Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
2.
Curr Pharm Des ; 20(30): 4807-15, 2014.
Article in English | MEDLINE | ID: mdl-24251671

ABSTRACT

Melatonin is a potent reactive oxygen metabolite scavenger and antioxidant that has been shown to influence many physiological functions of the gastrointestinal (GI) tract including secretion, motility, digestion and absorption of nutrients. The role of melatonin in gastroduodenal defense and ulcer healing has been the subject of recent investigations. Melatonin produced in the GI mucosa plays an important role in protection against noxious agents thus contributing to the maintenance of GI integrity and to esophageal protection, gastroprotection and ulcer healing. This review was designed to summarize the involvement of melatonin, conventionally considered as a major hormone of the pineal gland, in the maintenance of gastric mucosal integrity, gastroprotection, ulcer healing and intestinal disorders. Melatonin was originally shown to attenuate gastric mucosal lesions but controversy exists in the literature as to whether melatonin derived from the pineal gland, considered as the major source of this indole, or rather gastrointestinal melatonin plays predominant role in gastroprotection. Intragastric and central administration of exogenous melatonin and L-tryptophan, this indoleamine precursor, affords protection against gastric hemorrhagic damage caused by the exposure of gastric mucosa to variety of non-topical and topical ulcerogens such as stress, ethanol and ischemia-reperfusion. The speed of ulcer healing in experimental animals and humans is accelerated by melatonin. This indoleamine could be also effective against the esophageal lesions provoked by reflux esophagitis in animal models and prevents the incidence of GERD in humans. The melatonin-induced gastroprotection is accompanied by an increase in gastric blood flow, plasma melatonin concentration, enhancement in mucosal generation of PGE2, luminal NO content and plasma gastrin levels. Melatonin scavenges reactive oxygen metabolites, exerts anti-oxidizing and anti-inflammatory actions and inhibits the formation of metalloproteinases- 3 and -9; both implicated in the pathogenesis of gastrointestinal injury and formation of gastric ulcers. Blockade of MT2 receptors by luzindole, significantly attenuated melatonin- and L-tryptophan-induced protection and increased the speed of ulcer healing and these effects were accompanied by an increase in the GBF and luminal content of NO suggesting that melatonin exhibits gastroprotection and hyperemia via activation of MT2 receptors and release of NO. The accumulated evidence indicates that the melatonin-induced gastroprotection and the enhancement in healing rate of gastric ulcers may involve the gastroprotective factors derived from the activation of PG/COX and NO/NOS systems as well as gastrin which also was shown to exhibit protective and trophic effects in the upper GItract. Interestingly, pinealectomy, which suppressed plasma melatonin levels, markedly exacerbated gastric lesions induced by topical and non-topical ulcerogens and these effects are counteracted by a concurrent supplementation with melatonin. Evidence is provided that exogenous melatonin and that converted from its precursor, L-tryptophan, attenuates acute gastric lesions and accelerates ulcer healing via interaction with MT2 receptors due to an enhancement of gastric microcirculation, probably mediated by NO and PG derived from NOS and COX-1 and COX-2 overexpression and activity. The pineal gland plays an important role in the limitation of gastric mucosal injury and the acceleration of ulcer healing via releasing endogenous melatonin, which attenuates oxidative stress and exerts anti-inflammatory action.


Subject(s)
Esophagus/drug effects , Gastroesophageal Reflux/drug therapy , Melatonin/therapeutic use , Peptic Ulcer/drug therapy , Humans , Melatonin/pharmacology
3.
Peptides ; 49: 9-20, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23978788

ABSTRACT

Nesfatin-1 belongs to a family of anorexigenic peptides, which are responsible for satiety and are identified in the neurons and endocrine cells within the gut. These peptides have been implicated in the control of food intake; however, very little is known concerning its contribution to gastric secretion and gastric mucosal integrity. In this study the effects of nesfatin-1 on gastric secretion and gastric lesions induced in rats by 3.5h of water immersion and restraint stress (WRS) were determined. Exogenous nesfatin-1 (5-40µg/kg i.p.) significantly decreased gastric acid secretion and attenuated gastric lesions induced by WRS, and this was accompanied by a significant rise in plasma NUCB2/nefatin-1 levels, the gastric mucosal blood flow (GBF), luminal NO concentration, generation of PGE2 in the gastric mucosa, an overexpression of mRNA for NUBC2 and cNOS, as well as a suppression of iNOS and proinflammatory cytokine IL-1ß and TNF-α mRNAs. Nesfatin-1-induced protection was attenuated by suppression of COX-1 and COX-2 activity, the inhibition of NOS with L-NNA, the deactivation of afferent nerves with neurotoxic doses of capsaicin, and the pretreatment with capsazepine to inhibit vanilloid VR1 receptors. This study shows for the first time that nesfatin-1 exerts a potent protective action in the stomach of rats exposed to WRS and these effects depend upon decrease in gastric secretion, hyperemia mediated by COX-PG and NOS-NO systems, the activation of vagal and sensory nerves and vanilloid receptors.


