Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Zhongguo Dang Dai Er Ke Za Zhi ; 12(7): 563-8, 2010 Jul.
Article in Zh | 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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Endocrinology ; 141(11): 4255-61, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11089560

ABSTRACT

Ghrelin, a novel GH-releasing acylated peptide, was recently isolated from rat stomach. It stimulated the release of GH from the anterior pituitary through the GH secretagogue receptor (GHS-R). Ghrelin messenger RNA and the peptide are present in rat stomach, but its cellular source has yet to be determined. Using two different antibodies against the N- and C-terminal regions of rat ghrelin, we identified ghrelin-producing cells in the gastrointestinal tracts of rats and humans by light and electron microscopic immunohistochemistry and in situ hybridization combined with immunohistochemistry. Ghrelin-immunoreactive cells, which are not enterochromaffin-like cells, D cells, or enterochromaffin cells, accounted for about 20% of the endocrine cell population in rat and human oxyntic glands. Rat ghrelin was present in round, compact, electron-dense granules compatible with those of X/A-like cells whose hormonal product and physiological functions have not previously been clarified. The localization, population, and ultrastructural features of ghrelin-producing cells (Gr cells) indicate that they are X/A-like cells. Ghrelin also was found in enteric endocrine cells of rats and humans. Using two RIAs for the N- and C-terminal regions of ghrelin, we determined its content in the rat gastrointestinal tract. Rat ghrelin was present from the stomach to the colon, with the highest content being in the gastric fundus. Messenger RNAs of ghrelin and GHS-R also were found in these organs. Ghrelin probably functions not only in the control of GH secretion, but also in the regulation of diverse processes of the digestive system. Our findings provide clues to additional, as yet undefined, physiological functions of this novel gastrointestinal hormone.


Subject(s)
Enteroendocrine Cells/metabolism , Peptide Hormones , Peptides/analysis , Animals , Chromatography, High Pressure Liquid , Gastric Fundus/chemistry , Ghrelin , Humans , Immunohistochemistry , In Situ Hybridization , Intestine, Large/chemistry , Intestine, Small/chemistry , Jejunum/chemistry , Male , Microscopy, Immunoelectron , Peptides/genetics , RNA, Messenger/analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
8.
J Clin Endocrinol Metab ; 84(7): 2568-73, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10404838

ABSTRACT

The role of mineralocorticoids in human gastrointestinal tract is well established. In the stomach, aldosterone is thought to regulate electrolyte transport associated with gastric acid secretion. In mineralocorticoid target organs, the action of the glucocorticoid inactivating enzyme 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2) facilitates aldosterone binding to a nonselective mineralocorticoid receptor (MR) in the face of high levels of circulating glucocorticoids. In the present study, we examined 25 specimens of human stomach for the presence of MR and 11beta-HSD2 using a [3H]aldosterone binding assay, Northern blot analysis, RT-PCR, and immunohistochemistry. Specific [3H]aldosterone binding sites were detected in gastric fundic mucosa, but not in the antrum. In fundic mucosa the Kd was 0.72+/-0.05 nmol/L (mean +/- SE), and Bmax was 6.0+/-1.4 fmol per milligram of protein. Northern blot analysis demonstrated a faint band for MR mRNA at 6.0 kb, although message for 11beta-HSD2 was undetectable. However, RT-PCR demonstrated specific PCR products for both MR and 11beta-HSD2. Immunohistochemistry demonstrated the colocalization of MR and 11beta-HSD2 only in parietal cells. MR-positive cells were further characterized by electron microscopy, confirming the identity of parietal cells. This study shows that parietal cells contain both MR and 11beta-HSD2, suggesting that the human stomach is a novel target organ for mineralocorticoids. Aldosterone may, therefore, regulate biological functions of parietal cells including gastric acid secretion.


