Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
1.
Vet Parasitol ; 105(4): 285-301, 2002 May 10.
Article in English | MEDLINE | ID: mdl-11983304

ABSTRACT

Infection with the bovine abomasal nematode, Ostertagia ostertagi, results in a loss of acid-secreting parietal cells and an increase in gastric pH. The effects of an experimental infection with Ostertagia and/or daily treatment with omeprazole (OMP) at 2mgkg(-1) bodyweight for four consecutive days (experiment days 24-27, inclusive) on voluntary feed intake, blood and tissue gastrin concentrations, abomasal G-cell numbers, gastric pH, and blood cholecystokinin (CCK) and pepsinogen concentrations were investigated in the calf. Ostertagia-infected calves demonstrated a significant drop in feed intake between days 24 and 27 post-infection (38%; P<0.001) and in G-cell numbers (42%; P<0.05) and significant increases in abomasal pH (P<0.001), fundic mucosal weight (99%; P<0.01), and blood gastrin (P<0.05) and pepsinogen (P<0.0001). OMP treatment of worm-free animals resulted in a significant drop in intake between days 24 and 27 (30%; P<0.001) and in G-cell numbers (17%; P<0.05) and significant increases in abomasal pH (P<0.01) and blood gastrin (P<0.001). OMP treatment of Ostertagia-infected animals with an existing hypergastrinaemia had no effect on feed intake, abomasal pH, blood gastrin or pepsinogen or abomasal G-cell numbers. Blood CCK concentrations were also unaffected by either Ostertagia infection or OMP treatment. These data suggest that: (a) the depression in feed intake associated with OMP in worm-free calves was not due to a side effect of drug treatment; (b) inappetance in Ostertagia-infected animals is closely associated with the parasite-induced hypergastrinaemia; and (c) the elevation in abomasal pH was a major factor responsible for the elevated blood gastrin concentrations seen in parasitised and OMP-treated animals.


Subject(s)
Cattle Diseases/parasitology , Enzyme Inhibitors/pharmacology , Gastrins/metabolism , Omeprazole/pharmacology , Ostertagia/growth & development , Ostertagiasis/veterinary , Abomasum/parasitology , Abomasum/pathology , Animals , Cattle , Cattle Diseases/drug therapy , Cattle Diseases/metabolism , Cattle Diseases/pathology , Cholecystokinin/blood , Eating/drug effects , Feces/parasitology , Female , Gastrins/blood , Hydrogen-Ion Concentration , Immunohistochemistry/veterinary , Ostertagia/metabolism , Ostertagiasis/blood , Ostertagiasis/drug therapy , Ostertagiasis/metabolism , Parasite Egg Count , Pepsinogen A/blood
3.
Gut ; 49(1): 23-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11413106

ABSTRACT

BACKGROUND AND AIMS: The main goal of our study was to characterise the activity of BIM26226 as a peripheral gastrin releasing peptide (GRP) receptor antagonist in healthy human subjects and to determine if endogenous GRP is a physiological regulator of gastric acid secretion and gastrin release. METHODS: Our study consisted of three parts. In part I, subjects received saline or BIM26226 followed by graded doses of intravenous human GRP in a four period crossover design. In part II, subjects received BIM26226 or saline during oral meal ingestion or modified sham feeding. In part III, subjects received an acidified meal in the presence and absence of BIM26226 in a two period crossover design. In addition, gastrin and somatostatin mRNA were measured in biopsy specimens during saline and BIM26226 infusion. RESULTS: BIM26226 dose dependently inhibited GRP induced acid output. Acid secretion after oral liquid meal intake and sham feeding was significantly inhibited by BIM26226 (p<0.01) whereas plasma gastrin release remained unchanged. Gastrin and somatostatin mRNAs were not significantly different after saline or BIM26226. CONCLUSIONS: BIM26226 is a potent GRP antagonist in humans. Endogenous GRP may be a physiological regulator of gastric acid secretion. Gastrin release does not seem to be under the control of GRP.


Subject(s)
Bombesin/analogs & derivatives , Bombesin/pharmacology , Gastric Acid/metabolism , Gastrin-Releasing Peptide/physiology , Peptide Fragments/pharmacology , Adult , Analysis of Variance , Blotting, Northern , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Eating/physiology , Gastric Acidity Determination , Gastrin-Releasing Peptide/antagonists & inhibitors , Gastrins/analysis , Humans , Male , Middle Aged , RNA, Messenger/analysis , Somatostatin/analysis , Statistics, Nonparametric
4.
Aliment Pharmacol Ther ; 15(7): 1077-84, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11421885