Subject(s)
Calcium-Binding Proteins/pharmacology , DNA-Binding Proteins/pharmacology , Gastric Mucosa/drug effects , Nerve Tissue Proteins/pharmacology , Nitric Oxide/physiology , Prostaglandins/physiology , Satiation/drug effects , Stress, Physiological , TRPV Cation Channels/physiology , Animals , Blotting, Western , Calcium-Binding Proteins/administration & dosage , DNA-Binding Proteins/administration & dosage , Gastric Acid/metabolism , Gastric Mucosa/injuries , Gastric Mucosa/innervation , Gastrins/blood , Injections, Intraventricular , Nerve Tissue Proteins/administration & dosage , Nucleobindins , Rats , Reverse Transcriptase Polymerase Chain Reaction
4.
Pharmacol Rep ; 65(2): 494-504, 2013.
Article in English | MEDLINE | ID: mdl-23744434

ABSTRACT

BACKGROUND: Serotonin (5-HT) is released from enterochromaffin cells in the gastrointestinal tract. 5-HT, via the activation of 5-HT2 and 5-HT3 receptors on vagal fibers, mediates pancreatic secretion through the mechanism independent from cholecystokinin. Melatonin (5-HT derivative) or L-tryptophan (melatonin or 5-HT precursor) given systemically or intraduodenally to the rats stimulate amylase secretion, but the mechanism is not clear. The aim of this study was to investigate the involvement of 5-HT in the pancreatostimulatory effect of melatonin or L-tryptophan, administered intraduodenally. METHODS: Wistar rats were surgically equipped with silicone catheters; inserted into pancreato-biliary duct and into the duodenum. Melatonin, L-tryptophan or 5-HT were given to the rats as a bolus. Combination of 5-HT2 or 5-HT3 receptor antagonists: ketanserin (100 µg/kg) and MDL72222 (250 µg/kg) was given intraperitoneally to the animals, 15 min. prior to the administration of the examined substances. The role of the vagal nerve, sensory fibers and CCK in the control of pancreatic exocrine function were determined. Blood samples were taken for the determination of 5-HT. RESULTS: Melatonin, 5-HT or L-tryptophan increased pancreatic amylase secretion. The stimulatory effect of the above substances was decreased by pretreatment of the rats with ketanserin and MDL72222. Bilateral vagotomy completely abolished the increase of amylase output caused by 5-HT, while capsaicin deactivation of sensory nerves or blockade of CCK1 receptor only partially reversed the stimulatory effect of 5-HT on the pancreas. Intraduodenal L-tryptophan, but not melatonin, increased plasma 5-HT concentrations in a dose- and time-dependent manner. CONCLUSION: Stimulation of pancreatic exocrine function caused by intraluminal administration of melatonin, or L-tryptophan is modified, at least in part, by serotoninergic mechanisms and vagal nerves.


Subject(s)
Amylases/metabolism , Melatonin/pharmacology , Serotonin/metabolism , Tryptophan/pharmacology , Animals , Dose-Response Relationship, Drug , Duodenum/metabolism , Ketanserin/pharmacology , Melatonin/administration & dosage , Pancreas/drug effects , Pancreas/enzymology , Rats , Rats, Wistar , Receptor, Cholecystokinin A/metabolism , Serotonin Antagonists/pharmacology , Time Factors , Tropanes/pharmacology , Tryptophan/administration & dosage , Vagus Nerve/metabolism
5.
J Pineal Res ; 55(1): 46-57, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23480366

ABSTRACT

Reflux esophagitis is a common clinical entity in western countries with approximately 30% of the population experiencing the symptoms at least once every month. The imbalance between the protective and aggressive factors leads to inflammation and damage of the esophageal mucosa. We compared the effect of exogenous melatonin and melatonin derived endogenously from L-tryptophan with that of pantoprazole or ranitidine in acid reflux esophagitis due to ligation of the rat pylorus and the limiting ridge between the forestomach and the corpus. Four hours after the induction of gastric reflux, an increase in mucosal lesions associated with edema of the submucosa and with the infiltration of numerous neutrophils and the fall in esophageal blood flow (EBF) were observed. Both melatonin and L-tryptophan or pantoprazole significantly reduced the lesion index (LI) and raised the EBF. Pinealectomy that significantly decreased plasma melatonin levels aggravated LI and these effects were reduced by melatonin and L-tryptophan. Luzindole, the MT2 receptor antagonist, abolished the melatonin-induced reduction in LI and the rise in EBF. L-NNA and capsaicin that augmented LI and decreased EBF, also significantly reduced melatonin-induced protection and hyperemia; both were restored with L-arginine and calcitonin gene-related peptide (CGRP) added to melatonin. Upregulation of IL-1ß and TNF-α mRNAs and plasma IL-1ß and TNF-α levels were significantly attenuated by melatonin and L-tryptophan. We conclude that melatonin protects against acid reflux-induced damage via activation of MT2 receptors mediated by NO and CGRP released from sensory nerves and the suppression of expression and release of TNF-α and IL-1ß.