Subject(s)
Gastric Mucosa/metabolism , Gene Expression , Hydroxysteroid Dehydrogenases/genetics , Receptors, Mineralocorticoid/genetics , 11-beta-Hydroxysteroid Dehydrogenases , Aldosterone/metabolism , Blotting, Northern , Gastric Fundus/chemistry , Gastric Fundus/metabolism , Gastric Mucosa/chemistry , Humans , Hydroxysteroid Dehydrogenases/analysis , Hydroxysteroid Dehydrogenases/metabolism , Immunohistochemistry , Microscopy, Electron , RNA, Messenger/analysis , Receptors, Mineralocorticoid/analysis , Receptors, Mineralocorticoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/metabolism , Tritium
9.
J Mol Endocrinol ; 11(2): 223-9, 1993 Oct.
Article in English | MEDLINE | ID: mdl-7507679

ABSTRACT

To investigate the hypothesis that gastrin might be synthesized by tumour tissues in cancer of the colon, samples from six human colon tumours, one hepatic metastasis, four normal colonic mucosal samples and two antral and one fundic gastric mucosal samples from nine patients were analysed to determine whether gastrin mRNA was present. RNA was extracted from surgical specimens by ultracentrifugation on a CsCl cushion, purified using the guanidinium thiocyanate method, reverse-transcribed and amplified by polymerase chain reaction. Gastrin mRNA was detected in each colonic carcinoma sample (including the hepatic metastasis), while no such signal was observed in normal colon biopsies. Positive and negative controls (gastric antrum and fundus respectively) gave the expected results. In each of the positive samples, the chemiluminescent revelation of amplified products after Southern blotting corresponded to gastrin mRNA without the intron. These findings demonstrate the ability of primary and metastatic human colonic tumours to produce gastrin mRNA. Since malignant cell lines have been reported to produce gastrin peptide, and since gastrin receptors were present in some cases, our results support the idea that gastrin may be involved in an autocrine mechanism.


Subject(s)
Colonic Neoplasms/genetics , Gastrins/genetics , Neoplasm Proteins/genetics , RNA, Messenger/analysis , Base Sequence , Colon/chemistry , Colonic Neoplasms/chemistry , Gastric Fundus/chemistry , Gastric Mucosa/chemistry , Humans , Intestinal Mucosa/chemistry , Liver Neoplasms/chemistry , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Luminescent Measurements , Molecular Sequence Data , Organ Specificity , Polymerase Chain Reaction , Pyloric Antrum/chemistry , RNA, Messenger/genetics , RNA-Directed DNA Polymerase
10.
Am J Med ; 91(2A): 52S-57S, 1991 Aug 08.
Article in English | MEDLINE | ID: mdl-1679296

ABSTRACT

To explore the mechanisms of the effects of sucralfate on the stomach, we investigated the action of sucrose octasulfate (SOS), a constituent of sucralfate, on the function of canine gastric parietal cells and somatostatin cells and in the isolated perfused intact rat stomach. Somatostatin cells from the canine gastric fundus were isolated by EDTA-collagenase dispersion and counterflow elutriation, and somatostatin-like immunoreactivity (SLI) release in response to SOS was measured by radioimmunoassay. Similar methods were used to isolate gastric parietal cells, in which gastric acid secretion was measured by uptake of a radiolabeled weak base, [14C]aminopyrine. SLI release by the intact rat stomach was examined in an isolated vascularly perfused rat stomach model. SOS, either alone or co-administered with epinephrine or gastrin heptadecapeptide (G17), dose-dependently stimulated SLI release by isolated canine fundic D-cells. At the highest doses, SOS potentiated the effect of epinephrine but not G17. Similarly, SOS potentiated the stimulating effect of dibutyryl cyclic adenosine 3',5'-monophosphate (DBcAMP), but not 12-O-tetradecanoylphorbol 13-acetate (TPA). The effect of SOS on SLI release could be inhibited by octreotide, a somatostatin analogue. SOS did not alter acid secretion by cultured canine parietal cells either in the basal state or when coadministered with acid secretagogues. In isolated perfused rat stomach studies, SOS produced a significant (60% greater than basal) increase in SLI secretion. There was a similar effect when SOS was perfused against a background of isoproterenol. SOS stimulates SLI release from gastric somatostatin cells and from the isolated perfused stomach but has no direct effect on gastric parietal cells. These actions of SOS may mediate in part the apparent ability of sucralfate to enhance gastric mucosal defense.


