Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
1.
Endocr Dev ; 25: 41-8, 2013.
Article in English | MEDLINE | ID: mdl-23652390

ABSTRACT

Ghrelin is the endogenous ligand for the growth hormone secretagogue receptor (GHS-R) which was identified in the pituitary gland and is now named the ghrelin receptor. However, the peptide is most abundant in the stomach and ghrelin receptors are present in all major organ systems and tissues. Ghrelin forms with motilin, a new gut peptide family and the sequence similarities of peptides and receptors suggest they evolved by gene duplication. Nevertheless, no cross-reactivity exits between both peptides. Ghrelin shares with motilin motor effects in the gut, in particular gastric emptying and the induction of the migrating motor complex, but ghrelin also affects gastric acid secretion, offers gastroprotection and may modulate intestinal inflammation. The effects of ghrelin result from the activation of central, vagal and enteric neural receptors and receptors on immune cells. Ghrelin agonists have been developed for the treatment of hypomotility disorders and the peptidomimetic TZP-102 is in phase 2 clinical trials for diabetic gastroparesis.


Subject(s)
Gastrointestinal Tract/metabolism , Ghrelin/metabolism , Animals , Cross Reactions , Cytoprotection/drug effects , Cytoprotection/physiology , Gastroenteritis/etiology , Gastroenteritis/prevention & control , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/physiology , Ghrelin/agonists , Ghrelin/pharmacology , Humans , Motilin/immunology , Motilin/metabolism , Motilin/physiology , Peptic Ulcer/etiology , Peptic Ulcer/prevention & control , Receptors, Ghrelin/immunology , Receptors, Ghrelin/metabolism
2.
Neurogastroenterol Motil ; 21(1): 59-70, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18823291

ABSTRACT

Ghrelin is an important orexigenic peptide that not only exerts gastroprokinetic but also immunoregulatory effects. This study aimed to assess the role of endogenous and exogenous ghrelin in the pathogenesis of colitis and in the disturbances of gastric emptying and colonic contractility during this process. Dextran sodium sulphate colitis was induced for 5 days in (i) ghrelin(+/+) and ghrelin(-/-) mice and clinical and histological parameters were monitored at days 5, 10 and 26 and (ii) in Naval Medical Research Institute non-inbred Swiss (NMRI) mice treated with ghrelin (100 nmol kg(-1)) twice daily for 5 or 10 days. Neural contractility changes were measured in colonic smooth muscle strips, whereas gastric emptying was measured with the (14)C octanoic acid breath test. Inflammation increased ghrelin plasma levels. Body weight loss, histological damage, myeloperoxidase activity and IL-1beta levels were attenuated in ghrelin(-/-) mice. Whereas absence of ghrelin did not affect changes in colonic contractility, gastric emptying in the acute phase was accelerated in ghrelin(+/+) but not in ghrelin(-/-) mice. In agreement with the studies in ghrelin knockout mice, 10 days treatment of NMRI mice with exogenous ghrelin enhanced the clinical disease activity and promoted infiltration of neutrophils and colonic IL-1beta levels. Unexpectedly, ghrelin treatment decreased excitatory and inhibitory neural responses in the colon of healthy but not of inflamed NMRI mice. Endogenous ghrelin enhances the course of the inflammatory process and is involved in the disturbances of gastric emptying associated with colitis. Treatment with exogenous ghrelin aggravates colitis, thereby limiting the potential therapeutic properties of ghrelin during intestinal inflammation.


Subject(s)
Colitis/physiopathology , Ghrelin/metabolism , Animals , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate/toxicity , Gastric Emptying/drug effects , Gastric Emptying/physiology , Ghrelin/pharmacology , Male , Mice , Mice, Knockout , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/drug effects , Muscle, Smooth/physiology
4.
Br J Pharmacol ; 153(7): 1550-7, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18204478