Subject(s)
Esophagitis, Peptic/pathology , Melatonin/pharmacology , Protective Agents/pharmacology , Analysis of Variance , Animals , Arginine/pharmacology , Capsaicin/pharmacology , Digestive System Surgical Procedures , Disease Models, Animal , Esophagus/blood supply , Esophagus/drug effects , Esophagus/pathology , Male , Melatonin/metabolism , Mucous Membrane/pathology , Nitric Oxide/metabolism , Pineal Gland/surgery , Protective Agents/metabolism , Rats , Rats, Wistar , Tryptophan/pharmacology
6.
J Pineal Res ; 54(2): 154-61, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22804755

ABSTRACT

Insulin resistance, oxidative stress, and an abnormal production of adipokines and cytokines are implicated in the pathogenesis of nonalcoholic steatohepatitis (NASH). Recently, we reported a significant improvement in plasma liver enzymes among patients with NASH treated with melatonin. In this study, we investigated the effect of melatonin, administered at a dose of 10 mg/day for 28 days to 16 patients with histologically proven NASH on insulin resistance (HOMA-IR), on the plasma levels of adiponectin, leptin, ghrelin, and resistin. Additionally, plasma levels of aminotransferases and gamma glutamyltranspeptidase as well as plasma concentrations of melatonin were evaluated. Median baseline values of HOMA-IR, leptin (ng/mL), and resistin (pg/mL) in patients with NASH were significantly higher in comparison with controls: 4.90 versus 1.60, 10.70 versus 4.30, and 152 versus 91, respectively. Median adiponectin level (µg/mL) was decreased in patients compared to controls: 6.40 versus 16.25; no significant difference in ghrelin levels between patients and controls was found. After melatonin treatment, the median value of HOMA-IR was significantly reduced by 60% as compared to baseline values, whereas adiponectin, leptin, and ghrelin plasma levels rose significantly by 119%, 33%, and 20%, respectively; the difference between pre-/posttreatment in plasma resistin levels was not significant. These findings make melatonin a suitable candidate for testing in patients with NASH in the large controlled clinical trials.


Subject(s)
Adiponectin/blood , Fatty Liver/blood , Fatty Liver/drug therapy , Ghrelin/blood , Insulin/blood , Leptin/blood , Melatonin/therapeutic use , Resistin/blood , Adult , Female , Humans , Male
7.
Int J Inflam ; 2012: 354904, 2012.
Article in English | MEDLINE | ID: mdl-22685683

ABSTRACT

Introduction. Lipopolysaccharide endotoxin (LPS) is responsible for septic shock and multiorgan failure, but pretreatment of rats with low doses of LPS reduced pancreatic acute damage. Aim. We investigated the effects of the endotoxemia induced in the early period of life on Toll-like receptor 4 (TLR4), heat shock protein 60 (HSP60) and proapoptotic Bax, caspase-9 and -3 or antiapoptotic Bcl-2 protein expression in the pancreatic acinar cells of adult animals. Material and Methods. Newborn rats (25 g) were injected with endotoxin (Escherichia coli) for 5 consecutive days. Two months later, pancreatic acinar cells were isolated from all groups of animals and subjected to caerulein stimulation (10(-8) M). Protein expression was assessed employing Western blot. For detection of apoptosis we have employed DNA fragmentation ladder assay. Results. Preconditioning of newborn rats with LPS increased TLR4, Caspase-9 and -3 levels, but failed to affect basal expression of HSP60, Bax, and Bcl-2. Subsequent caerulein stimulation increased TLR4, Bcl-2, and caspases, but diminished HSP60 and Bax proteins in pancreatic acinar cells. Endotoxemia dose-dependently increased TLR4, Bax, HSP60, and both caspases protein signals in the pancreatic acini, further inhibiting antiapoptotic Bcl-2. Conclusions. Endotoxemia promoted the induction of HSP60 via TLR4 in the infant rats and participated in the LPS-dependent pancreatic tissue protection against acute damage.

8.
Curr Pharm Des ; 17(16): 1541-51, 2011.
Article in English | MEDLINE | ID: mdl-21548865

ABSTRACT

Previous studies revealed that prostaglandins contribute to the mechanism of maintenance of gastrointestinal integrity and mediate various physiological aspects of mucosal defense. The suppression of prostaglandin synthesis in the stomach is a critical event in terms of the development of mucosal injury after administration of various NSAID including aspirin (ASA). A worldwide use of ASA is now accepted due to its remarkable analgesic, antipyretic and anti-thrombotic prophylactics against myocardial infarct and coronary disorders despite the fact that the use of NSAIDs is associated with the risk of gastrointestinal bleedings, haemorrhagic lesions and ulcerations. It has become clear that other mediators besides prostaglandins can similarly act to protect the gastrointestinal mucosa of experimental animals and humans from injury induced by ASA. For instance, nitric oxide (NO) released from vascular epithelium, epithelial cells of gastrointestinal tract and sensory nerves can influence many of the same components of mucosal defense as do prostaglandins. This review was designed to provide an updated overview based on the experimental and clinical evidence on the involvement COX-2 derived products, lipoxins in the mechanism of gastric defense, gastroprotection and gastric adaptation to ASA. Lipoxins were recently considered as another group of lipid mediators that can protect the stomach similarly as NO-donors known to exert protective influence on the stomach from the injury under condition where the mucosal prostaglandin levels are suppressed. The new class of NO-releasing NSAIDs, including NO-aspirin or NO-naproxen, represent a very promising approach to reducing the toxicity of their parent NSAIDs. Aspirin-triggered lipoxin (ATL) synthesis, via COX-2, acts to reduce the severity of damage induced by this NSAID. Lipoxin analogues may prove to be useful for preventing mucosal injury and for modulating mucosal inflammation. Evidence presented in this review documents that ATL also play in important role in gastric adaptation during chronic ASA administration. Suppression of COX-2 activity by selective COX-2 inhibitors such as rofecoxib or celecoxib was shown to abolish the production of ATL and to diminish the gastric tolerability of ASA and gastric adaptation developed in response to repetitive administration of this NSAID. Synthetic analogues of lipoxins as well as newer class of NSAIDs releasing NO may be used in the future as the therapeutic approach to counteract adverse effects in the stomach associated with NSAIDs ingestion.