Subject(s)
Parietal Cells, Gastric/drug effects , Peptides/metabolism , Somatostatin/metabolism , Sucrose/analogs & derivatives , Aminopyrine/pharmacokinetics , Animals , Bucladesine/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Epinephrine/pharmacology , Gastric Fundus/chemistry , Gastric Fundus/drug effects , Gastric Fundus/metabolism , Gastrins/pharmacology , Isoproterenol/pharmacology , Male , Octreotide/pharmacology , Parietal Cells, Gastric/chemistry , Parietal Cells, Gastric/metabolism , Peptides/chemistry , Radioimmunoassay , Rats , Rats, Inbred Strains , Somatostatin/chemistry , Sucrose/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
11.
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
12.
Biochem Pharmacol ; 44(4): 733-40, 1992 Aug 18.
Article in English | MEDLINE | ID: mdl-1510720

ABSTRACT

Clonidine-displacing substance (CDS) from brain is biologically active in the kidney and stomach and on platelets. To determine whether CDS is contained in these and other peripheral tissues, homogenates of fresh brain, eight other organs and serum from rat were ultrafiltered (less than 10,000 mol. wt only), dried and extracted with methanol. Evaluation by radioimmunoassay (RIA) using antibodies to p-aminoclonidine showed that adrenal gland and gastric fundus (GF) contained significantly greater amounts of CDS-like radioimmunoactivity than brain; intermediate-to-low activity was present in heart, small intestine, serum, kidney and liver; lung and skeletal muscle values were near-background. RIA-positive extracts elicited well-correlated contractile activity in a GF smooth muscle bioassay; contractions persisted in the presence of antagonists of various transmitters and modulators, but were abolished by low concentrations of the calcium channel blocker verapamil. Serum levels of CDS were profoundly reduced following removal of the adrenal glands. We conclude that a CDS-like substance is present not only in brain as previously reported, but also in peripheral organs and in the circulation.


Subject(s)
Adrenal Glands/metabolism , Clonidine/antagonists & inhibitors , Adrenal Glands/chemistry , Adrenalectomy , Animals , Brain Chemistry , Clonidine/analogs & derivatives , Clonidine/analysis , Clonidine/blood , Clonidine/metabolism , Gastric Fundus/chemistry , Gastric Fundus/drug effects , Gastric Fundus/metabolism , Male , Muscle Contraction/drug effects , Radioimmunoassay , Rats , Rats, Inbred Strains , Tissue Extracts/pharmacology , Verapamil/pharmacology
13.
Int J Oncol ; 12(6): 1253-8, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9592182

ABSTRACT

The expression of CD44 splice variant containing exon 14 (variant exon 9: CD44v9) was examined immunohistochemically in non-neoplastic mucosa, adenoma and adenocarcinoma of the stomach and analyzed the relation with the expression of Ki-67 antigen and p53 protein. In non-neoplastic gastric mucosa, basolateral membrane of the epithelial cells in the pyloric glands showed the expression of CD44v9. The epithelial cells in the intestinal metaplastic mucosa of the stomach sometimes expressed CD44v9. In the neoplastic lesions, the expression of CD44v9 was detected in 20% (34/170) of the adenomas and 28% (132/478) of the adenocarcinomas, respectively. The incidence of CD44v9 expression did not differ among histological type of gastric carcinoma. Twelve per cent of the adenocarcinomas showed strong expression of CD44v9, whereas non of the adenomas did. The incidence of CD44v9 expression was significantly higher in carcinomas invading into muscularis propria or the cases of stages 3 and 4 in comparison with that in carcinomas limited to submucosa or the stages 1 and 2 cases (p<0.05). The incidence of positive cases was higher in carcinomas with lymph node metastasis than those without metastasis (p<0.05). The expression of CD44v9 was significantly correlated with the expression of Ki-67 (p<0.05). It was also correlated with the expression of p53 protein in the tumor cells (p<0.01). These findings overall suggest that the expression of CD44v9 may be associated with the development as well as progression of the gastric carcinomas.


