/physiology , Gastric Fundus/chemistry , Gastric Mucosa/chemistry , Nerve Tissue Proteins , Animals , Blotting, Northern , Cloning, Molecular , Gene Expression , In Situ Hybridization , Lasers , Lithostathine , Male , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
20.
J Histochem Cytochem ; 50(3): 385-93, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11850440

ABSTRACT

To evaluate the distribution of smooth muscle myosin heavy chain isoforms (SMB, with head insert), we examined frozen sections from the various regions of swine stomachs using isoform-specific antibodies. We previously reported variable SMB myosin heavy chain (MHC) expression in stomach cells that correlates with unloaded shortening velocities. This is consistent with the generalization of tonic fundic muscle having low expression and phasic antral muscle having high expression of the SMB MHC isoform. Using immunohistochemistry (IHC), we show a progression of the SMB MHC from very low immunoreactivity in the fundus to very intense immunoreactivity in the antrum. In the body, the average level of SMB MHC immunoreactivity lies between that of the antrum and fundus. Intercellular heterogeneity was observed in all stomach regions to a similar extent. However, the intercellular range in SMB MHC immunoreactivity decreases from fundus to antrum. All stomach regions show isolated pockets or clusters of cells with similar SMB MHC immunoreactivity. There is a non-uniform intracellular immunoreactivity in SMB MHC, with many cells showing greater-intensity staining of SMB MHC in their cell peripheries. This information may prove useful in helping to elucidate possible unique physiological roles of SMB MHC.


Subject(s)
Muscle, Smooth/chemistry , Myosin Heavy Chains/analysis , Stomach/chemistry , Swine , Animals , Fluorescent Antibody Technique , Gastric Fundus/chemistry , Immunohistochemistry , Microscopy, Fluorescence , Protein Isoforms/analysis , Pyloric Antrum/chemistry , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...