Subject(s)
Adaptation, Physiological , Aspirin/adverse effects , Lipoxins/physiology , Stomach Ulcer/prevention & control , Stomach/drug effects , Humans , Stomach/physiopathology , Stomach Ulcer/physiopathology
9.
J Pineal Res ; 50(4): 389-94, 2011 May.
Article in English | MEDLINE | ID: mdl-21362032

ABSTRACT

Melatonin and L-tryptophan (Trp) are highly gastroprotective in humans, but no study has assessed their impact on healing of chronic gastroduodenal ulcers in humans. Three groups (A, B and C) of 14 idiopathic patients in each treatment group with gastroduodenal chronic ulcers were treated with omeprazole (20 mg twice daily) combined either with placebo (group A), melatonin (group B) or with Trp (group C). The rate of ulcer healing was determined by gastroduodenoscopy at day 0, 7, 14 and 21 after initiation of therapy. Plasma melatonin, gastrin, ghrelin and leptin were measured by RIA. On day 7, omeprazole by itself (group A) had not healed any ulcers, but four ulcers were healed with omeprazole plus melatonin and two with omeprazole plus tryptophan. At day 21, all ulcers were healed in patients treated with melatonin or Trp, but only 10-12 ulcers were healed in placebo-treated patients. After treatment with omeprazole plus melatonin (group B) or Trp (group C), plasma melatonin levels rose several-fold above initial values. Plasma gastrin level also rose significantly during treatment with omeprazole plus melatonin or Trp, but it was also significantly increased in patients treated with omeprazole plus placebo. Plasma ghrelin levels did not change significantly after treatment with melatonin or Trp, while plasma leptin increased significantly in patients treated with melatonin or Trp but not with placebo. We conclude that melatonin or Trp, when added to omeprazole treatment, accelerates ulcer healing and this likely depends mainly upon the significant increments in plasma melatonin.


Subject(s)
Melatonin/therapeutic use , Omeprazole/therapeutic use , Peptic Ulcer/drug therapy , Tryptophan/therapeutic use , Adult , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Radioimmunoassay
10.
World J Gastroenterol ; 17(4): 449-58, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21274374

ABSTRACT

AIM: To investigate plasma ghrelin, gastrin and growth hormone secretagogue receptor (GHS-R) expression in advanced gastric cancer (GC) before and after resection. METHODS: Seventy subjects in whom endoscopy of the upper gastrointestinal tract was performed in the Department of General Surgery at Cracow University during the past decade: (1) 25 patients with GC associated with Helicobacter pylori (H. pylori) infection; (2) 10 patients with GC 4-5 years after (total or subtotal) gastrectomy; (3) 25 healthy H. pylori-negative controls, matched by age and BMI to the above two groups; and (4) 10 GC patients 4-5 years after total gastrectomy. Ghrelin and gastrin plasma concentrations were measured by specific radioimmunoassay under fasting conditions and postprandially at 60 and 90 min after ingestion of a mixed meal. GHS-R expression was examined in biopsy samples from intact healthy mucosa and GC tissue using semi-quantitative reverse transcription-polymerase chain reaction. RESULTS: In healthy controls, fasting plasma ghrelin levels were significantly elevated and declined markedly at 60 and 90 min after a mixed meal. The concomitant enhanced ghrelin, GHS-R and gastrin expression in GC tissue over that recorded in intact mucosa, and the marked rise in plasma gastrin in these subjects under fasting conditions indicate the role of these hormonal factors in GC formation. Fasting plasma levels and postprandial response of ghrelin and gastrin appear to be inversely correlated in healthy subjects. Feeding in the controls resulted in a significant fall in plasma ghrelin with a subsequent rise in plasma gastrin, but in H. pylori-positive GC patients submitted to total or distal gastrectomy, feeding failed to affect significantly the fall in plasma ghrelin that was recorded in these patients before surgery. Fasting ghrelin concentrations were significantly lower in patients 4-5 years after total gastrectomy compared to those in healthy controls and to these in GC patients before surgery. CONCLUSION: Elevated plasma gastrin and suppression of fasting ghrelin in patients with GC suggest the existence of a close relationship between these two hormones in gastric carcinogenesis.


Subject(s)
Gastrectomy , Gastrins/blood , Ghrelin/blood , Stomach Neoplasms/blood , Stomach Neoplasms/surgery , Adult , Aged , Aged, 80 and over , Fasting/blood , Female , Helicobacter Infections/blood , Humans , Male , Middle Aged , Postprandial Period , Radioimmunoassay , Receptors, Ghrelin/blood , Stomach Neoplasms/pathology
11.
Curr Protoc Toxicol ; Chapter 21: Unit 21.6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20967748

ABSTRACT

The protocols described in this unit are designed to present the major endogenous gastric mediators involved in the control of gastric acid secretion, namely gastrin and histamine, and in the regulation of gastric motility, which include motilin and ghrelin, under physiological and pathological conditions. The measurement of these mediators in plasma or serum of humans and animals by radioimmunoassay are described and their pathophysiological role is discussed.