Subject(s)
Adenocarcinoma/chemistry , Adenoma/chemistry , Hyaluronan Receptors/genetics , Stomach Neoplasms/chemistry , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenoma/genetics , Adenoma/pathology , Biomarkers/analysis , Cell Division , Disease Progression , Exons/genetics , Gastric Fundus/chemistry , Gastric Mucosa/chemistry , Gene Expression , Genetic Variation/genetics , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Neoplasm Staging , Severity of Illness Index , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Suppressor Protein p53/analysis
14.
Hum Pathol ; 27(9): 896-903, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8816883

ABSTRACT

Fundic gland polyps are benign lesions, composed of a disorderly arrangement of normal gastric corpus cell types, that occur in a large proportion of patients with familial adenomatous polyposis (FAP) but also develop sporadically in non-FAP patients as well. In this study, the authors evaluated and compared the endoscopic, histological, mucin histochemical, and microscopic stereologic features of 77 fundic gland polyps (FGPs) (15 FAP; 62 non-FAP) to determine if FAP-associated and sporadic lesions are histologically distinct. The authors also analyzed the distribution of mitotically active cells and smooth muscle cells in these lesions using MIB-1 and smooth muscle alpha-actin immunohistochemistry in an effort to determine the pathogenesis of these lesions. The results show that, compared with non-FAP cases, FAP patients with FGPs have a lower male-to-female ratio, a younger mean age at diagnosis, and a higher proportion of cases with multiple polyps. However, no differences were detected between FAP and non-FAP-associated FGPs with respect to any endoscopic, morphological, mucin histochemical, or stereometric features. Eighty-six percent of FGPs showed an increase in smooth muscle content, often in a pericystic distribution. MIB-1-positive proliferative cells were observed not only in the foveolar stem cell region, as expected, but also in the epithelium lining the microcysts and in the gland buds located directly adjacent to the microcysts. The authors conclude that FAP and non-FAP-associated FGPs are histologically identical, and propose that proliferation and subsequent differentiation of aberrantly located proliferative cells in these lesions may explain the histogenesis of FGPs.


Subject(s)
Gastric Fundus/chemistry , Gastric Fundus/pathology , Mucins/chemistry , Nuclear Proteins/chemistry , Polyps/chemistry , Polyps/pathology , Stomach Neoplasms/chemistry , Stomach Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Antigens, Nuclear , Female , Gastric Fundus/immunology , Humans , Immunohistochemistry , Ki-67 Antigen , Male , Microscopy , Middle Aged , Mucins/immunology , Nuclear Proteins/immunology , Polyps/immunology , Stomach Neoplasms/immunology
15.
J Cancer Res Clin Oncol ; 119(3): 155-9, 1993.
Article in English | MEDLINE | ID: mdl-8418088

ABSTRACT

The effect of N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) on the mucin phenotype of non-metaplastic gastric mucosa in the rat was studied histochemically. Animals were exposed to MNNG in drinking water (83 mg/l) for 12 weeks. Carcinogen treatment was then discontinued and the animals (27 in the treatment group and 25 in the control group) were examined after another 44 weeks. Glycosylation was analysed with histochemical stains for sialomucins and sulphomucins and with peroxidase-conjugated lectins (GS-II, SBA, DBA, UEA-I, and WGA). Sialo- and sulphomucins remained quantitatively unchanged, only a slight increase of acid mucins in the antral glands was observed. The analysis of the lectin binding patterns, however, revealed a significant increase for WGA-binding glycoproteins in the surface mucous cells and gastric pits, while DBA binding was significantly decreased (P < 0.05). GS-II lectin bound specifically to the proliferative compartment in the gastric fundus, consisting of mucous neck cells, and was significantly increased after MNNG treatment. No specific alterations were detected in lectin binding to parietal or chief cells. It is concluded, therefore, that treatment of gastric mucosa with MNNG alters the glycoprotein metabolism before intestinal metaplasia can be observed.