ABSTRACT

BACKGROUND: Concanavalin-A, the lectin present in Jack beans, binds to mannose- and glucose-containing residues and can interact with the epidermal growth factor receptor and moderate cell proliferation in vitro. AIM: To compare the actions of concanavalin-A and epidermal growth factor on the gastrointestinal tract in vivo. METHODS: Rats maintained on total parenteral nutrition were given intragastric concanavalin-A, intravenous epidermal growth factor or concanavalin-A and epidermal growth factor. Cell proliferation and crypt fission were assayed in 'micro-dissected' crypts. RESULTS: Concanavalin-A and epidermal growth factor both significantly elevated proliferation in the small intestine and colon. No significant interaction between the effects of these two agents was seen, except in the mid small intestine where there was a synergistic interaction. Concanavalin-A had no effect on crypt branching. Epidermal growth factor significantly reduced branching in the distal small intestine and mid colon. CONCLUSION: The effects of the two agents appeared to be separate, except in the mid small intestine where they were additive. This is in marked contrast with the actions reported in vitro, where concanavalin-A is a powerful inhibitor of epidermal growth factor-induced cell proliferation. Concanavalin-A thus has potential for enhancing the functions of the small intestine.


Subject(s)
Cell Division/drug effects , Concanavalin A/pharmacology , Epidermal Growth Factor/pharmacology , Intestines/drug effects , Intestines/growth & development , Animals , Drug Interactions , Infusions, Intravenous , Male , Parenteral Nutrition, Total , Rats , Rats, Sprague-Dawley
5.
Helicobacter ; 6(1): 1-14, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11328360

ABSTRACT

BACKGROUND: Further elucidation of the consequences of Helicobacter pylori infection on gastric mucosal inflammation and gastric secretory function would be facilitated by an animal model that is susceptible to infection with H. pylori, is broadly similar in gastric physiology and pathology to people, and is amenable to repeated non-invasive evaluation. The goal of this study was to examine the interrelationship of bacterial colonization, mucosal inflammation and gastric secretory function in cats with naturally acquired H. pylori infection. MATERIALS AND METHODS: Twenty clinically healthy cats with naturally acquired H. pylori infection (cagA-, picB) and 19 Helicobacter-free cats were evaluated. Gastric colonization was determined by tissue urease activity, light microscopy, culture and PCR. The mucosal inflammatory response was evaluated by light microscopy, and by RT-PCR of the pro-inflammatory cytokines IL-1alpha, IL-1beta, IL-8 and TNF-alpha in gastric mucosa. Gastric secretory function was assessed by measuring pentagastrin-stimulated acid secretion, fasting plasma gastrin, and antral mucosal gastrin and somatostatin immunoreactivity. RESULTS: H. pylori colonized the pylorus, fundus and cardia in similar density. Bacteria were observed free in the lumen of gastric glands and were also tightly adherent to epithelial cells where they were associated with microvillus effacement. Mononuclear inflammation, lymphoid follicle hyperplasia, atrophy and fibrosis were observed primarily in H. pylori-infected cats, with the pylorus most severely affected. Neutrophilic and eosinophilic infiltrates, epithelial dysplasia, and up-regulation of mucosal IL-1beta and IL-8 were observed solely in infected cats. Fasting plasma gastrin concentrations and pentagastrin-stimulated acid output were similar in both infected and uninfected cats. There was no relationship of bacterial colonization density or gastric inflammation to plasma gastrin concentrations or gastric acid output. CONCLUSIONS: The pattern of colonization and the mucosal inflammatory response in cats with naturally acquired H. pylori are broadly similar to those in infected people, particularly children, and non-human primates. The upregulation of IL-8 in infected cats was independent of cagA and picB. Our findings argue against a direct acid-suppressing effect of H. pylori on the gastric secretory-axis in chronically infected cats.


Subject(s)
Antigens, Bacterial , Cat Diseases/microbiology , Gastric Mucosa/microbiology , Gastritis/veterinary , Helicobacter Infections/veterinary , Helicobacter pylori , Animals , Bacterial Proteins/metabolism , Cardia/microbiology , Cardia/pathology , Cat Diseases/metabolism , Cat Diseases/pathology , Cats , Disease Models, Animal , Female , Gastric Acidity Determination , Gastric Fundus/microbiology , Gastric Fundus/pathology , Gastric Mucosa/metabolism , Gastrins/metabolism , Gastritis/metabolism , Gastritis/microbiology , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Interleukin-1/biosynthesis , Interleukin-8/biosynthesis , Male , Pyloric Antrum/metabolism , Pyloric Antrum/microbiology , Pyloric Antrum/pathology , Reverse Transcriptase Polymerase Chain Reaction , Somatostatin/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
6.
Gut ; 48(6): 782-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11358895

ABSTRACT

BACKGROUND: Interleukin 1beta (IL-1beta) is a potent inhibitor of gastric acid secretion. Regulatory actions at several levels have previously been demonstrated, including direct inhibition of parietal cell acid secretion. Although IL-1beta may activate several intracellular signalling pathways, the mechanisms responsible for inhibition of carbachol stimulated acid secretion have not been determined. AIMS: To investigate the roles of protein kinase C (PKC) and the sphingomyelinase signalling pathways in the regulation of acid secretion by IL-1beta. METHODS: Rabbit parietal cells were obtained by collagenase-EDTA digestion and centrifugal elutriation. Acid secretion stimulated by carbachol and A23187 (to mimic elevations in intracellular calcium) was assessed by 14C aminopyrine uptake in response to IL-1beta, PKC, and sphingomyelinase manipulation. RESULTS: IL-1beta inhibited carbachol and A23187 stimulated acid secretion in a dose dependent manner. The inhibitory actions were completely reversed by each of three different PKC inhibitors, staurosporine, H-7, and chelerythrine, as well as by PKC depletion with high dose phorbol ester pretreatment. IL-1beta did not downregulate parietal cell muscarinic receptor. IL-1beta significantly increased membrane PKC activity. Activation of the sphingomyelinase/ceramide pathway had no effect on basal or stimulated acid secretion. The inhibitory action of IL-1beta was independent of protein kinase A and protein kinase G activity. CONCLUSIONS: IL-1beta directly inhibits parietal cell carbachol stimulated acid secretion. This action occurs distal to muscarinic receptor activation and elevations in intracellular calcium and requires PKC.


Subject(s)
Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Gastric Acid/physiology , Interleukin-1/physiology , Parietal Cells, Gastric/drug effects , Protein Kinase C/physiology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Alkaloids , Analysis of Variance , Animals , Benzophenanthridines , Carbachol/antagonists & inhibitors , Carcinogens/pharmacology , Cell Communication/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Parietal Cells, Gastric/physiology , Phenanthridines/pharmacology , Rabbits , Sphingomyelin Phosphodiesterase/physiology , Staurosporine/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
7.
J Appl Physiol (1985) ; 90(1): 105-13, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11133899

ABSTRACT

The purpose of the present investigation was to determine the independent effects of hypoxia and physical exercise on peripheral cholecystokinin (CCK) metabolism in humans. Thirty-two physically active men were randomly assigned in a double-blind manner to either a normoxic (N; n = 14) or hypoxic (H; n = 18) group. During the acute study, subjects in the H group only participated in two tests, separated by 48 h, which involved a cycling test to exhaustion in normobaric normoxia and normobaric hypoxia (inspired O(2) fraction = 0.21 and 0.16, respectively). In the intermittent study, N and H groups cycle-trained for 4 wk at the same relative exercise intensity in both normoxia and hypoxia. Acute normoxic exercise consistently raised plasma CCK during both studies by 290-723%, which correlated with increases in the plasma ratio of free tryptophan to branched chain amino acids (r = 0.58-0.71, P < 0.05). In contrast, acute hypoxic exercise decreased CCK by 7.0 +/- 5.5 pmol/l, which correlated with the decrease in arterial oxygen saturation (r = 0.56, P < 0.05). In the intermittent study, plasma CCK response at rest and after normoxic exercise was not altered after physical training, despite a slight decrease in adiposity. We conclude that peripheral CCK metabolism 1) is more sensitive to acute changes than chronic changes in energy expenditure and 2) is potentially associated with acute changes in tissue PO(2) and metabolic precursors of cerebral serotoninergic activity.


Subject(s)
Cholecystokinin/blood , Exercise/physiology , Hypoxia/blood , Adult , Amino Acids, Branched-Chain/blood , Anthropometry , Atmospheric Pressure , Blood Glucose/analysis , Diet , Double-Blind Method , Energy Intake , Fatty Acids, Nonesterified/blood , Glycerol/blood , Humans , Lactic Acid/blood , Male , Oxygen Consumption , Physical Education and Training , Reference Values , Tryptophan/blood
8.
Clin Chim Acta ; 301(1-2): 181-92, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11020472

ABSTRACT

Infection with the bacterium Helicobacter pylori is associated with altered gastric acid secretion and gastrointestinal disease. Recent work has suggested that N alpha-methylhistamine, produced by the bacterium and acting on histamine receptors in gastric tissue, might be involved. Gastric juice and tissue biopsies from infected patients have been analysed for the presence of N alpha-methylhistamine using a specific and sensitive assay based on gas chromatography mass spectrometry. N alpha-Methylhistamine was detected in five of seven samples of gastric juice from infected patients (5-180 pmol/ml) but was absent in nine uninfected subjects. The compound was not found in fundic and antral biopsies from both subject groups. Helicobacter pylori, cultured on agar and in broth with and without added histamine, was found not to produce detectable levels of N alpha-methylhistamine. Instillation of this compound at 10(-5) mol/l into the gastric lumen produced a significant increase in acid secretion in vivo while plasma gastrin concentration remained unchanged. N alpha-Methylhistamine in gastric juice appears therefore to be associated with infection, although this product is not generated directly by the bacterium. The concentrations found are below those required to affect acid secretion or gastrin production in vivo, although higher local concentrations may exist around a site of infection.


Subject(s)
Gastric Acid/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori/pathogenicity , Methylhistamines/metabolism , Gas Chromatography-Mass Spectrometry , Gastric Mucosa/metabolism , Gastrins/blood , Humans , Sensitivity and Specificity
9.
Eur J Gastroenterol Hepatol ; 12(6): 641-4, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10912482

ABSTRACT

OBJECTIVE: To investigate the mechanisms underlying the hypergastrinaemia of Helicobacter pylori by examining the effects of H. pylori on basal and stimulated gastrin release from cultured canine G-cells. METHODS: Canine antral G-cells were prepared by collagenase-EDTA digestion and cultured for 40 h. G-cells were then cultured for a further 24 h with two different H. pylori sonicates before basal and bombesin-stimulated gastrin release were measured by radioimmunoassay. RESULTS: Treatment of G-cells with both H. pylori sonicates significantly enhanced basal gastrin release (by 17-27%) and bombesin-stimulated gastrin release (by 115-133%). This effect was independent of cagA and vacuolating cytotoxin status. Control treatment with Escherichia coli sonicate had no effect on gastrin release. There was no change in the cellular content of gastrin. CONCLUSIONS: Incubation of antral G-cells with H. pylori constituents enhances subsequent basal and bombesin-stimulated gastrin release. Direct contact between H. pylori and G-cells in the gastric antrum may be responsible for the hypergastrinaemia seen with the infection.


Subject(s)
Gastrin-Secreting Cells/metabolism , Gastrins/metabolism , Helicobacter pylori/physiology , Animals , Bombesin/pharmacology , Cells, Cultured , Dogs , Gastrin-Secreting Cells/drug effects , Gastrin-Secreting Cells/microbiology
10.
Aliment Pharmacol Ther ; 14(6): 835-40, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10848670

ABSTRACT

BACKGROUND: Ingested peanut agglutinin stimulates colonic proliferation in humans. In rats, ingested peanut agglutinin stimulates hormone release and proliferation in the small and large intestines. Peanut agglutinin is absorbed into the circulation but little is known about the systemic effect of this lectin. Therefore, we studied the effect of intravenous peanut agglutinin on hormone release and intestinal growth. METHOD: Six rats per group received peanut agglutinin infusion at 0, 2, 20 or 200 microg/rat/day for 6 days via the right jugular vein. Organ weights were measured, pancreatic enzymes, DNA, RNA and protein levels were analysed. Plasma hormones were measured by radioimmunoassay. All tissues were examined histologically. Small intestinal and colonic proliferation rates were estimated by metaphase arrest. RESULTS: High-dose peanut agglutinin significantly reduced the wet weight of the stomach by 7% (P < 0.05) and large intestine by 10% (P < 0.05). Peanut agglutinin dose-dependently released enteroglucagon; low-, medium- and high-dose by 64%, 126% (P < 0.01) and 180% (P < 0.01), respectively, and glucagon-like peptide-1 by 127% (P < 0.01), 169% (P < 0.01) and 315% (P < 0.001), respectively. Peanut agglutinin had no effect on cholesystokinin, gastrin or insulin levels. Peanut agglutinin, low-, medium- and high-dose stimulated proliferation in the mid colon by 42% (P < 0.01), 30% and 38%, respectively. Only high-dose peanut agglutinin stimulated proliferation in the distal colon by 54% (P < 0.01). No histological changes were evident in any tissue. CONCLUSION: Intravenous peanut agglutinin released hormones and stimulated colonic proliferation. Proliferation of the small intestine seen after ingestion of peanut agglutinin in previous studies appears to require luminal contact between enterocytes and the lectin. Possible clinical applications include reversal of atrophy during total parenteral nutrition, anastomotic healing after surgery and restoration of mucosa integrity in colitis.


Subject(s)
Cell Division/drug effects , Colon/drug effects , Gastrointestinal Hormones/metabolism , Intestine, Small/drug effects , Peanut Agglutinin/pharmacology , Animals , Colon/cytology , Colon/physiology , Infusions, Intravenous , Intestine, Small/cytology , Intestine, Small/physiology , Peanut Agglutinin/administration & dosage , Rats
11.
J Chromatogr B Biomed Sci Appl ; 739(2): 337-44, 2000 Mar 10.
Article in English | MEDLINE | ID: mdl-10755377

ABSTRACT

A gas chromatography-electron capture mass spectrometry assay has been developed for the histamine H3 receptor agonist, N(alpha)-methylhistamine (N(alpha)-MH). The assay is linear from 50 pg-10 ng, with a limit of detection of 50 pg/ml for gastric juice and plasma, and 50 pg/sample for bacteria (10(7)-10(8) CFU) and gastric tissue (5-10 mg wet weight). The limits of quantification are 100 pg/ml for gastric juice (%RSD=1.4) and plasma (%RSD=9.4), and 100 pg/sample for bacteria (%RSD=3.9) and tissue (%RSD=5.8). N(alpha)-MH was not present in human plasma, but low levels (1.4 ng/ml and 0.4 ng/ml) were detected in two samples of human gastric juice obtained from patients infected with Helicobacter pylori.


Subject(s)
Gas Chromatography-Mass Spectrometry/methods , Gastric Juice/metabolism , Methylhistidines/metabolism , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/isolation & purification , Humans , Methylhistidines/blood , Sensitivity and Specificity
12.
Infect Immun ; 68(2): 779-90, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10639446

ABSTRACT

The relationship of Helicobacter felis, a bacterium observed in the stomachs of cats, to gastric disease is unclear. The objective of this study was to determine if H. felis infection alters gastric histopathology, proinflammatory cytokine expression, and secretory function and evokes a humoral immune response in cats. Five specific-pathogen-free (SPF) Helicobacter-free cats were studied before and for 1 year after oral inoculation with H. felis (ATCC 49179). Four SPF H. felis-uninfected cats served as controls. The stomachs of all five H. felis-inoculated cats became colonized, as determined by urease activity, histopathology, PCR, culture, and transmission electron microscopy of serial gastric biopsies at 0, 3, 5, 8, and 12 months. Uninoculated cats remained Helicobacter free. Lymphoid follicular hyperplasia, atrophy, and fibrosis were observed primarily in the pylorus of infected cats. Mild mononuclear inflammation was detected in both infected and uninfected cats, but was more extensive in infected cats, with pangastric inflammation, eosinophilic infiltrates, and cardia gastritis observed only in infected cats. No upregulation of antral mucosal interleukin 1alpha (IL-1alpha), IL-1beta, or tumor necrosis factor alpha was detected by reverse transcription-PCR in any cat. The gastric secretory axes, assessed by fasting plasma gastrin, antral mucosal gastrin and somatostatin immunoreactivity, and pentagastrin-stimulated gastric acid secretion, were similar in both infected and uninfected cats. Gradual seroconversion (immunoglobulin G) was observed in four of five infected cats, with enzyme-linked immunosorbent assay values reaching 4x to 12x baseline 12 months postinfection. These findings indicate that H. felis infection in cats induces lymphoid follicular hyperplasia, mild gastritis, and seroconversion, but is associated with normal gastric secretory function.


Subject(s)
Gastric Mucosa/pathology , Gastritis/etiology , Helicobacter Infections/pathology , Lymphoid Tissue/pathology , Animals , Antibodies, Bacterial/blood , Cats , Cytokines/biosynthesis , Gastric Mucosa/metabolism , Gastrins/analysis , Hyperplasia , Immunohistochemistry , Male , Polymerase Chain Reaction , Somatostatin/analysis , Urease/metabolism
13.
High Alt Med Biol ; 1(1): 9-23, 2000.
Article in English | MEDLINE | ID: mdl-11258590

ABSTRACT

The aims of the present study were to measure the satiety neuropeptide cholecystokinin (CCK) in humans at terrestrial high altitude to investigate its possible role in the pathophysiology of anorexia, cachexia, and acute mountain sickness (AMS). Nineteen male mountaineers aged 38 +/- 12 years participated in a 20 +/- 5 day trek to Mt. Kanchenjunga basecamp (BC) located at 5,100 m, where they remained for 7 +/- 5 days. Subjects were examined at rest and during a maximal exercise test at sea-level before/after the expedition (SL1/SL2) and during the BC sojourn. There was a mild increase in Lake Louise AMS score from 1.1 +/- 1.2 points at SL1 to 2.3 +/- 2.3 points by the end of the first day at BC (P < 0.05). A marked increase in resting plasma CCK was observed on the morning of the second day at BC relative to sea-level control values (62.9 +/- 42.2 pmol/L(-1) vs. SL1: 4.3 +/- 8.3 pmol/L(-1), P < 0.05 vs. SL2: 26.5 +/- 25.2 pmol/L(-1), P < 0.05). Maximal exercise increased CCK by 78.5 +/- 24.8 pmol/L(-1), (P < 0.05 vs. resting value) during the SL1 test and increased the plasma concentration of non-esterified fatty acids and glycerol at BC (P < 0.05 vs. SL1/SL2). The CCK response was not different in five subjects who presented with anorexia on Day 2 compared with those with a normal appetite. While there was no relationship between the increase in CCK and AMS score at BC, a more pronounced increase in resting CCK was observed in subjects with AMS (> or =3 points at the end of Day 1 at BC) compared with those without (+98.9 +/- 1.4 pmol/L(-1) vs. +67.6 +/- 37.2 pmol/L(-1), P < 0.05). Caloric intake remained remarkably low during the stay at BC (8.9 +/- 1.4 MJ.d(-1)) despite a progressive decrease in total body mass (-4.5 +/- 2.1 kg after 31 +/- 13 h at BC, P < 0.05 vs. SL1/SL2), which appeared to be due to a selective loss of torso adipose tissue. These findings suggest that the satiogenic effects of CCK may have contributed to the observed caloric deficit and subsequent cachexia at high altitude despite adequate availability of palatable foods. The metabolic implications of elevated CCK in AMS remain to be elucidated.


Subject(s)
Altitude Sickness/blood , Anorexia/blood , Cachexia/blood , Cholecystokinin/blood , Mountaineering , Adult , Altitude Sickness/complications , Analysis of Variance , Anorexia/etiology , Anthropometry , Appetite , Blood Glucose , Cachexia/etiology , Energy Intake , Exercise , Fatty Acids, Nonesterified/blood , Glycerol/blood , Hand Strength , Humans , Male
14.
Aliment Pharmacol Ther ; 13(12): 1669-74, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10594403

ABSTRACT

BACKGROUND: Gastrin release by Helicobacter pylori may be an important step in the pathway leading to duodenal ulceration. A histamine H3-receptor agonist was found to release gastrin from antral mucosal fragments; this was interpreted as being due to suppression of somatostatin release. H. pylori is reported to produce Nalpha-methyl histamine (NalphaMH), which is an agonist of H3 as well as other histamine receptors. H. pylori infection also recruits mast cells, which release histamine. AIM: To determine the direct effects of histamine receptor agonists on isolated gastrin cells. METHODS: Rabbit G-cells were prepared by countercurrent elutriation and cultured on 24-well plates. RESULTS: NalphaMH (10-6-10-4 M) caused a dose-dependent increase in gastrin release from a basal level of 2.3 +/- 0.2% total cell content (TCC; mean +/- S.E.M.) to a maximum of 5.1 +/- 0.7%, an increase of 117% (P < 0. 005) above basal. This was abolished by the H2-antagonist ranitidine (10-5 M), but not by immunoblockade with anti-somatostatin antibody, the H1-antagonist chlorpheniramine (10-5 M) or the H3-antagonist thioperamide (10-4 M). The histamine H2-receptor agonist dimaprit (10-6-10-4 M) increased gastrin release from 2.4 +/- 0.2% to 3.6 +/- 0.2% TCC (P < 0.001). Gastrin release was also stimulated by histamine (10-7-10-4 M) from a basal value of 3.0 +/- 0.3% to 5.4 +/- 0.5% TCC (P < 0.001). This also was inhibited by ranitidine (10-5 M) (P < 0.01). CONCLUSION: NalphaMH and histamine release gastrin from G-cells via H2-receptors; this might contribute to H. pylori-associated hypergastrinaemia.


Subject(s)
Gastrin-Secreting Cells/drug effects , Gastrin-Secreting Cells/metabolism , Gastrins/metabolism , Histamine Agonists/pharmacology , Histamine/pharmacology , Pyloric Antrum/metabolism , Animals , Antibodies, Monoclonal/immunology , Cells, Cultured , Chlorpheniramine/pharmacology , Dimaprit/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Histamine/analogs & derivatives , Histamine Antagonists/pharmacology , Histamine H2 Antagonists/pharmacology , Piperidines/pharmacology , Pyloric Antrum/drug effects , Rabbits , Ranitidine/pharmacology , Receptors, Histamine/physiology
15.
Am J Physiol ; 276(5): G1235-42, 1999 05.
Article in English | MEDLINE | ID: mdl-10330015

ABSTRACT

Total parenteral nutrition (TPN) causes atrophy of gastrointestinal epithelia, so we asked whether lectins that stimulate epithelial proliferation can reverse this effect of TPN. Two lectins stimulate pancreatic proliferation by releasing CCK, so we asked whether lectins that stimulate gastrointestinal proliferation also release hormones that might mediate their effects. Six rats per group received continuous infusion of TPN and a once daily bolus dose of purified lectin (25 mg. rat-1. day-1) or vehicle alone (control group) for 4 days via an intragastric cannula. Proliferation rates were estimated by metaphase arrest, and hormones were measured by RIAs. Phytohemagglutinin (PHA) increased proliferation by 90% in the gastric fundus (P < 0.05), doubled proliferation in the small intestine (P < 0.001), and had a small effect in the midcolon (P < 0.05). Peanut agglutinin (PNA) had a minor trophic effect in the proximal small intestine (P < 0.05) and increased proliferation by 166% in the proximal colon (P < 0.001) and by 40% in the midcolon (P < 0.001). PNA elevated circulating gastrin and CCK by 97 (P < 0.05) and 81% (P < 0.01), respectively, and PHA elevated plasma enteroglucagon by 69% and CCK by 60% (both P < 0.05). Only wheat germ agglutinin increased the release of glucagon-like peptide-1 by 100% (P < 0.05). PHA and PNA consistently reverse the fall in gastrointestinal and pancreatic growth associated with TPN in rats. Both lectins stimulated the release of specific hormones that may have been responsible for the trophic effects. It is suggested that lectins could be used to prevent gastrointestinal atrophy during TPN. Their hormone-releasing effects might be involved.


Subject(s)
Cell Division , Digestive System Physiological Phenomena , Digestive System/cytology , Lectins/pharmacology , Parenteral Nutrition, Total , Animals , Cholecystokinin/blood , Colon/cytology , Gastrins/blood , Glucagon/blood , Glucagon-Like Peptide 1 , Glucagon-Like Peptides/blood , Insulin/blood , Intestine, Small/cytology , Male , Organ Size , Pancreas/cytology , Peanut Agglutinin/pharmacology , Peptide Fragments/blood , Phytohemagglutinins/pharmacology , Protein Precursors/blood , Rats , Rats, Sprague-Dawley , Stomach/cytology
16.
Gut ; 44(5): 709-14, 1999 May.
Article in English | MEDLINE | ID: mdl-10205210

ABSTRACT

BACKGROUND: Dietary lectins can alter the proliferation of colonic cells. Differentiation is regulated by adhesion molecules which, being glycosylated, are targets for lectin binding. AIMS: To examine the effects of dietary lectins on differentiation, adhesion, and proliferation of colorectal cancer cells. METHODS: Differentiation was assessed in three dimensional gels, adhesion by aggregation assay, and proliferation by 3H thymidine incorporation. The role of the epithelial cell adhesion molecule (epCAM) was studied using a specific monoclonal antibody in blocking studies and Western blots. The human colon cancer cell lines LS174T, SW1222, and HT29 were studied. RESULTS: The cell line LS174T differentiated in the presence of Vicia faba agglutinin (VFA) into gland like structures. This was inhibited by anti-epCAM monoclonal antibody. Expression of epCAM itself was unaffected. VFA as well as wheat germ agglutinin (WGA) and the edible mushroom lectin (Agaricus bisporus lectin, ABL) significantly aggregated LS174T cells but peanut agglutinin (PNA) and soybean agglutinin (SBA) did not. All lectins aggregated SW1222 and HT29 cells. Aggregation was blocked by the corresponding sugars. Aggregation of cells by VFA was also inhibited by anti-epCAM. VFA, ABL, and WGL inhibited proliferation of all the cell lines; PNA stimulated proliferation of HT29 and SW1222 cells. In competition studies all sugars blocked aggregation and proliferation of all cell lines, except that the addition of mannose alone inhibited proliferation. CONCLUSION: VFA stimulated an undifferentiated colon cancer cell line to differentiate into gland like structures. The adhesion molecule epCAM is involved in this. Dietary or therapeutic VFA may slow progression of colon cancer.


Subject(s)
Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , Fabaceae/chemistry , Lectins/pharmacology , Plants, Medicinal , Antigens, Neoplasm/metabolism , Blotting, Western , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Differentiation/drug effects , Cell Division/drug effects , Epithelial Cell Adhesion Molecule , Humans , Plant Lectins , Tumor Cells, Cultured/drug effects
17.
Yale J Biol Med ; 72(2-3): 195-202, 1999.
Article in English | MEDLINE | ID: mdl-10780581

ABSTRACT

Helicobacter pylori plays major causative roles in peptic ulcer disease and gastric cancer. Elevated acid secretion in patients with duodenal ulcers (DUs) contributes to duodenal injury, and diminished acid secretion in patients with gastric cancer allows carcinogen-producing bacteria to colonize the stomach. Eradication of H. pylori normalizes acid secretion both in hyper-secreting DU patients and hypo-secreting relatives of gastric cancer patients. Therefore, we and others have asked how H. pylori causes these disparate changes in acid secretion. H. pylori gastritis more or less restricted to the gastric antrum in DU patients is associated with increased acid secretion. This is probably because gastritis increases release of the antral acid-stimulating hormone gastrin and diminished mucosal expression of the inhibitory peptide somatostatin. Bacterial products and inflammatory cytokines including TNFalpha may cause these changes in endocrine function. Gastritis involving the gastric corpus tends to diminish acid secretion, probably because bacterial products and cytokines including IL-1 inhibit parietal cells. Pharmacological inhibition of acid secretion increases corpus gastritis in H. pylori-infected subjects, so it is envisaged that gastric hypo-secretion of any cause might become self-perpetuating. H. pylori-associated mucosal atrophy will also contribute to acid hypo-secretion and is more likely in when the diet is high in salt or lacking in antioxidant vitamins. Data on gastric acid secretion in patients with esophagitis are limited but suggest that acid secretion is normal or slightly diminished. Nevertheless, H. pylori infection may be relevant to the management of esophagitis because: (i) H. pylori infection increases the pH-elevating effect of acid inhibiting drugs; (ii) proton pump inhibitors may increase the tendency of H. pylori to cause atrophic gastritis; and (iii) successful eradication of H. pylori is reported to increase the likelihood of esophagitis developing in patients who had DU disease. Points (ii) and (iii) remain controversial and more work is clearly required to elucidate the relationship between H. pylori, acid secretion, gastric mucosa atrophy and esophagitis.


Subject(s)
Digestive System/cytology , Digestive System/microbiology , Gastric Acid/metabolism , Gastroesophageal Reflux/etiology , Helicobacter Infections/metabolism , Helicobacter pylori , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/microbiology , Humans
18.
Peptides ; 19(9): 1541-7, 1998.
Article in English | MEDLINE | ID: mdl-9864061

ABSTRACT

We have studied the mechanism of soybean agglutinin (SBA) mediated cholecystokinin (CCK) release in enriched cultured cholecystokinin-secreting cells. 12-O-Tetradecanoylphorbol-13-acetate 1 mM significantly stimulated release of CCK-like-immunoreactivity (CCK-LI) by 55%+/-17% (p < 0.05), which was blocked by the protein kinase C inhibitor staurosporine 100 nM. Forskolin 10 mM stimulated CCK-LI by 82%+/-12% (p < 0.05) and this was inhibited by somatostatin 1 nM. 1-Phenylalanine 20 mM and Bay K 8644 1 mM stimulated CCK-LI by 69%+/-22% and 60%+/-19% respectively (p < 0.05), these responses were completely abolished by the L-type calcium channel antagonist verapamil 10 mM. SBA 10 and 100 microg/ml stimulated CCK-LI by 65%+/-22% and 74%+/-24% respectively (p < 0.05). The effect of SBA was inhibited by verapamil and N-acetylgalactosamine. We conclude that SBA stimulates CCK-LI through calcium flux via L-type calcium channels.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Cholecystokinin/metabolism , Jejunum/drug effects , Lectins/pharmacology , Plant Lectins , Soybean Proteins , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Adenylyl Cyclases/metabolism , Animals , Calcium Channels, L-Type , Cell Separation , Cells, Cultured , Cholecystokinin/immunology , Colforsin/pharmacology , Enzyme Activation , Jejunum/cytology , Male , Phenylalanine/pharmacology , Protein Kinase C/metabolism , Rabbits , Radioimmunoassay , Somatostatin/pharmacology , Staurosporine/pharmacology , Verapamil/pharmacology
19.
Eur J Gastroenterol Hepatol ; 10(4): 281-3, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9855041

ABSTRACT

Tham et al. show that Helicobacter pylori infection lowers the density of immunoreactive somatostatin cells (D-cells) in the antral mucosa and elevates plasma gastrin concentrations. According to current hypothesis, the lack of inhibition by somatostatin allows excessive release of gastrin, which stimulates acid secretion and thus causes duodenal ulcers. The cytokine tumour necrosis factor-alpha which is released in H. pylori gastritis inhibits D-cells in culture and may be responsible. Why do not all infected persons get duodenal ulcers? Recent work shows that more aggressive strains of H. pylori have greater effects on somatostatin/gastrin physiology. Another variable is whether the infection causes corpusitis or not. Inflammation of the gastric corpus diminishes acid secretion, which greatly decreases the likelihood of duodenal ulcers but increases the risk of gastric cancer. Factors which promote corpusitis include diets with high salt content or lacking in antioxidant vitamins. Work in this area is elucidating how H. pylori causes different diseases. Hopefully this will allow us to predict and prevent its serious sequelae.


Subject(s)
Gastrins/metabolism , Helicobacter Infections/physiopathology , Helicobacter pylori/pathogenicity , Somatostatin-Secreting Cells/physiology , Somatostatin/metabolism , Stomach Diseases/microbiology , Helicobacter Infections/pathology , Humans , Somatostatin-Secreting Cells/microbiology , Stomach Diseases/metabolism , Stomach Diseases/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...