Subject(s)
Gastric Acid/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/physiopathology , Gastrins/blood , Gastrins/physiology , Histamine/blood , Histamine/physiology , Animals , Ghrelin/blood , Humans , Motilin/blood , Radioimmunoassay/methods , Toxicology/methods
12.
Med Sci Monit ; 16(10): CR493-500, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20885354

ABSTRACT

BACKGROUND: Ghrelin is a hormone produced by neuroendocrine cells of gastric mucosa. Its concentration changes in cases of Helicobacter pylori (H. pylori) infection of stomach, but the relation between the expression of plasma and mucosal ghrelin content and H. pylori infection is not completely defined. This study was designed to determine the changes in plasma and gastric mucosa ghrelin concentrations in diseases of gastric mucosa depending upon the H. pylori infection. MATERIAL/METHODS: The following groups were included; Group 1. patients with gastric cancer and concomitant H. pylori infection (N=25); Group 2. patients with antral gastritis with H. pylori infection and concomitant duodenal peptic ulcer (N=18); Group 3. patients with atrophic gastritis of both the fundus and the body of the stomach without H. pylori infection (N=10); Group 4. control group consisting of patients without morphological and histological lesions of gastric mucosa and without H. pylori infection (n=25). Endoscopic biopsies of gastric mucosa of fundus, body and pyloric region were obtained in all tested groups. In patients suffering from gastric cancer biopsies of tumor were also taken. The ghrelin concentrations were measured by specific RIA. Biopsy specimens were examined to assess ghrelin mRNA expression in intact gastric mucosa and gastric cancer. RESULTS: The study showed significant influence of H. pylori infection on ghrelin plasma concentrations. The highest ghrelin concentrations were found in patients of the group 2 (average 503 pg/mL (95% CI: 285-886). Ghrelin concentrations were found to fall to the lowest values in the group 3 (average 144 pg/mL (95% CI: 93-222). In the group 1, the ghrelin concentration averaged 203 pg/mL (95% CI: 161-257), while in the group 4 (control group) - 255 pg/mL; 95% (CI: 160-406). The study proved that gastric cancer does not exert any ghrelin-production activity, as confirmed by RT-PCR examination of biopsy specimens of the cancer. CONCLUSIONS: This study shows that the presence of H. pylori in the stomach with peptic ulcer increases plasma ghrelin levels, whereas in gastric cancer and atrophic gastritis it is accompanied by a marked decrease in plasma and cancer tissue levels of ghrelin.


Subject(s)
Gastric Mucosa/pathology , Ghrelin/blood , Ghrelin/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori , Adult , Biopsy , Case-Control Studies , Duodenal Ulcer/complications , Duodenal Ulcer/metabolism , Duodenal Ulcer/pathology , Female , Gastric Fundus/metabolism , Gastric Fundus/pathology , Gastric Mucosa/metabolism , Gastritis/metabolism , Gastritis/pathology , Gastritis, Atrophic/complications , Gastritis, Atrophic/metabolism , Gastritis, Atrophic/pathology , Helicobacter Infections/complications , Helicobacter Infections/pathology , Humans , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radioimmunoassay , Reverse Transcriptase Polymerase Chain Reaction , Stomach/pathology , Stomach Neoplasms/complications , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
13.
Curr Pharm Des ; 16(10): 1214-23, 2010.
Article in English | MEDLINE | ID: mdl-20166991

ABSTRACT

Orexigenic peptides are group of endocrine hormones exerting a pleiotropic influence on many physiological functions including regulation of the feeding behaviour and energy expenditure, release of growth hormone (GH) and inotropic effects on the heart. Some of these peptides such as ghrelin, originally identified in the gastric mucosa, has been involved not only in control of food intake and growth hormone release but also exerts the immunomodulatory and anti-inflammatory properties. This review summarizes the recent attempts to prove the concept that orexigenic peptides such as ghrelin, orexin-A and obestatin besides playing an important role in the mechanism of food intake, exhibit a potent gastroprotective action against the formation of acute gastric mucosal injury induced by various ulcerogens. This protective effect depends upon vagal activity and hyperemia mediated by NOS/NO and COX/PG systems and CGRP released from sensory afferent nerves. In addition, the appetite peptides such as ghrelin and orexin-A are implicated in the mechanism of the healing of preexisting gastric ulcers due to an activation of specific GHS-R1a and OX-R1 receptors and PG/COX system.


Subject(s)
Appetite Stimulants/pharmacology , Gastric Mucosa/drug effects , Ghrelin/pharmacology , Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptides/pharmacology , Stomach Ulcer/drug therapy , Animals , Cyclooxygenase 2 Inhibitors/pharmacology , Gastric Mucosa/blood supply , Gastric Mucosa/innervation , Ghrelin/administration & dosage , Ghrelin/physiology , Helicobacter Infections/pathology , Helicobacter pylori , Humans , Intracellular Signaling Peptides and Proteins/administration & dosage , Intracellular Signaling Peptides and Proteins/physiology , Neuropeptides/administration & dosage , Neuropeptides/physiology , Neurotransmitter Agents/pharmacology , Orexins , Rats , Stomach Ulcer/pathology
14.
Regul Pept ; 148(1-3): 6-20, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18378017

ABSTRACT

Orexin-A, identified in the neurons and endocrine cells in the gut, has been implicated in control of food intake and sleep behavior but little is known about its influence on gastric secretion and mucosal integrity. The effects of orexin-A on gastric secretion and gastric lesions induced in rats by 3.5 h of water immersion and restraint stress (WRS) or 75% ethanol were determined. Orexin-A (5-80 microg/kg i.p.) increased gastric acid secretion and attenuated gastric lesions induced by WRS and this was accompanied by the significant rise in plasma orexin-A, CGRP and gastrin levels, the gastric mucosal blood flow (GBF), luminal NO concentration and an increase in mRNA for CGRP and overexpression of COX-2 protein and the generation of PGE(2) in the gastric mucosa. Orexin-A-induced protection was abolished by selective OX-1 receptor antagonist, vagotomy and attenuated by suppression of COX-1 and COX-2, deactivation of afferent nerves with neurotoxic dose of capsaicin, pretreatment with CCK(2)/gastrin antagonist, CGRP(8-37) or capsazepine and by inhibition of NOS with L-NNA. This study shows for the first time that orexin-A exerts a potent protective action on the stomach of rats exposed to non-topical ulcerogens such as WRS or topical noxious agents such as ethanol and these effects depend upon hyperemia mediated by COX-PG and NOS-NO systems, activation of vagal nerves and sensory neuropeptides such as CGRP released from sensory nerves probably triggered by an increase in gastric acid secretion induced by this peptide.


Subject(s)
Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptides/metabolism , Nitric Oxide/metabolism , Prostaglandins/metabolism , Stomach Diseases/prevention & control , Stress, Physiological/physiopathology , Animals , Blotting, Western , Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Gastric Acid/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastrins/antagonists & inhibitors , Male , Neuropeptides/pharmacology , Orexin Receptors , Orexins , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stomach Diseases/metabolism
15.
J Pharmacol Exp Ther ; 326(1): 105-16, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18385449

ABSTRACT

1-Methylnicotinamide (MNA) is one of the major derivatives of nicotinamide, which was recently shown to exhibit antithrombotic and antiinflammatory actions. However, it is not yet known whether MNA affects gastric mucosal defense. The effects of exogenous MNA were studied on gastric secretion and gastric lesions induced in rats by 3.5 h of water immersion and water restraint stress (WRS) or in rats administered 75% ethanol. MNA [6.25-100 mg/kg intragastrically (i.g.)] led to a dose-dependent rise in the plasma MNA level, inhibited gastric acid secretion, and attenuated these gastric lesions induced by WRS or ethanol. The gastroprotective effect of MNA was accompanied by an increase in the gastric mucosal blood flow and plasma calcitonin gene-related peptide (CGRP) levels, the preservation of prostacyclin (PGI(2)) generation (measured as 6-keto-PGF1alpha), and an overexpression of mRNAs for cyclooxygenase (COX)-2 and CGRP in the gastric mucosa. R-3-(4-Fluoro-phenyl)-2-[5-(4-fluoro-phenyl)-benzofuran-2-ylmethoxycarbonylamino]-propionic acid (RO 324479), which is the selective antagonist of IP/PGI(2) receptors, reversed the effects of MNA on gastric lesions and GBF. MNA-induced gastroprotection was attenuated by suppression of COX-1 [5-(4-chlorophenyl)-1-(4-methoxyphenyl)-3-(trifluoromethyl)-1H-pyrazole; SC-560] and COX-2 [4-(4-methylsulfonylphenyl)-3-phenyl-5H-furan-2-one; rofecoxib] activity, capsaicin denervation, and by the pretreatment with CGRP(8-37) or capsazepine. Addition of exogenous PGI(2) or CGRP restored the MNA-induced gastroprotection in rats treated with COX-1 and COX-2 inhibitors or in those with capsaicin denervation. WRS enhanced MDA content while decreasing superoxide dismutase (SOD) activity in the gastric mucosa, but pretreatment with MNA reversed these changes. MNA exerts potent gastroprotection against WRS damage via mechanisms involving cooperative action of PGI(2) and CGRP in preservation of microvascular flow, antioxidizing enzyme SOD activity, and reduction in lipid peroxidation.


Subject(s)
Epoprostenol/physiology , Neurons, Afferent/drug effects , Niacinamide/analogs & derivatives , Stomach Ulcer/drug therapy , Stress, Physiological/drug therapy , Acute Disease , Animals , Gastric Acid/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastric Mucosa/physiology , Male , Neurons, Afferent/metabolism , Neurons, Afferent/physiology , Niacinamide/pharmacology , Niacinamide/therapeutic use , Rats , Rats, Wistar , Stomach Ulcer/etiology , Stress, Physiological/complications
16.
J Pineal Res ; 45(2): 180-90, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18298459

ABSTRACT

Following induction of gastric ulcer in rats by serosal application of acetic acid, local mucosal necrosis ensues accompanied by a reduction in mucosal microcirculation and by almost immediate expression of inducible nitric oxide (NO) synthase (iNOS) and proinflammatory cytokines. Daily application of melatonin (20 mg/kg) or l-tryptophan (100 mg/kg) accelerates ulcer healing by affecting the cyclooxygenase-2 (COX-2)-prostaglandin (PG) system with excessive production of protective PG, especially in later period of ulcer healing. Furthermore, expression of hypoxia inducible factor, vascular-endothelial growth factor, an activation of cNOS-NO system and the stimulation of sensory nerves with the expression and release of calcitonin gene related peptide (CGRP) appear to aid the restoration of mucosal repair and microcirculation in the ulcer bed. The enhanced expression of the melatonin MT(2) receptors (MT(2)-R) combined with overexpression of key enzymes involved in biosynthesis of melatonin such as N-acetyltransferase and hydroxyindole-O-methyltransferase contribute to the acceleration of ulcer healing by this indole. Melatonin-induced acceleration of ulcer healing is also mediated by release of gastrin and ghrelin, the most potent stimulants of gastric mucosal cell proliferation and mucosal repair. These sequential steps in ulcer healing accelerated by melatonin can be interfered with by the blockade of MT(2)R, COX-2/PG and cNOS/NO systems, and by reduction in the inflammatory iNOS/NO system. Thus, melatonin and its precursor l-tryptophan, trigger the cascade of molecular events leading to the functional improvement in ulcer healing.


Subject(s)
Melatonin/pharmacology , Stomach Ulcer/drug therapy , Tryptophan/pharmacology , Acetates , Actins/genetics , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacology , Cyclooxygenase 2/genetics , Dinoprostone/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Interleukin-1beta/genetics , Male , Melatonin/administration & dosage , Nitric Oxide Synthase Type III/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Melatonin, MT2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stomach Ulcer/chemically induced , Stomach Ulcer/metabolism , Tryptophan/administration & dosage , Tumor Necrosis Factor-alpha/genetics , Wound Healing/drug effects
17.
J Pineal Res ; 44(4): 408-15, 2008 May.
Article in English | MEDLINE | ID: mdl-18086146

ABSTRACT

The formation of acute gastric lesions depends upon the balance between the aggressive factors promoting mucosal damage and the natural defense mechanisms. Previous studies have shown that melatonin inhibits gastric acid secretion, enhances the release of gastrin, augments gastric blood flow (GBF), increases the cyclooxygenase-2 (COX-2)-prostaglandin (PG) system and scavenges free radicals, resulting in the prevention of stress-induced gastric lesions. Besides the pineal gland, melatonin is also generated in large amounts in the gastrointestinal tract and due to its antioxidant and anti-inflammatory properties; this indole might serve as local protective endogen preventing the development of acute gastric damage. The results of the present study indicate that stress-induced gastric lesions show circadian variations with an increase in the day time and a decline at night. These changes are inversely related to plasma melatonin levels. Following pinealectomy, stress-induced gastric mucosal lesions were more pronounced both during the day and at night, and were accompanied by markedly reduced plasma melatonin levels with a pronounced reduction in mucosal generation of prostaglandin E(2) (PGE(2)), GBF and increased free radical formation and by small rise in plasma melatonin during the dark phase. We conclude that stress-induced gastric ulcerations exhibit a circadian variation with an increase in the day and attenuation at night and that these fluctuations of gastric stress ulcerogenesis occur also after pinealectomy, depending upon the interaction of COX-PG and free radicals, probably mediated by the changes in local gastric melatonin.


Subject(s)
Circadian Rhythm , Melatonin/metabolism , Pineal Gland/surgery , Stomach Ulcer/blood , Stress, Physiological/blood , Animals , Cyclooxygenase 2/metabolism , Dinoprostone/blood , Free Radicals/metabolism , Gastrins/biosynthesis , Male , Rats , Rats, Sprague-Dawley , Regional Blood Flow , Stomach Ulcer/etiology , Stomach Ulcer/pathology , Stress, Physiological/complications , Stress, Physiological/pathology
18.
Przegl Lek ; 64(3): 124-9, 2007.
Article in Polish | MEDLINE | ID: mdl-17941462

ABSTRACT

Present-day methods of successful treatment of inflammatory bowel diseases (IBD) result from a better understanding of their pathophysiology due to advances in preclinical studies in this area of knowledge. Until recently microbiological studies have been focused on the bacterial aspects in pathogenesis of GI disorders, however in the last years an interest in the presence of fungi in the gastrointestinal tract has also increased. In this study using an animal model of ulcerative colitis, the impact of fungal colonization of the colon on the intensity of inflammatory changes in the colonic mucosa and the course of their healing was carried out. The macroscopic and microscopic criteria relating to the changes of weight of examined fragments of the colon were evaluated while assessing differences between groups tested. The intensity of intestinal inflammatory changes was determined by assessment of such parameters, as colonic blood flow (CBF), the level of MPO as a marker of colonic neutrophil infiltration intensity and the plasma levels of IL-1beta; and TNF-alpha concentrations. Results at the 3rd day after TNBS rectal administration revealed an increase of weight of isolated segments of inflammed colon, a decrease of CBF and the 4-5 fold increase of plasma MPO activity. Candida colonization of colon mucosa of rats delayed healing of colonic ulcers, induced by TNBS and this was associated with the increased expression of plasma IL-1beta and TNF-alpha levels. Administration of antifungal (fluconazole) or probiotic (Lacidofil) treatment to C. albicans infected rats exerted favorable effect on healing of inflammatory changes in the colon because the area of ulcerations in groups of rats treated with fluconazole or Lacidofil was significantly smaller in comparison with those inoculated with Candida solution only. Administration of fluconazole or Lacidofil significantly decreased the weight of colon segments, the MPO activity and the plasma IL-1beta and TNF-alpha levels, as compared with respective values in the group receiving Candida only. The results of our studies indicate the deteriorating influence of Candida on the healing process of inflamed colon in the animal model of ulcerative colitis. Concomitant therapy with probiotic or antifungal treatment improved healing of colonic lesions, decreased the weight of inflamed colonic tissue and also attenuated the MPO activity and plasma proinflammatory cytokines IL-1beta and TNF-alpha levels.


Subject(s)
Candidiasis/complications , Candidiasis/physiopathology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/physiopathology , Wound Healing/drug effects , Administration, Rectal , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antifungal Agents/therapeutic use , Candidiasis/drug therapy , Disease Models, Animal , Fluconazole/therapeutic use , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/drug therapy , Interleukin-1beta/blood , Male , Probiotics/therapeutic use , Rats , Rats, Wistar , Trinitrobenzenesulfonic Acid , Tumor Necrosis Factor-alpha/blood
19.
Regul Pept ; 143(1-3): 56-63, 2007 Oct 04.
Article in English | MEDLINE | ID: mdl-17462749

ABSTRACT

Ghrelin, a 28-amino-acid peptide produced predominantly by oxyntic mucosa has been reported to affect the pancreatic exocrine function but the mechanism of its secretory action is not clear. The effects of intraduodenal (i.d.) infusion of ghrelin on pancreatic amylase outputs under basal conditions and following the stimulation of pancreatic secretion with diversion of pancreato-biliary juice (DPBJ) as well as the role of vagal nerve, sensory fibers and CCK in this process were determined. Ghrelin given into the duodenum of healthy rats at doses of 1.0 or 10.0 microg/kg increased pancreatic amylase outputs under basal conditions or following the stimulation of pancreatic secretion with DPBJ. Bilateral vagotomy as well as capsaicin deactivation of sensory fibers completely abolished all stimulatory effects of luminal ghrelin on pancreatic exocrine function. Pretreatment with lorglumide, a CCK(1) receptor blocker, reversed the stimulation of amylase release produced by intraduodenal application of ghrelin. Intraduodenal ghrelin at doses of 1.0 or 10.0 microg/kg increased plasma concentrations of CCK and ghrelin. In conclusion, ghrelin given into the duodenum stimulates pancreatic enzyme secretion. Activation of vagal reflexes and CCK release as well as central mechanisms could be implicated in the stimulatory effect of luminal ghrelin on the pancreatic exocrine functions.


Subject(s)
Amylases/metabolism , Pancreas/drug effects , Peptide Hormones/pharmacology , Animals , Capsaicin/administration & dosage , Capsaicin/pharmacology , Cholecystokinin/blood , Duodenum/drug effects , Duodenum/metabolism , Ghrelin , Humans , Male , Pancreas/enzymology , Pancreas/metabolism , Peptide Hormones/administration & dosage , Proglumide/administration & dosage , Proglumide/analogs & derivatives , Proglumide/pharmacology , Radioimmunoassay , Rats , Rats, Wistar , Receptor, Cholecystokinin A/antagonists & inhibitors , Vagotomy
20.
J Clin Gastroenterol ; 41(2): 145-51, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17245212

ABSTRACT

BACKGROUND AND AIMS: Helicobacter pylori (H. pylori) is an important pathogen in gastritis, peptic ulcer and possibly gastric cancer, but several questions remain unanswered. Particularly how the organism is transmitted and what is the relationship between oral presence of H. pylori and the gastric infection. Accordingly, we aimed to characterize the H. pylori in oral cavity and to evaluate its relationship to gastric H. pylori infection. MATERIALS AND METHODS: Out of total 100 screened for H. pylori infection female subjects (40 to 85 y), 49 patients (pts), who had positive C-urea breath test (UBT) and dyspeptic symptoms, agreed for 1 week regimen of triple anti-H. pylori therapy. The presence of H. pylori in oral cavity was assessed using bacterial culture from saliva and gingival pockets. Gastric H. pylori infection was estimated using capsulated C-urea breath test and plasma anti-H. pylori IgG and saliva IgA antibodies. In addition, plasma gastrin, ghrelin, and pepsinogen I were measured by radioimmunoassay. In selected patients, gastroscopy was additionally performed and gastric biopsy samples were taken for H. pylori random amplification of polymorphic DNA genetic profiling. RESULTS: The triple therapy resulted in gastric H. pylori eradication in 79% pts, along with significant decrease of plasma gastrin combined with an increase in plasma ghrelin and pepsinogen I (PgI) levels and a marked alleviation of dyspeptic symptoms. In contrast to gastric effects, the eradication therapy failed to cause any changes in the presence of H. pylori in oral cavity. Moreover no relationship was observed between the presence of H. pylori in oral cavity and the gastric H. pylori eradication. In line with these findings, no relationship between gastric and oral H. pylori was found using genetic profiling by random amplification of polymorphic DNA. CONCLUSIONS: H. pylori was detected both in the oral cavity and the stomach but oral H. pylori had no relation to gastric H. pylori and remained unaffected by eradication of gastric H. pylori.


Subject(s)
Helicobacter Infections/therapy , Helicobacter pylori/isolation & purification , Mouth/microbiology , Stomach/microbiology , Adult , Aged , Aged, 80 and over , DNA, Bacterial/analysis , DNA, Bacterial/genetics , Female , Gastrins/blood , Ghrelin , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Humans , Middle Aged , Pepsinogen A/blood , Peptide Hormones/blood , Random Amplified Polymorphic DNA Technique , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...