Subject(s)
Gastric Mucosa/metabolism , Methylnitronitrosoguanidine/pharmacology , Mucins/chemistry , Animals , Gastric Fundus/chemistry , Gastric Mucosa/drug effects , Lectins , Male , Mucins/metabolism , Phenotype , Rats , Rats, Wistar
16.
Histol Histopathol ; 12(3): 719-27, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9225154

ABSTRACT

The proliferation and migration of stem cells in the developing and adult rat fundic gland have been studied using BrdU immunohistochemistry and BrdU-GSA II (Griffonia-simplicifolia agglutinin-II) double staining. In the developing rat fundic gland, stem cells were first scattered throughout all levels of the epithelia and then concentrated in the depth of the pits. With the elongation and maturation of the fundic glands, stem cells left the gland base and moved upward. By 4 weeks after birth, the development of the fundic gland was completed and stem cells were confined to a narrow proliferative zone in the isthmus, reaching the adult distribution pattern. In the adult rat fundic gland, stem cells in the isthmus differentiated and migrated upward and downward, replacing the surface mucous cells and glandular cells respectively. For upward migration, it took about one week for stem cells to migrate from the isthmus to the surface. For downward migration, it took about two weeks for stem cells to migrate from the isthmus to the neck, and it took 30-36 weeks to reach the gland unit's blind end. Finally stem cells were lost at the deepest level of the glands. The results obtained by simple topographical distribution in the present experiment agreed well with those obtained by quantitative analysis, suggesting the usefulness of BrdU immunohistochemistry for cell kinetic studies.


Subject(s)
Gastric Fundus/cytology , Gastric Mucosa/cytology , Immunohistochemistry/methods , Plant Lectins , Stem Cells/cytology , Aging , Animals , Bromodeoxyuridine/analysis , Cell Division , Cell Movement , Evaluation Studies as Topic , Gastric Fundus/chemistry , Gastric Mucosa/chemistry , Lectins/analysis , Rats , Rats, Wistar , Stem Cells/chemistry
17.
Peptides ; 19(8): 1359-64, 1998.
Article in English | MEDLINE | ID: mdl-9809649

ABSTRACT

The effect of post-mortem delay on the affinity and density of tachykinin NK1 and NK2 receptors was examined in the rat submandibular gland and gastric fundus, respectively, using saturation binding studies with the radioligands [125I]Bolton-Hunter [Sar9, Met(O2)11]SP and [125I][Lys5, Tyr(I2)7, MeLeu9, Nle10]NKA(4-10). For NK1 receptors, no significant changes were seen in either Kd (control 375 +/- 35 pM, n = 5; 32 h post-mortem 390 +/- 59 pM, n = 5) or Bmax (control 96 +/- 16 fmol/mg protein, n = 5; 32 h post-mortem 62 +/- 10 fmol/mg protein, n = 5). For NK2 receptors, no alterations were seen up to 16 h post-mortem. However, significant (p < 0.001) changes were seen at 32 h post-mortem (n = 4), where values for Kd were increased (3.0 +/- 0.2 nM) and those for Bmax were reduced (42 +/- 5.9 fmol/mg protein), relative to control (Kd = 1.3 +/- 0.2 nM; Bmax = 208 +/- 30 fmol/mg protein, n = 5). These changes are probably related to observed histological deterioration. This study demonstrates the stability of tachykinin receptors in these peripheral tissues and indicates the suitability of post-mortem tissue as a valid control in future tachykinin receptor studies.