ABSTRACT

BACKGROUND AND PURPOSE: The finding that obestatin, a peptide encoded by the ghrelin gene, opposes ghrelin's stimulatory effect on food intake and gastric emptying has been questioned. The effect of obestatin has been mostly investigated in fasted rodents, a condition associated with high blood levels of ghrelin which may mask the effect of obestatin. We therefore investigated the effect of obestatin on food intake, gastric emptying and gastric contractility in ghrelin knockout mice. EXPERIMENTAL APPROACH: The effect of obestatin on 6-h cumulative food intake was studied in fasted wildtype (ghrelin+/+) and ghrelin knockout (ghrelin-/-) mice. In both genotypes, the effect of obestatin and/or ghrelin was studied in vivo on gastric emptying measured with the (14)C-octanoic acid breath test and in vitro on neural responses elicited by electrical field stimulation (EFS) of fundic smooth muscle strips. KEY RESULTS: Administration of obestatin did not influence fasting-induced hyperphagia or gastric emptying in both genotypes. Injection of ghrelin accelerated gastric emptying in ghrelin+/+ and ghrelin-/- mice but the effect was not reversed by co-injection with obestatin. In fundic strips from ghrelin+/+ and ghrelin-/- mice, ghrelin increased EFS-induced contractions, but obestatin was without effect. However, co-administration with obestatin tended to reduce the excitatory effect of ghrelin in both genotypes. CONCLUSIONS AND IMPLICATIONS: In ghrelin-/- mice, obestatin failed to affect food intake and gastric motility. These results suggest that endogenous ghrelin does not mask the effect of obestatin and confirm that obestatin administered peripherally is not a major regulator of satiety signalling or gut motility.


Subject(s)
Eating/drug effects , Gastric Emptying/drug effects , Ghrelin/pharmacology , Animals , Breath Tests , Caprylates , Cross-Over Studies , Electric Stimulation , Gastric Fundus/drug effects , Gastric Fundus/metabolism , Genotype , Ghrelin/administration & dosage , Ghrelin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction/drug effects , Muscle Contraction/physiology , Satiety Response/drug effects , Satiety Response/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
5.
J Vet Pharmacol Ther ; 30(6): 541-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17991222

ABSTRACT

The present study examines comparatively the effects of theophylline and its metabolites, 1-methylxanthine (1-MX), 3-methylxanthine (3-MX), 1,3-dimethyluric acid (1,3-DMU) and 1-methyluric acid (1-MU) along the rabbit intestine, and explores the underlying mechanism(s). In the small intestine, theophylline produces atropine- and hexamethonium-sensitive increases in both the amplitude of phasic contractions and the basal tone. All metabolites mimic the theophylline's stimulating effect. In particular, concerning the phasic contractions, all metabolites are more potent than theophylline in the duodenum and jejunum, while in the ileum, only 1-MU is more potent. Regarding the basal tone, the metabolites show, in most cases, higher efficacy in all small intestinal regions, the maximum effects of 3-MX and 1-MU on the duodenum and ileum being double or triple the one of theophylline. In the ascending colon, while lower concentrations of theophylline produce an atropine- and hexamethonium-sensitive increase in the basal tone, higher ones produce a postsynaptic, nonadrenergic noncholinergic (NANC) relaxing effect. 1-MU mimics, in a weaker manner, theophylline's effect, while the other metabolites produce only relaxation, the potency rank of order being 3-MX>1-MX=1,3-DMU>theophylline. It is suggested that the theophylline and its metabolites stimulatory effect involves a cholinergic pathway, while the relaxing one is due to 3('),5(')-cyclic adenosine monophosphate (cAMP) elevation mediated by the theophylline and its metabolites inhibitory action on phosphodiesterases (PDEs).


Subject(s)
Bronchodilator Agents/pharmacology , Colon/drug effects , Intestine, Small/drug effects , Theophylline/pharmacology , Animals , Bronchodilator Agents/administration & dosage , Colon/physiology , Dose-Response Relationship, Drug , Female , Intestine, Small/physiology , Male , Muscle Contraction/drug effects , Muscle Contraction/physiology , Rabbits , Theophylline/administration & dosage
6.
Neurogastroenterol Motil ; 19(3): 211-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17300291

ABSTRACT

Obestatin has recently been discovered in the rat stomach. It is encoded by the ghrelin gene and has been claimed to be a functional opponent of ghrelin and to be the natural ligand of the GPR39 receptor. The latter could not be confirmed by Holst et al. (Endocrinology, 2006). Yet, in GPR39 knockout mice, gastric emptying is accelerated. We verified the effects of obestatin on gastric emptying and intestinal contractility in rodents. Gastric emptying was measured with the (14)C octanoic breath test in mice. In vitro, the effect of obestatin was studied on electrically stimulated and non-stimulated strips from the fundus and small intestine of mice and rats. Obestatin (60, 125, 250 nmol kg(-1)) did not affect gastric emptying parameters (T(half) and T(lag)) and did not inhibit the prokinetic effects of ghrelin. Mouse and rat intestinal and fundic smooth muscle strips did not respond to obestatin either in the absence or in the presence of electrical field stimulation. Obestatin (125 nmol kg(-1)) did not inhibit fasting-induced hyperphagia. Our results suggest that peripheral obestatin is not a satiety signal that plays a role in the regulation of gastric emptying and do not support the concept that obestatin is a physiological opponent of ghrelin.


Subject(s)
Gastric Emptying/physiology , Gastrointestinal Motility/physiology , Muscle, Smooth/physiology , Peptide Hormones/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/physiology , Rats , Rats, Wistar
7.
Endocrinology ; 147(6): 2634-42, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16484325

ABSTRACT

Ghrelin is an orexigenic peptide involved in the regulation of energy homeostasis. To investigate the role of ghrelin in the hyperphagia associated with uncontrolled streptozotocin-induced diabetes, food intake was followed in diabetic ghrelin knockout (ghrelin(-/-)) and control wild-type (ghrelin(+/+)) mice and diabetic Naval Medical Research Institute noninbred Swiss mice treated with either saline or the ghrelin receptor antagonist, D-Lys3-GH-releasing peptide-6 (D-Lys3-GHRP-6) for 5 d. In diabetic ghrelin(-/-) mice, hyperphagia was attenuated, and the maximal increase in food intake was 50% lower in mutant than in wild-type mice. The increased food intake observed during the light period (1000-1200 h) in ghrelin(+/+) mice was abolished in mutant mice. Diabetic ghrelin(-/-) mice lost 12.4% more body weight than ghrelin(+/+) mice. In diabetic ghrelin(+/+) mice, but not in ghrelin(-/-) mice, the number of neuropeptide Y (NPY)-immunoreactive neurons was significantly increased. Diabetic Naval Medical Research Institute noninbred Swiss mice were hyperphagic and had increased plasma ghrelin levels. Treatment with D-Lys3-GHRP-6 reduced daily food intake by 23% and reversed the increased food intake observed during the light period. The change in the number of NPY- (2.4-fold increase) and alpha-MSH (1.7-fold decrease)-immunoreactive hypothalamic neurons induced by diabetes was normalized by D-Lys3-GHRP-6 treatment. Our results suggest that enhanced NPY and reduced alpha-MSH expression are secondary to the release of ghrelin, which should be considered the underlying trigger of hyperphagia associated with uncontrolled diabetes.


Subject(s)
Diabetes Mellitus, Experimental/complications , Hyperphagia/etiology , Peptide Hormones/physiology , Animals , Arcuate Nucleus of Hypothalamus/physiology , Blood Glucose/analysis , Body Weight , Ghrelin , Glucagon/blood , Male , Mice , Mice, Inbred C57BL , Neuropeptide Y/analysis , Neuropeptide Y/physiology , Oligopeptides/pharmacology , Peptide Hormones/blood , Streptozocin , alpha-MSH/analysis
9.
J Pharmacol Exp Ther ; 316(1): 431-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16203827

ABSTRACT

To elucidate the role of endogenous ghrelin in the regulation of energy homeostasis and gastric emptying, ghrelin knockout mice (ghrelin(-/-)) were generated. Body weight, food intake, respiratory quotient, and heat production (indirect calorimetry), and gastric emptying ((14)C breath test) were compared between ghrelin(+/+) and ghrelin(-/-) mice. In both strains, the effect of exogenous ghrelin on gastric emptying and food intake was determined. Ghrelin(-/-) mice showed some subtle phenotypic changes. Body weight gain and 24-h food intake were not affected, but interruption of the normal light/dark cycle triggered additional food intake in old ghrelin(+/+) but not in ghrelin(-/-) mice. Exogenous ghrelin increased food intake in both genotypes with a bell-shaped dose-response curve that was shifted to the left in ghrelin(-/-) mice. During the dark period, young ghrelin(-/-) mice had a lower respiratory quotient, whereas their heat production was higher than that of the wild-type littermates, inferring a leaner body composition of the ghrelin(-/-) mice. Absence of ghrelin did not affect gastric emptying, and the bell-shaped dose-response curves of the acceleration of gastric emptying by exogenous ghrelin were not shifted between both strains. In conclusion, ghrelin is not an essential regulator of food intake and gastric emptying, but its loss may be compensated by other redundant inputs. In old mice, meal initiation triggered by the light/dark cue may be related to ghrelin. In young animals, ghrelin seems to be involved in the selection of energy stores and in the partitioning of metabolizable energy between storage and dissipation as heat.


Subject(s)
Energy Metabolism/genetics , Gastric Emptying/genetics , Homeostasis/genetics , Peptide Hormones/genetics , Peptide Hormones/physiology , Aging/physiology , Animals , Body Temperature Regulation/genetics , Body Temperature Regulation/physiology , Body Weight/genetics , Body Weight/physiology , Breath Tests , Calorimetry, Differential Scanning , Caprylates , Eating/drug effects , Eating/genetics , Eating/physiology , Energy Metabolism/physiology , Gastric Emptying/physiology , Gastrointestinal Motility/genetics , Gastrointestinal Motility/physiology , Ghrelin , Homeostasis/physiology , Immunohistochemistry , Mice , Mice, Knockout , Oxygen Consumption/genetics , Oxygen Consumption/physiology , Peptide Hormones/pharmacology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Ghrelin
11.
Gut ; 54(8): 1078-84, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15843418

ABSTRACT

BACKGROUND AND AIMS: The gastroprokinetic activities of ghrelin, the natural ligand of the growth hormone secretagogue receptor (GHS-R), prompted us to compare the effect of ghrelin with that of synthetic peptide (growth hormone releasing peptide 6 (GHRP-6)) and non-peptide (capromorelin) GHS-R agonists both in vivo and in vitro. METHODS: In vivo, the dose dependent effects (1-150 nmol/kg) of ghrelin, GHRP-6, and capromorelin on gastric emptying were measured by the 14C octanoic breath test which was adapted for use in mice. The effect of atropine, N(G)-nitro-L-arginine methyl ester hydrochloride (L-NAME), or D-Lys3-GHRP-6 (GHS-R antagonist) on the gastroprokinetic effect of capromorelin was also investigated. In vitro, the effect of the GHS-R agonists (1 microM) on electrical field stimulation (EFS) induced responses was studied in fundic strips in the absence and presence of L-NAME. RESULTS: Ghrelin, GHRP-6, and capromorelin accelerated gastric emptying in an equipotent manner, with bell-shaped dose-response relationships. In the presence of atropine or l-NAME, which delayed gastric emptying, capromorelin failed to accelerate gastric emptying. D-Lys3-GHRP-6 also delayed gastric emptying but did not effectively block the action of the GHS-R agonists, but this may be related to interactions with other receptors. EFS of fundic strips caused frequency dependent relaxations that were not modified by the GHS-R agonists. L-NAME turned EFS induced relaxations into cholinergic contractions that were enhanced by ghrelin, GHRP-6, and capromorelin. CONCLUSION: The 14C octanoic breath test is a valuable technique to evaluate drug induced effects on gastric emptying in mice. Peptide and non-peptide GHS-R agonists accelerate gastric emptying of solids in an equipotent manner through activation of GHS receptors, possibly located on local cholinergic enteric nerves.


Subject(s)
Oligopeptides/pharmacology , Peptide Hormones/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Stomach/drug effects , Animals , Atropine/pharmacology , Bethanechol/pharmacology , Breath Tests , Cholinergic Agents/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gastric Emptying/drug effects , Ghrelin , Growth Hormone-Releasing Hormone/metabolism , Male , Mice , Mice, Inbred Strains , Muscle Contraction/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Peptide Hormones/agonists
12.
Neurogastroenterol Motil ; 16(4): 439-46, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15305999

ABSTRACT

Ghrelin is an orexigenic peptide with prokinetic effects in the rat. We investigated the effect of ghrelin and growth hormone-releasing hormone 6 (GHRP-6) on gastric emptying and transit in control and septic mice. Mice were injected i.p. with lipopolysaccharides (LPS) or saline (control). After 16-17 h mice were pretreated with saline, ghrelin or GHRP-6 1 h before intragastric administration of Evans blue. Fifteen minutes later, after assessment of the behaviour scale, mice were killed and gastric emptying, transit and rectal temperature were measured. In control mice, ghrelin (100 microg kg(-1)) and GHRP-6 (20-100 microg kg(-1)) accelerated gastric emptying, whereas ghrelin and GHRP-6 failed to increase transit significantly. Septic mice developed a delay in gastric emptying and transit, hypothermia and a deterioration of the behaviour scale. In septic mice, ghrelin (20 microg kg(-1)) accelerated gastric emptying without effect on transit while GHRP-6 significantly accelerated gastric emptying dose-dependently and failed to increase transit significantly. Ghrelin and GHRP-6 had no effect on the endotoxin-induced hypothermia or deterioration of behaviour scale. Therefore, the beneficial prokinetic effect of ghrelin but mainly of GHRP-6 offers potential therapeutic options in the treatment of septic gastric ileus.


Subject(s)
Ileus/drug therapy , Oligopeptides/therapeutic use , Peptide Hormones/therapeutic use , Sepsis/drug therapy , Animals , Gastric Emptying/drug effects , Gastric Emptying/physiology , Ghrelin , Ileus/microbiology , Male , Mice , Oligopeptides/pharmacology , Peptide Hormones/pharmacology , Sepsis/microbiology
13.
Neurogastroenterol Motil ; 15(2): 103-11, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12680909

ABSTRACT

Motilin was infused in this study with the aim of examining refractory characteristics for motilin stimulation of antral phase III and fasting gallbladder emptying. Moreover, interdigestive pyloric and small intestinal motility from duodenum to ileum were studied, as these may be target organs for motilin. Eight fasting, healthy male volunteers received, on separate subsequent days, repeated infusions of 13leucine-motilin (8 pmol (kg min)(-1) for 5 min) or saline at 30 min after phase IIIs in the duodenum. Interdigestive motility of the antrum, pylorus, duodenum, jejunum and ileum was measured for maximum 10 h by using a 21-lumen perfused catheter. Gallbladder motility was measured by ultrasonography. Motilin infusions induced antral phase IIIs, but only after a preceding phase III of duodenal origin. Under this condition, time-interval to phase III at the duodenal recording site was 30 +/- 13 (SEM) min after motilin, compared with 79 +/- 14 min after saline (P < 0.01), and compared with 121 +/- 13 min for motilin infusion following an antral phase III (P < 0.001). Motilin did not affect small intestinal motility or isolated pyloric pressure waves (IPPWs). However, the number of IPPWs was significantly affected by the origin of the preceding phase III, irrespective of whether motilin or saline was infused. Gallbladder volume decreased significantly within 10 min after each motilin infusion. We conclude that this study clearly demonstrates differential regional effects of motilin. Motilin initiates antral phase IIIs, but stimulation is subject to a refractory period which is clearly prolonged after a preceding antral phase III. Motilin induced gallbladder emptying, however, is not subject to a refractory state. Small intestinal phase IIIs as well as pyloric IPPWs are not affected by motilin.


Subject(s)
Gallbladder Emptying/drug effects , Gastrointestinal Motility/drug effects , Motilin/pharmacology , Adult , Cross-Over Studies , Double-Blind Method , Gallbladder Emptying/physiology , Gastrointestinal Motility/physiology , Humans , Infusions, Intravenous , Male , Manometry , Motilin/administration & dosage , Motilin/blood , Ultrasonography
14.
Neurogastroenterol Motil ; 15(2): 149-59, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12680914

ABSTRACT

Intestinal schistosomiasis is accompanied by motility-related dysfunctions but the underlying mechanisms are not well-known. Therefore, the presence and effects on intestinal contractility of somatostatin (SOM) and its receptor, SSTR2A, were investigated in the ileum of normal and infected mice. The distribution of SOM and SSTR2A was visualized using immunocytochemistry. Radioimmunoassay combined with oogram studies was performed to determine SOM levels and contractility measurements were determined in organ bath experiments. Schistosomiasis resulted in a significant decrease in somatostatin-positive endocrine cells, whereas the number of somatostatin-immunoreactive (IR) neuronal cell bodies did not change. From 8 weeks postinfection onwards, an increase was noted in somatostatin-IR nerve fibres in both villi and granulomas. The staining intensity for SSTR2A, expressed in somatostatin-negative myenteric cholinergic neurones, increased during infection suggesting an upregulation of this receptor. SOM levels were negatively correlated with the number of eggs during the acute phase, and were elevated during the chronic phase. Pharmacological experiments revealed that schistosomiasis diminished the inhibitory effect of SOM on neurogenic contractions. We can conclude that schistosomiasis influences the distribution and expression levels of SOM and SSTR2A in the murine ileum, which might explain the changed motility pattern.


Subject(s)
Ileum/parasitology , Receptors, Somatostatin/biosynthesis , Schistosomiasis mansoni/metabolism , Somatostatin/biosynthesis , Adrenergic Fibers/metabolism , Adrenergic Fibers/parasitology , Animals , Cholinergic Fibers/metabolism , Cholinergic Fibers/parasitology , Electric Stimulation , Gastrointestinal Motility/physiology , Ileum/cytology , Ileum/innervation , Ileum/metabolism , Immunohistochemistry , Male , Mice , Muscle Contraction/physiology , Organ Culture Techniques , Radioimmunoassay , Schistosoma mansoni , Schistosomiasis mansoni/physiopathology , Time Factors , Up-Regulation
15.
J Physiol Pharmacol ; 54 Suppl 4: 95-103, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15075452

ABSTRACT

Ghrelin is the recently discovered endogenous ligand for the growth hormone secretagogue receptor. This receptor had previously been characterized based on the stimulatory effect of synthetic peptides, enkephalin analogues, on growth hormone secretion by pituitary somatotrophs. Surprisingly, ghrelin is most abundant in the stomach, suggesting that it may have effects beyond the stimulation of growth hormone in the pituitary and that it is a new brain-gut peptide. There is now increasing evidence that ghrelin stimulates motor activity in the gastrointestinal tract. Thus ghrelin induces the migrating motor complex and accelerates gastric emptying. These are effects typical for motilin, the only peptide structurally related to ghrelin. Moreover, the receptors of both peptides are structurally related as well. The motor effects of ghrelin require rather high concentrations, while motilin at high concentrations stimulates growth hormone release. These data suggest cross-reactivity. However, in vitro binding and contractility studies in the rabbit, the classical model to study motilin agonists, show that ghrelin has very weak if any interaction with the motilin receptor. Similarly, in cell lines expressing the receptors for both peptides there is no evidence for cross-reactivity. This corresponds to the fact that the pharmacophore of both peptides is quite different. Therefore, the motor effects must be due to the stimulation of specific central or peripheral ghrelin receptors. In the guinea pig there is evidence from electrophysiology, immunohistochemistry and calcium imaging studies for ghrelin receptors on myenteric neurons. This provides the morphological basis for peripheral effects of ghrelin. In rats, ghrelin, but not motilin, enhances the response of muscle strips to electrical field stimulation by activating cholinergic pathways. In rabbits the opposite is true but some synthetic ghrelin agonists have weak effects which cannot be blocked by motilin antagonists. Apparently ghrelin is the functional equivalent of motilin in the rat, but in rabbits the motilin-ghrelin family may have yet unknown members. In vivo the effect of ghrelin can be blocked by vagotomy and there is evidence for ghrelin receptors on vagal afferents and in the nodose ganglion. Studies in the rat suggest that under physiological conditions circulating ghrelin does not activate the myenteric plexus, but is able to do so following vagotomy. Finally, centrally administered ghrelin also accelerates gastric emptying and ghrelin changes the activity of neurons of the central nuclei involved in signalling information from the gastrointestinal tract. It is concluded that ghrelin may affect gastrointestinal motility via specific ghrelin receptors located on myenteric, vagal and central neurons. Vagal and central pathways appear to be most important. The fact that ghrelin may reverse the effect of ileus on gastric emptying suggests that ghrelin agonists could find therapeutical application as prokinetics.


Subject(s)
Central Nervous System/physiology , Gastrointestinal Motility/physiology , Ghrelin/physiology , Peripheral Nerves/physiology , Animals , Gastric Emptying/physiology , Humans
16.
Neurogastroenterol Motil ; 14(2): 151-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11975715

ABSTRACT

A plasma motilin peak and a partial gallbladder emptying precede the antral phase III of the migrating motor complex (MMC). To clarify the causal relationship between these factors, we aimed to study the role of motilin in interdigestive gastrointestinal and gallbladder motility simultaneously. In addition, involvement of 5HT3 receptors in the action of motilin was studied. Eight fasting, healthy male volunteers received 13Leu-motilin or 0.9% NaCl i.v. for 30 min, in randomized order on two separate occasions, from 30 min after phase III. Seven of the eight subjects also received the 5HT3 receptor antagonist ondansetron in addition to motilin, on a third occasion. Antroduodenal motility, gallbladder volumes and plasma motilin were measured. The interval between the start of infusion and phase III was 95.0 (57.6-155.7) min for saline, 28.7 (21.0-33.2) min for motilin, and 39.3 (30.7-100.5) min for motilin + ondansetron (P < 0.05). Gallbladder volume decreased by one-third from 10 min after both motilin and motilin + ondansetron infusion (P < 0.05), and returned to baseline with duodenal passage of phase III. In two of the seven subjects phase III was absent after motilin + ondansetron, although gallbladder volume decreased and only refilled during a later spontaneous phase III. We conclude that motilin induces both partial gallbladder emptying and antral phase III. Indeed, although gallbladder emptying clearly precedes antral phase III, ondansetron only prevented phase III in some cases and had no effect on gallbladder emptying. Passage of phase III in the duodenum makes an important contribution to gallbladder refilling.


Subject(s)
Gallbladder/drug effects , Gastrointestinal Motility/drug effects , Motilin/pharmacology , Receptors, Serotonin/physiology , Adult , Analysis of Variance , Cross-Over Studies , Double-Blind Method , Gallbladder/physiology , Gastrointestinal Motility/physiology , Humans , Male , Motilin/blood , Motilin/physiology , Ondansetron/pharmacology , Receptors, Serotonin, 5-HT3 , Serotonin Antagonists/pharmacology
17.
Curr Opin Investig Drugs ; 2(4): 555-7, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11566017

ABSTRACT

GM-611 is an erythromycin derivative that acts as an agonist at the motilin receptor. It is being developed by Chugai as a potential treatment for gastric motility disorder [169036], as well as reflux esophagitis, non-ulcer dyspepsia and diabetic gastroparesis [347963]. GM-611 is in phase II trials in the US for reflux esophagitis [322624], [347955], [399349]. GM-611 acts by a novel mechanism whereby it stimulates and promotes peristalsis in the stomach and other segments of the gastrointestinal tract [334994]. The drug was shown to produce a dose-dependent sustained depolarization of rabbit duodenal smooth muscle. Depolarization appeared to be associated with activation of monovalent cation-selective channels [273336]. In December 2000, Credit Suisse First Boston predicted that successful development of GM-611 could lead to sales over $500 million [400228].


Subject(s)
Erythromycin/analogs & derivatives , Erythromycin/pharmacology , Gastrointestinal Motility/drug effects , Receptors, Gastrointestinal Hormone/agonists , Receptors, Neuropeptide/agonists , Animals , Clinical Trials as Topic , Erythromycin/chemical synthesis , Humans , Structure-Activity Relationship
18.
Dig Dis Sci ; 46(6): 1328-34, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11414312

ABSTRACT

The role of interdigestive gallbladder emptying in gallstone formation is unknown. In fasting healthy subjects, gallbladder emptying is associated with antral phase III of the migrating motor complex (MMC) and high plasma motilin. Therefore, gallbladder volumes and motilin levels were measured during 13 MMC cycles in 10 cholesterol gallstone patients and compared with 20 MMC cycles in 10 healthy subjects. MMC cycle length was longer in gallstone patients than in healthy subjects (158.2 +/- 17.0 vs 105.5 +/- 10.4 min, respectively; P < 0.05), due to longer phase I (39.8 +/- 5.7 vs 17.2 +/- 3.7 min, respectively; P < 0.05). In contrast to healthy subjects, gallstone patients had no significant fluctuations of gallbladder volume during the MMC cycle, and motilin concentrations were not different in MMC cycles with phase III originating in antrum or duodenum. During MMC cycles with phase III originating in the duodenum, motilin levels were twice as high in gallstone patients as in healthy subjects (P < 0.002). In conclusion, cholesterol gallstone patients have an abnormal MMC and motilin release pattern. Their interdigestive gallbladder emptying is reduced and dissociated from the MMC. These disturbances may contribute to gallstone formation.


Subject(s)
Cholelithiasis/physiopathology , Duodenum/physiopathology , Gallbladder Emptying/physiology , Gastrointestinal Motility/physiology , Motilin/blood , Cholelithiasis/blood , Cholelithiasis/chemistry , Cholesterol/analysis , Digestion , Female , Humans , Male , Middle Aged , Pyloric Antrum
19.
Digestion ; 63(3): 195-202, 2001.
Article in English | MEDLINE | ID: mdl-11351147

ABSTRACT

BACKGROUND/AIMS: The enterohepatic circulation of bile acids is related to normal inter-digestive gastrointestinal motility, with the gut peptide motilin also being involved. This study aimed to investigate the effect of intraduodenal artificial bile infusion on antroduodenal and gallbladder motility so as to further elucidate the controlling factors. METHODS: Twelve fasting, healthy male volunteers received artificial bile (80 mol% bile acids; 15 mol% phospholipids; 5 mol% cholesterol) or placebo (saline) intraduodenally for 10 min starting 30 min after the end of phase III, according to a double-blind, randomised, cross-over design. Antroduodenal motility, gallbladder volumes, and plasma motilin levels were measured. All values are means +/- SEM. RESULTS: The interval between infusion and the subsequent phase III, as well as the origin of this phase III were not significantly different between bile and saline. Antral pressure waves were significantly more frequent during and immediately after bile infusion compared with saline infusion (p < 0.05). The duration of phase I following infusion was significantly longer after bile (24.8 +/- 3.7 min) than after saline infusion (13.1 +/- 1.7 min; p < 0.05). The mean gallbladder volume tended to increase in the hours following bile infusion, but to decrease after saline infusion (p = 0.06). Plasma motilin increased after bile and saline infusion in an almost identical way. CONCLUSION: This study provides no clear evidence for a role of intraduodenal artificial bile (i.e. its main constituents) in the regulation of migrating motor complex cycling or feedback inhibition of inter-digestive gallbladder emptying.


Subject(s)
Bile Acids and Salts/pharmacology , Gallbladder Emptying/drug effects , Gastrointestinal Motility/drug effects , Motilin/blood , Adult , Bile Acids and Salts/metabolism , Cross-Over Studies , Double-Blind Method , Duodenum/drug effects , Duodenum/physiology , Gallbladder Emptying/physiology , Gastrointestinal Motility/physiology , Humans , Injections, Intralesional , Male , Manometry , Probability , Reference Values , Sensitivity and Specificity
20.
Brain Res ; 895(1-2): 119-28, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11259768

ABSTRACT

BACKGROUND: Our laboratory has described the presence of motilin receptors in the rabbit cerebellum. We discovered its presence in the human TE671 cell line, which is of cerebellar origin. METHODS: Cytosolic Ca(2+) fluxes were monitored on a confocal microscope in cells loaded with Indo-1 and stimulated with motilin under various conditions. Binding studies were performed with 125I-[Nle(13)]porcine motilin. Using primers, PCR for the motilin receptor was performed. RESULTS: Cells responded to motilin after 45+/-20 s. At different concentrations of motilin (10(-8), 10(-7), 10(-6.5), 10(-6) and 10(-5) M) the percentage of responding cells was 0+/-0, 0.6+/-1.5, 4.9+/-4.7, 21.7+/-15 and 35.7+/-12, respectively. The response was blocked by the motilin antagonists [Phe(3), Nle(13)]po-motilin (0.8+/-1.8%) and GM-109 (0.0+/-0.0%) and mimicked by the agonist ABT-229 (23.6+/-15%). After stimulation with motilin, ABT-229 or [Phe(3),Leu(13)]po-motilin, but not with the antagonist GM-109, cells were desensitized. The response to motilin persisted in Ca(2+)-free solution (22.8+/-14.7%), was not affected by nifedipine (44+/-11%) but was abolished by incubation with thapsigargin (0+/-0%). Neither ryanodine, nor a previous stimulation with caffeine (0+/-0%) in Ca(2+)-free Krebs, nor both could block the response to motilin (28, 32.0+/-5.7, 41.3+/-6.1%, respectively). Binding studies revealed two binding sites for motilin, with a pK(d) of 8.9+/-0.05 and 6.11+/-0.61 (n=4). There were 100 times more low than high affinity receptors per cell. The presence of receptor mRNA was confirmed by PCR. CONCLUSION: Functional motilin receptors are present in TE671 cells. The response requires intracellular IP(3)-sensitive Ca(2+) stores. These cells may serve as a model of the central motilin receptor.


Subject(s)
Cerebellum/metabolism , Motilin/metabolism , Neurons/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Neuropeptide/metabolism , Tumor Cells, Cultured/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cerebellum/cytology , Cerebellum/drug effects , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Drug Interactions/physiology , Humans , Iodine Radioisotopes/pharmacology , Medulloblastoma , Motilin/agonists , Motilin/antagonists & inhibitors , Neurons/cytology , Neurons/drug effects , Norleucine/pharmacology , Radioligand Assay , Receptors, Gastrointestinal Hormone/drug effects , Receptors, Neuropeptide/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Tachyphylaxis/physiology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...