Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Int J Obes Suppl ; 6(Suppl 1): S8-S14, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28685024

ABSTRACT

The microbiota-gut-brain axis is currently being explored in many types of rodent models, including models of behavioral, neurodegenerative and metabolic disorders. Our laboratory is interested in determining the mechanisms and consequences of activation of vagal afferent neurons that lead to activation of parasympathetic reflexes and changes in feeding behavior in the context of obesity. Obesity is associated with microbial dysbiosis, decreased intestinal barrier function, gut inflammation, metabolic endotoxemia, chronic low-grade systemic inflammation and desensitization of vagal afferent nerves. This review will present the evidence that altered gut microbiota together with decreased gut barrier function allows the passage of bacterial components or metabolites in obese individuals, leading to the disruption of vagal afferent signaling and consequently resulting in an increase in body weight. We first review the most recent descriptions of gut microbial dysbiosis due to a high fat diet and describe changes in the gut barrier and the evidence of increased intestinal permeability in obesity. We then will review the evidence to show how manipulating the gut microbiota via pre and probiotics can restore gut barrier function and prevent weight gain. Lastly, we present possible mechanisms by which the microbe-gut-brain axis may have a role in obesity. The studies mentioned in this review have provided new targets to treat and prevent obesity and have highlighted how the microbiota-gut-brain axis is involved.

2.
Neurogastroenterol Motil ; 25(7): 579-e460, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23566308

ABSTRACT

BACKGROUND: Ingestion of sweet food is driven by central reward circuits and restrained by endocrine and neurocrine satiety signals. The specific influence of sucrose intake on central affective and reward circuitry and alterations of these mechanisms in the obese are incompletely understood. For this, we hypothesized that (i) similar brain regions are engaged by the stimulation of sweet taste receptors by sucrose and by non-nutrient sweeteners and (ii) during visual food-related cues, obese subjects show greater brain responses to sucrose compared with lean controls. METHODS: In a double-blind, crossover design, 10 obese and 10 lean healthy females received a sucrose or a non-nutrient sweetened beverage prior to viewing food or neutral images. BOLD signal was measured using a 1.5 Tesla MRI scanner. KEY RESULTS: Viewing food images after ingestion of either drink was associated with engagement of similar brain regions (amygdala, hippocampus, thalamus, anterior insula). Obese differed from lean subjects in behavioral and brain responses rating both beverages as less tasteful and satisfying, yet demonstrating greater brain responses. Obese subjects also showed engagement of an additional brain network (including anterior insula, anterior cingulate, hippocampus, and amygdala) only after sucrose ingestion. CONCLUSIONS & INFERENCES: Obese subjects had a reduced behavioral hedonic response, yet a greater engagement of affective brain networks, particularly after sucrose ingestion, suggesting that in obese subjects, lingual and gut-derived signaling generate less central hedonic effects than food-related memories in response to visual cues, analogous to response patterns implicated in food addiction.


Subject(s)
Beverages , Brain/physiology , Food Preferences/physiology , Obesity/physiopathology , Sweetening Agents , Adolescent , Adult , Brain Mapping , Double-Blind Method , Female , Humans , Image Interpretation, Computer-Assisted , Magnetic Resonance Imaging , Young Adult
3.
Obes Rev ; 12(11): 984-94, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21729236

ABSTRACT

Bariatric surgery is the most effective method for promoting dramatic and durable weight loss in morbidly obese subjects. Furthermore, type 2 diabetes is resolved in over 80% of patients. The mechanisms behind the amelioration in metabolic abnormalities are largely unknown but may be due to changes in energy metabolism, gut peptides and food preference. The goal of this meeting was to review the latest research to better understand the mechanisms behind the 'magic' of bariatric surgery. Replication of these effects in a non-surgical manner remains one of the ultimate challenges for the treatment of obesity and diabetes. Promising data on energy metabolism, gastrointestinal physiology, hedonic response and food intake were reviewed and discussed.


Subject(s)
Bariatric Surgery/methods , Energy Metabolism/physiology , Obesity, Morbid/surgery , Weight Loss , Ghrelin/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Obesity, Morbid/metabolism , Peptide YY/metabolism , Weight Loss/physiology
4.
Neurogastroenterol Motil ; 23(7): e282-93, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21303432

ABSTRACT

BACKGROUND: Lumenal glucose initiates changes in gastrointestinal (GI) function, including inhibition of gastric emptying, stimulation of pancreatic exocrine and endocrine secretion, and intestinal fluid secretion. Glucose stimulates the release of GI hormones and 5-hydroxytryptamine (5-HT), and activates intrinsic and extrinsic neuronal pathways to initiate changes in GI function. The precise mechanisms involved in luminal glucose-sensing are not clear; studying gut endocrine cells is difficult due to their sparse and irregular localization within the epithelium. METHODS: Here we show a technique to determine activation of gut epithelial cells and the gut-brain pathway in vivo in rats using immunohistochemical detection of the activated, phosphorylated, form of calcium-calmodulin kinase II (pCaMKII). KEY RESULTS: Perfusion of the gut with glucose (60 mg) increased pCaMKII immunoreactivity in 5-HT-expressing enterochromaffin (EC) cells, cytokeratin-18 immunopositive brush cells, but not in enterocytes or cholecystokinin-expressing cells. Lumenal glucose increased pCaMKII in neurons in the myenteric plexus and nodose ganglion, nucleus of the solitary tract, dorsal motor nucleus of the vagus and the arcuate nucleus. pCaMKII expression in neurons, but not in EC cells, was significantly attenuated by pretreatment with the 5-HT(3) R antagonist ondansetron. Deoxynojirimycin, a selective agonist for the putative glucose sensor, sodium-glucose cotransporter-3 (SGLT-3), mimicked the effects of glucose with increased pCaMKII in ECs and neurons; galactose had no effect. CONCLUSIONS & INFERENCES: The data suggest that native EC cells in situ respond to glucose, possibly via SGLT-3, to activate intrinsic and extrinsic neurons and thereby regulate GI function.


Subject(s)
Brain/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Consciousness/physiology , Gastrointestinal Tract/physiology , Glucose/pharmacology , Intestinal Mucosa/physiology , Signal Transduction/drug effects , 1-Deoxynojirimycin/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/drug effects , Enterochromaffin Cells/cytology , Enterochromaffin Cells/physiology , Glucosamine/analogs & derivatives , Glucosamine/pharmacology , Intestinal Mucosa/cytology , Male , Models, Animal , Myenteric Plexus/physiology , Ondansetron/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Serotonin, 5-HT3/drug effects , Receptors, Serotonin, 5-HT3/physiology , Serotonin Antagonists/pharmacology , Signal Transduction/physiology , Sodium-Glucose Transport Proteins/drug effects , Sodium-Glucose Transport Proteins/physiology
5.
J. physiol. biochem ; 64(4): 349-356, oct.-dic. 2008.
Article in English | IBECS | ID: ibc-61830

ABSTRACT

Although it is well established that the presence of nutrients in the gut lumen canbring about changes in GI function, the mechanisms and pathways by which thesechanges occur has not been fully elucidated. It has been known for many years thatluminal nutrients stimulate the release of hormones and regulatory peptides from gutendocrine cells and that luminal nutrients activate intrinsic and extrinsic neural pathwaysinnervating the gut. Activation of gut endocrine cells and neural pathways bynutrients in the gut lumen is key in coordination of postprandial GI function and alsoin the regulation of food intake. Recent evidence suggests that these pathways can bemodified by long term changes in diet or by inflammatory processes in the gut wall.Thus it is important to determine the cellular and molecular mechanisms underlyingthese processes not only to increase our understanding of as part of basic physiologybut also to understand changes in these pathways that occur in the presence ofpathophysiology and disease. This review summarizes some of the latest data that wehave obtained, together with information from the other laboratories, which haveelucidated some of the mechanisms involved in nutrient detection in the gut wall. Thefocus is on monosaccharides and protein hydrolysates as there is some evidence fora role for nutrient transporters in detection of these nutrients (AU)


No disponible


Subject(s)
Humans , Animals , Male , Female , Digestive System Physiological Phenomena , Enteroendocrine Cells/metabolism , Enteroendocrine Cells/physiology , Gastrointestinal Tract/physiology , Membrane Transport Proteins/physiology , Gastrointestinal Hormones/physiology , Gastrointestinal Tract/innervation , Vagus Nerve/physiology , Food , Gastrointestinal Hormones/metabolism , Gastrointestinal Hormones/pharmacology , Gastrointestinal Tract/metabolism , Glucose/metabolism , Proteins/metabolism , Nutrients
6.
Physiol Behav ; 93(4-5): 930-6, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18234246

ABSTRACT

UNLABELLED: Food intake is modulated by ingestive (gastrointestinal) and post-ingestive signals; ingested fat is potent to produce short-term satiety (satiation) but this can be modified by long-term ingestion of a high fat diet. AIM: Determine whether altered lipid-induced satiation is dependent on the fat content of the diet, rather than increased caloric density or changes in adiposity. METHODS: Initial experiments determined the differences in the microstructure of meal patterns in rats fed a high fat diet (HF: 38% fat kcal) and in rats pair-fed an isocaloric, isonitrogenous low fat diet (LF: 10% fat kcal) and changes in meal patterns measured after long-term maintenance on the HF diet. RESULTS: Rats fed the HF diet had a significant 50% increase in meal frequency compared to rats fed the LF diet; in addition, there was a significant reduction in meal size (32%) and inter meal interval (38%) consistent with induction of satiation. After 8 weeks on the HF diet, these parameters tend to approach those of rats maintained on the LF diet. There was a significant 56% decrease in the activation of neurons in the NTS in response to intragastric gavage of lipid in rats maintained for 8 weeks on the HF compared to LF diet. CONCLUSION: Dietary fat alters meal patterns consistent with induction of a short-term satiety signal. This signal is attenuated with long-term exposure to dietary lipid, in the absence of ingestion of additional calories or changes in body weight. This adaptation of short-term satiety might contribute to diet-induced obesity.


Subject(s)
Adaptation, Physiological/physiology , Lipids/pharmacology , Satiation/drug effects , Analysis of Variance , Animals , Behavior, Animal/physiology , Body Composition/drug effects , Eating/drug effects , Feeding Behavior/physiology , Food Preferences/drug effects , Male , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Satiation/physiology , Time Factors
7.
J Physiol Biochem ; 64(4): 349-56, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19391461

ABSTRACT

Although it is well established that the presence of nutrients in the gut lumen can bring about changes in GI function, the mechanisms and pathways by which these changes occur has not been fully elucidated. It has been known for many years that luminal nutrients stimulate the release of hormones and regulatory peptides from gut endocrine cells and that luminal nutrients activate intrinsic and extrinsic neural pathways innervating the gut. Activation of gut endocrine cells and neural pathways by nutrients in the gut lumen is key in coordination of postprandial GI function and also in the regulation of food intake. Recent evidence suggests that these pathways can be modified by long term changes in diet or by inflammatory processes in the gut wall. Thus it is important to determine the cellular and molecular mechanisms underlying these processes not only to increase our understanding of as part of basic physiology but also to understand changes in these pathways that occur in the presence of pathophysiology and disease. This review summarizes some of the latest data that we have obtained, together with information from the other laboratories, which have elucidated some of the mechanisms involved in nutrient detection in the gut wall. The focus is on monosaccharides and protein hydrolysates as there is some evidence for a role for nutrient transporters in detection of these nutrients.


Subject(s)
Digestive System Physiological Phenomena , Enteroendocrine Cells/physiology , Food , Gastrointestinal Tract/physiology , Membrane Transport Proteins/physiology , Animals , Enteroendocrine Cells/metabolism , Gastrointestinal Hormones/metabolism , Gastrointestinal Tract/innervation , Gastrointestinal Tract/metabolism , Glucose/metabolism , Humans , Proteins/metabolism , Vagus Nerve/physiology
8.
Endocrinology ; 148(10): 4695-703, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17641001

ABSTRACT

Peptide YY (PYY)(3-36), released by intestinal lipid elicits functional effects that comprise the intestinal feedback response to luminal nutrients, but the pathway of action is not fully characterized. The aim of the present study was to determine the role of the apolipoprotein (apo) A-IV-cholecystokinin (CCK)(1) receptor (CCK(1)R) pathway in exogenous PYY(3-36)-induced activation of the gut-brain axis and inhibition of gastric emptying and food intake. PYY(3-36) (5 microg/100 g ip) significantly inhibited gastric emptying of a chow meal in wild-type but not A-IV(-/-) mice andCCK(1)R receptor blockade with devazepide (10 microg/100 g), abolished PYY(3-36)-induced inhibition of gastric emptying. PYY(3-36)-induced inhibition of food intake in both ad libitum-fed and 16-h fasted mice was unaltered in A-IV(-/-) mice, compared with wild-type controls, or by CCK(1)R receptor blockade with devazepide. PYY(3-36) activated neurons in the midregion of the nucleus of the solitary tract (bregma -7.32 to -7.76 mm) in A-IV(+/+) mice; this was measured by immunohistochemical localization of Fos protein. PYY(3-36)-induced Fos expression was significantly reduced by 65% in A-IV(+/+) mice pretreated systemically with the sensory neurotoxin capsaicin (5 mg/100 g), 78% by the CCK(1)R antagonist, devazepide (10 microg/100 g), and 39% by the Y2R antagonist, BIIE0246 (200 and 600 microg/100 g) and decreased by 67% in apo A-IV(-/-) mice, compared with A-IV(+/+) controls. The data suggest a role for apo A-IV and the CCK(1)R in PYY(3-36)-induced activation of the vagal afferent pathway and inhibition of gastric emptying, but this is likely not the pathway mediating the effects of PYY(3-36) on food intake.


Subject(s)
Apolipoproteins A/physiology , Feedback, Physiological/drug effects , Intestines/drug effects , Intestines/physiology , Peptide YY/pharmacology , Receptor, Cholecystokinin A/physiology , Animal Feed , Animals , Apolipoproteins A/deficiency , Brain/drug effects , Brain/physiology , Eating/drug effects , Eating/physiology , Fasting , Feedback, Physiological/physiology , Gastric Emptying/drug effects , Gastric Emptying/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Peptide Fragments , Proto-Oncogene Proteins c-fos/metabolism , Solitary Nucleus/cytology , Solitary Nucleus/drug effects , Solitary Nucleus/metabolism , Solitary Nucleus/physiology
9.
Am J Physiol Gastrointest Liver Physiol ; 291(3): G439-45, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16675747

ABSTRACT

The presence of glucose in the intestinal lumen elicits a number of changes in gastrointestinal function, including inhibition of gastric emptying and food intake and stimulation of pancreatic and intestinal secretion. The present study tested the hypothesis that Na(+)-glucose cotransporter (SGLT)-3, a member of the SGLT family of transport proteins, is involved in detection of luminal glucose in the intestine. Gastric emptying, measured in awake rats, was significantly inhibited by perfusion of the intestine with glucose (60 and 90 mg); this effect was mimicked by alpha-methyl glucose (nonmetabolizable substrate of SGLT-1 and -3) but not 2-deoxy-d-glucose (substrate for GLUT-2) or isoosmotic mannitol. Gastric motility and intestinal fluid secretion, measured in anesthetised rats, were significantly inhibited and stimulated, respectively, by duodenal glucose but not galactose, which has a much lower affinity for SGLT-3 than glucose. Duodenal glucose but not galactose stimulated the release of 5-HT into mesenteric lymph and stimulated the discharge of duodenal vagal afferent fibers. mRNA for SGLT-3 was identified in the duodenal mucosa. Together these data suggest that detection of glucose in the intestine may involve SGLT-3, possibly expressed by enterochromaffin cells in the intestinal mucosa, and release of 5-HT.


Subject(s)
Gastric Emptying/physiology , Gastrointestinal Motility/physiology , Glucose/metabolism , Intestinal Mucosa/metabolism , Sodium-Glucose Transport Proteins/metabolism , Animals , Intestines/cytology , Rats , Rats, Sprague-Dawley
10.
Am J Physiol Gastrointest Liver Physiol ; 291(1): G156-62, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16574983

ABSTRACT

Cholecystokinin (CCK), acting at CCK1 receptors (CCK1Rs) on intestinal vagal afferent terminals, has been implicated in the control of gastrointestinal function and food intake. Using CCK1R(-/-) mice, we tested the hypothesis that lipid-induced activation of the vagal afferent pathway and intestinal feedback of gastric function is CCK1R dependent. In anesthetized CCK1R(+/+) ("wild type") mice, meal-stimulated gastric acid secretion was inhibited by intestinal lipid infusion; this was abolished in CCK1R(-/-) mice. Gastric emptying of whole egg, measured by nuclear scintigraphy in awake mice, was significantly faster in CCK1R(-/-) than CCK1R(+/+) mice. Gastric emptying of chow was significantly slowed in response to administration of CCK-8 (22 pmol) in CCK1R(+/+) but not CCK1R(-/-) mice. Activation of the vagal afferent pathway was measured by immunohistochemical localization of Fos protein in the nucleus of the solitary tract (NTS; a region where vagal afferents terminate). CCK-8 (22 pmol ip) increased neuronal Fos expression in the NTS of fasted CCK1R(+/+) mice; CCK-induced Fos expression was reduced by 97% in CCK1R(-/-) compared with CCK1R(+/+) mice. Intralipid (0.2 ml of 20% Intralipid and 0.04 g lipid), but not saline, gavage increased Fos expression in the NTS of fasted CCK1R(+/+) mice; lipid-induced Fos expression was decreased by 47% in CCK1R(-/-) compared with CCK1R(+/+)mice. We conclude that intestinal lipid activates the vagal afferent pathway, decreases gastric acid secretion, and delays gastric emptying via a CCK1R-dependent mechanism. Thus, despite a relatively normal phenotype, intestinal feedback in response to lipid is severely impaired in these mice.


Subject(s)
Gastric Emptying/physiology , Lipids/administration & dosage , Receptor, Cholecystokinin B/metabolism , Stomach/innervation , Stomach/physiology , Vagus Nerve/physiology , Animals , Feedback/drug effects , Feedback/physiology , Gastric Emptying/drug effects , Gene Silencing , Male , Mice , Mice, Knockout , Receptor, Cholecystokinin B/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Stomach/drug effects , Vagus Nerve/drug effects
11.
J Physiol ; 569(Pt 3): 949-58, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16239275

ABSTRACT

Long chain triglyceride (>C12) in the intestinal lumen potently inhibits gastric emptying and acid secretion via the vagal afferent pathway. While the mechanism of inhibition involves the formation of chylomicrons, the essential role of the apolipoprotein apo A-IV is unclear. Using apo A-IV(-/-) mice, we tested the hypothesis that inhibition of gastric emptying and gastric acid secretion in response to dietary lipid is dependent upon apo A-IV. As measured by nuclear scintigraphy in awake mice, gastric emptying of an ingested whole-egg meal was significantly faster in apo A-IV(-/-) knockout versus A-IV(+/+) controls (34 +/- 1 versus 54 +/- 3 min, P < 0.0001). In anaesthetized A-IV(+/+) mice, meal-stimulated gastric acid secretion was 59% inhibited by intestinal lipid infusion; this was abolished in apo A-IV(-/-) mice. Oral gavage of lipid in awake mice activated neurones throughout the nucleus of the solitary tract (NTS) in A-IV(+/+) mice, measured by immunohistochemical localization of Fos protein expression. However, in the mid region of the NTS (bregma -7.32 to -7.76 mm), Fos expression in response to intestinal lipid was significantly decreased by 50% in apo A-IV(-/-) mice compared to A-IV(+/+) controls. We conclude that activation of the vagal afferent pathway and inhibition of gastric function in response to dietary lipid is partly dependent upon apo A-IV.


Subject(s)
Apolipoproteins A/physiology , Intestines/drug effects , Lipids/pharmacology , Animals , Apolipoproteins A/genetics , Cholecystokinin/administration & dosage , Cholecystokinin/pharmacology , Devazepide/pharmacology , Gastric Acid/metabolism , Gastric Emptying/drug effects , Hormone Antagonists/pharmacology , Intestinal Mucosa/metabolism , Intestines/innervation , Intubation, Gastrointestinal , Lipids/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Solitary Nucleus/drug effects , Solitary Nucleus/metabolism , Triglycerides/blood , Vagus Nerve/drug effects
12.
J Nutr ; 135(6): 1491-5, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15930458

ABSTRACT

Intestinal infusion of protein digests activates a vago-vagal reflex inhibition of gastric motility. Protein digests release cholecystokinin (CCK) from enteroendocrine cells; however, the precise cellular mechanisms leading to vagal afferent activation is unclear. The hypothesis that the oligopeptide transporter PepT1 plays a major role in the initiation of this vago-vagal reflex was tested by recording activation of duodenal vagal afferent activity and inhibition of gastric motility in response to protein hydrolysates in the presence of 4-aminomethylbenzoic acid (4-AMBA), a competitive inhibitor of PepT1, or 4-aminophenylacetic acid (4-APAA), an inactive 4-AMBA analog. Duodenal infusion of the protein hydrolysate increased vagal afferent discharge and inhibited gastric motility; these responses were abolished by concomitant infusion of 4-AMBA, but not 4-APAA. Duodenal infusion with Cefaclor, a substrate of PepT1, increased duodenal vagal afferent activity; Cefaclor and protein hydrolysates selectively activated CCK-responsive vagal afferents. This study demonstrates that products of protein digestion increase spontaneous activity of CCK-sensitive duodenal vagal afferents via a mechanism involving the oligopeptide transporter PepT1.


Subject(s)
Digestion , Duodenum/innervation , Neurons, Afferent/physiology , Proteins/metabolism , Symporters/physiology , Vagus Nerve/physiology , para-Aminobenzoates , 4-Aminobenzoic Acid/pharmacology , Animals , Electrophysiology , Gastrointestinal Motility/drug effects , Male , Nerve Fibers/physiology , Neurons, Afferent/drug effects , Peptide Transporter 1 , Peptones/pharmacology , Rats , Rats, Sprague-Dawley , Symporters/antagonists & inhibitors , Vagus Nerve/cytology , Vagus Nerve/drug effects
13.
Neurogastroenterol Motil ; 16(4): 421-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15305997

ABSTRACT

The ability to make repetitive non-invasive measurements of gastric emptying of nutritive solids in awake, unstressed mice is highly desirable. The aim of the present study was to develop such a technique using nuclear scintigraphy and diets differing in triglyceride content. Awake mice were accustomed to light restraint and to feeding cooked, egg white (0.00 g fat g(-1)), whole egg (0.10 g fat g(-1)), or egg yolk (0.31 g fat g(-1)). Gastric emptying of each diet was measured by labelling the test meals with Technetium(99m) Mebrofenin and using a conventional gamma camera equipped with a high resolution, parallel hole collimator. Gastric emptying of cooked whole egg was also determined following administration of either vehicle or CCK A receptor antagonist, devazepide. The half-emptying time (t(1/2)) significantly increased with increasing triglyceride content from 14 +/- 5 min to 51 +/- 6 min and 82 +/- 4 min for egg white, whole egg and egg yolk, respectively. Administration of devazepide significantly decreased t(1/2) of whole egg to 28 +/- 2 min. These results demonstrate the sensitivity and predictability of this technique in mice and importantly, provide an opportunity to alter the macronutrient or caloric content of the meal to determine effects on gastric emptying.


Subject(s)
Devazepide/pharmacology , Dietary Fats/pharmacology , Gastric Emptying/physiology , Receptor, Cholecystokinin A/antagonists & inhibitors , Receptor, Cholecystokinin A/physiology , Animals , Gastric Emptying/drug effects , Male , Mice , Mice, Inbred C57BL , Radionuclide Imaging/methods , Reproducibility of Results
14.
Neurogastroenterol Motil ; 16(4): 429-38, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15305998

ABSTRACT

Galanin actions are mediated by distinct galanin receptors (GAL-R), GAL-R1, -R2 and -R3. We investigated the role of GAL-R1 in gastric motility and the expression of GAL-R1 in the rat stomach. In vivo, in urethane-anaesthetized rats, galanin (equipotent for all GAL-Rs) induced a short inhibition of gastric motility, followed by increase in tonic and phasic gastric motility; the latter was significantly reduced by the GAL-R1 antagonist, RWJ-57408. Galanin 1-16 (high affinity for GAL-R1 and -R2) induced a long-lasting decrease of intragastric pressure, which was not modified by RWJ-57408. In vitro, galanin and galanin 1-16 induced increase of intragastric pressure that was not affected by RWJ-57408. Tetrodotoxin (TTX) did not suppress the galanin excitatory effect, whereas the effect of galanin 1-16 on gastric contraction was increased by TTX- or N-nitro-L-arginine, an inhibitor of nitric oxide synthase. GAL-R1 immunoreactivity was localized to cholinergic and tachykinergic neurons and to neurons immunoreactive for nitric oxide synthase or vasoactive intestinal polypeptide. This study suggests that an extrinsic GAL-R1 pathway mediates the galanin excitatory action, whereas an extrinsic, non GAL-R1 pathway is likely to mediate the galanin inhibitory effect in vivo. GAL-R1 intrinsic neurons do not appear to play a major role in the control of gastric motility.


Subject(s)
Gastrointestinal Motility/physiology , Receptor, Galanin, Type 1/physiology , Animals , Dose-Response Relationship, Drug , Galanin/pharmacology , Gastrointestinal Motility/drug effects , In Vitro Techniques , Male , Nerve Net/drug effects , Nerve Net/physiology , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Galanin, Type 1/agonists
15.
Am J Physiol Regul Integr Comp Physiol ; 287(2): R354-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15117731

ABSTRACT

Apolipoprotein A-IV (apo A-IV), a peptide expressed by enterocytes in the mammalian small intestine and released in response to long-chain triglyceride absorption, may be involved in the regulation of gastric acid secretion and gastric motility. The specific aim of the present study was to determine the pathway involved in mediating inhibition of gastric motility produced by apo A-IV. Gastric motility was measured manometrically in response to injections of either recombinant purified apo A-IV (200 microg) or apo A-I, the structurally similar intestinal apolipoprotein not regulated by triglyceride absorption, close to the upper gastrointestinal tract in urethane-anesthetized rats. Injection of apo A-IV significantly inhibited gastric motility compared with apo A-I or vehicle injections. The response to exogenous apo A-IV injections was significantly reduced by 77 and 55%, respectively, in rats treated with the CCK(1) receptor blocker devazepide or after functional vagal deafferentation by perineural capsaicin treatment. In electrophysiological experiments, isolated proximal duodenal vagal afferent fibers were recorded in vitro in response to close-arterial injection of vehicle, apo A-IV (200 microg), or CCK (10 pmol). Apo A-IV stimulated the discharge of duodenal vagal afferent fibers, significantly increasing the discharge in 4/7 CCK-responsive units, and the response was abolished by CCK(1) receptor blockade with devazepide. These data suggest that apo A-IV released from the intestinal mucosa during lipid absorption stimulates the release of endogenous CCK that activates CCK(1) receptors on vagal afferent nerve terminals initiating feedback inhibition of gastric motility.


Subject(s)
Apolipoproteins A/pharmacology , Duodenum/innervation , Gastric Emptying/physiology , Receptor, Cholecystokinin A/metabolism , Vagus Nerve/drug effects , Animals , Apolipoproteins A/physiology , Cholecystokinin/pharmacology , Denervation , Devazepide/pharmacology , Duodenum/physiology , Hormone Antagonists/pharmacology , Male , Rats , Rats, Sprague-Dawley , Vagus Nerve/physiology , Visceral Afferents/drug effects , Visceral Afferents/physiology
16.
Neurogastroenterol Motil ; 16 Suppl 1: 60-3, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15066007

ABSTRACT

The enteric nervous system in combination with inputs from parasympathetic and sympathetic nerves regulate the contractile, secretory and vasomotor activity of the gastrointestinal track via neural reflexes. Sensory elements which may be present in specialized neurones, enteroendocrine cells or mast cells detect changes in force, chemical composition or even foreign antigens. Sensory elements signal the enteric nervous system to correct these changes by altering contractile activity, secretion and blood flow. Advances have been made in understanding the sensory mechanisms that are involved in 5-hydroxytryptamine (5-HT) release from enterochromaffin cells (EC) or a model for EC cells. These advances relate to roles for ATP and its metabolites ADP and adenosine in mechanotransduction and a role for a sodium glucose cotransporter, a SGLT-like protein, in chemotransduction.


Subject(s)
Digestive System/innervation , Enteric Nervous System/physiology , Enterochromaffin Cells/physiology , Mechanotransduction, Cellular/physiology , Sensory Receptor Cells/physiology , Animals , Enteric Nervous System/cytology , Gastrointestinal Motility/physiology , Humans , Reflex/physiology
17.
Neuroscience ; 123(1): 101-9, 2004.
Article in English | MEDLINE | ID: mdl-14667445

ABSTRACT

Immunohistochemistry and confocal microscopy were used to investigate mu opioid receptor (muOR) internalization in enteric neurons of the guinea-pig ileum following abdominal surgery. The following surgical procedures were performed under halothane or isofluorane anesthesia: a) midline abdominal skin incision, b) laparotomy or c) laparotomy with intestinal manipulation. Gastrointestinal transit was evaluated by using a non-absorbable marker and measuring fecal pellet output. In neurons from normal and control (anesthesia alone) animals, muOR was predominantly at the cell surface. muOR endocytosis following skin incision was not significantly different from controls (21.2+/-3.5% vs. 13.7+/-2.1%, mean+/-S.E.M.), whereas it was significantly increased by laparotomy (46.5+/-6.1%; P<0.01 vs. controls) or laparotomy plus intestinal manipulation (40.5+/-6.1%; P<0.01 vs. controls) 30 min following surgery compared with controls. muOR endocytosis remained elevated at 4 h (38.6+/-1.2%; P<0.01 vs. controls), whereas it was similar to controls at 6 and 12 h (17.5+/-5.8% and 11.2+/-3.0%). muOR endocytosis occurred in cholinergic and nitrergic neurons. Gastrointestinal transit was significantly delayed by laparotomy or laparotomy plus intestinal manipulation (12.8+/-1.2 and 13.8+/-0.6 h vs. 7.0+/-0.5 in controls; P<0.01), but was not significantly changed by skin incision (8.2+/-0.6 h). The findings of the present study support the concept that the noxious stimulation caused by abdominal surgery induces release of endogenous opioids thus resulting in muOR endocytosis in neurochemically distinct enteric neurons. muOR internalization can serve as indirect evidence of opioid release and as a means to visualize neuronal pathways activated by opioids.


Subject(s)
Endocytosis/physiology , Enteric Nervous System/metabolism , Ileum/metabolism , Ileum/surgery , Receptors, Opioid, mu/metabolism , Abdomen/physiology , Abdomen/surgery , Animals , Enteric Nervous System/chemistry , Guinea Pigs , Male , Neurons/chemistry , Neurons/metabolism , Receptors, Opioid, mu/analysis
18.
Neurogastroenterol Motil ; 14(4): 403-8, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12213108

ABSTRACT

Abstract In a number of different experimental paradigms of somatic pain, there is evidence for a vagally mediated antinociceptive system. This pathway probably involves opioid mechanisms. However, whether this pathway is activated in visceral pain or if it involves subdiaphragmatic vagal afferents is unclear. The aim of the present study was to determine whether subdiaphragmatic vagal afferents mediate antinociception in response to a visceral stimulus and whether this involves an opioid pathway. Colorectal distension was performed in fasted, conscious male Sprague-Dawley rats using a balloon catheter connected to an electronic distension device. The number of abdominal contractions (visceromotor response) in response to a tonic colorectal distension (60 mmHg for 10 min) was recorded. Experiments were performed in sham or subdiaphragmatically vagotomized, perineural vehicle- or capsaicin-treated rats (to functionally denervate vagal afferents) before and after administration of naloxone (25 mg kg(-1) bodyweight intraperitoneally). Vagotomy, capsaicin and naloxone pretreatments all significantly enhanced the visceromotor response to colorectal distension. The effect of naloxone in capsaicin-treated rats did not appear to be additive. These results suggest that activation of subdiaphragmatic afferents, which can be blocked by capsaicin, may play a role in opioid-dependent antinociceptive pathways activated by a noxious visceral stimulus.


Subject(s)
Colon/physiology , Diaphragm/innervation , Opioid Peptides/physiology , Pain Measurement/methods , Rectum/physiology , Vagus Nerve/physiology , Afferent Pathways/drug effects , Afferent Pathways/physiology , Animals , Colon/drug effects , Colon/innervation , Diaphragm/drug effects , Diaphragm/physiology , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Male , Naloxone/pharmacology , Pain Measurement/drug effects , Pain Measurement/statistics & numerical data , Rats , Rats, Sprague-Dawley , Rectum/drug effects , Rectum/innervation , Vagotomy/methods , Vagotomy/statistics & numerical data , Vagus Nerve/drug effects
19.
Gut ; 51 Suppl 1: i11-4, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12077056

ABSTRACT

The possible mechanisms that may be involved in nutrient detection in the wall of the gastrointestinal tract are reviewed. There is strong functional and electrophysiological evidence that both intrinsic and extrinsic primary afferent neurones mediate mechano- and chemosensitive responses in the gastrointestinal tract. This review focuses on the extrinsic afferent pathways as these are the ones that convey information to the central nervous system which is clearly necessary for perception to occur.


Subject(s)
Food , Nociceptors/physiology , Sensation/physiology , Visceral Afferents/physiology , Epithelial Cells/metabolism , Humans , Intestinal Mucosa/metabolism , Neurons, Afferent/physiology
20.
Gastroenterology ; 121(6): 1400-6, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11729119

ABSTRACT

BACKGROUND & AIMS: 5-Hydroxytryptamine (5-HT) is released from enterochromaffin cells and activates neural reflex programs regulating motility and secretion. Although sugars are reported to release 5-HT in vivo, it is unclear whether they act directly on enterochromaffin cells or indirectly through an intermediary messenger. The aim was to determine if D-glucose is a stimulus for 5-HT release. METHODS: Human BON cells, derived from enterochromaffin cells, were treated with D-glucose, galactose, and the nonmetabolizable methyl alpha-D-glucopyranoside, or with fructose. RESULTS: Reverse-transcription polymerase chain reaction together with Western blot analysis revealed an SGLT-like protein. D-glucose caused a concentration-dependent increase in 5-HT release, which was mimicked by methyl alpha-D-glucopyranoside and galactose but not fructose. D-glucose-stimulated 5-HT release was significantly reduced by phloridzin. Concentrations of mannitol below 75 mmol/L were ineffective in releasing 5-HT. Brefeldin A abolished forskolin-stimulated 5-HT release without affecting basal or constitutive release. CONCLUSIONS: The results show that high concentrations of metabolizable and nonmetabolizable hexoses activate signal transduction pathways, leading to release of 5-HT. These findings imply a role for enterochromaffin cells as "glucose sensors" during ingestion of a meal.


Subject(s)
Enterochromaffin Cells/drug effects , Enterochromaffin Cells/metabolism , Glucose/pharmacology , Serotonin/metabolism , Brefeldin A/pharmacology , Cell Line , Colforsin/pharmacology , Fructose/pharmacology , Galactose/pharmacology , Gene Expression , Humans , Mannitol/pharmacology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Methylglucosides/pharmacology , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Phlorhizin/pharmacology , Sodium-Glucose Transporter 1
SELECTION OF CITATIONS
SEARCH DETAIL
...