Subject(s)
Gastric Fundus/chemistry , Receptors, Tachykinin/analysis , Submandibular Gland/chemistry , Tissue Extracts/chemistry , Animals , Male , Postmortem Changes , Radioligand Assay , Rats , Rats, Wistar
18.
Peptides ; 22(12): 2055-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11786190

ABSTRACT

A novel protein expressed by entero-endocrine cells of the mouse stomach was named prepromotilin Related Peptide (ppMTLRP) since it shares sequence similarities with the prepromotilin (Tomasetto et al.). The mouse ppMTLRP was found identical to the rat precursor of ghrelin (ppghrelin), an endogenous ligand specific for the Growth Hormone Secretagogue receptor identified from rat stomach (Kojima et al.). In the present study the cDNA encoding the dog counterpart of ppMTLRP/Ghrelin has been isolated and sequenced. The dog ppMTLRP/Ghrelin cDNA showed scores of respectively 80% and 75% homology with its human and mouse counterparts. By translation of the dog ppMTLRP/Ghrelin cDNA sequences, two ORFs could be deduced encoding either a 117 amino acid ppMTLRP/Ghrelin or the deleted Gln14 ppMTLRP/Ghrelin, as it was also known in mouse, rat and man. The dog ppMTLRP/Ghrelin shared 91% similarity and 78% identity, and 89% similarity and 78% identity with the human and mouse ppMTLRP/Ghrelin proteins respectively. The best score of homology was found in the MTLRP/Ghrelin sequence itself. Indeed the dog MTLRP/Ghrelin peptide shared 100% similarity and 93% identity, and 96% identity and similarity, with the human and mouse MTLRP/Ghrelin. Using Northern blot analysis to study dog ppMTLRP/Ghrelin gene expression on dog adult gut tissues, maximal expression level was found in the stomach fundus and corpus, and no expression could be detected in the stomach antrum nor in the duodenum, jejunum, ileum, colon or liver. In conclusion, we have identified ppMTLRP/Ghrelin from dog, and found that it is highly conserved with man, mouse or rat. The expression pattern along the gastro-intestinal tract is similar to the expression pattern previously described in mouse.


Subject(s)
Gastric Fundus/chemistry , Motilin/genetics , Peptide Hormones , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary , Dogs , Ghrelin , Molecular Sequence Data , Motilin/chemistry , Motilin/isolation & purification , RNA, Messenger/genetics , Sequence Homology, Amino Acid
19.
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
20.
J Pharmacol Toxicol Methods ; 37(2): 97-103, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9174985

ABSTRACT

Nitric oxide (NO) is an inhibitory nonadrenergic, noncholinergic (NANC) neurotransmitter in the rat gastric fundus and is released upon electrical or pharmacological stimulation of the inhibitory NANC neurons. In this study, it was attempted to measure the release of NO from the rat gastric fundus upon electrical stimulation or administration of nicotine directly via an electrochemical probe (ISO-NO). The system was evaluated by adding exogenous NO. Addition of exogenous NO induced concentration-dependent relaxation of the rat gastric fundus and an increase in the ISO-NO probe baseline current. The concentration of NO detected by the ISO-NO probe was lower than the concentration of NO administered. When no tissue was present, higher concentrations of NO were detected than in the presence of a tissue. In the absence of 95% O2/5% CO2 the concentration of NO detected was highest. Electrical stimulation induced relaxations of the rat gastric fundus which were reduced by NG-nitro-L-arginine methylester (L-NAME). An increase in the ISO-NO probe baseline current was also observed, but this was duc to nonspecific effects as the response also occurred without a tissue present and was not sensitive to L-NAME. Nicotine induced relaxations, which were reduced by L-NAME, but the ISO-NO probe baseline current remained unaltered, even in the presence of L-arginine plus superoxide dismutase. It can be concluded that it is not possible to detect directly the NO release from the rat gastric fundus upon electrical or pharmacological stimulation of the NANC neurons with the ISO-NO probe.


Subject(s)
Gastric Fundus/chemistry , Nitric Oxide/analysis , Animals , Electric Stimulation , Electrochemistry/instrumentation , Electrochemistry/methods , Enzyme Inhibitors/pharmacology , Female , Gastric Fundus/drug effects , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nicotine/pharmacology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL