Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
J Physiol Pharmacol ; 74(2)2023 Apr.
Article in English | MEDLINE | ID: mdl-37453089

ABSTRACT

Melatonin (MEL) is produced and secreted by the pineal gland as well as the small intestine, liver, retina, lymphocytes, and melanocytes in the skin in both experimental animals as well as in humans. While pineal and retinas MEL is closely related to the light/dark cycle, the production of MEL by other so called extrapineal tissues is independent of such circadian rhythm. Among the primary mechanisms of action of MEL in humans, the most important are interaction of MEL with specific receptors (M1, M2, M3) and the MEL 'scavenging' activity against the formation of free oxygen metabolites as a result of MEL's ability to transfer free electrons and stimulation of the expression of redox reaction enzymes. In addition, MEL binds to intracellular proteins such as calmodulin, thereby affecting the course of cell cycle, and it has been shown to activate of nuclear receptors belonging to the retinoid orphan receptors/retinoid Z receptors (ROR/RZR) subfamily. MEL exerts regulatory effects on the master clock regulating diurnal rhythms. This updated review presents current view on the synthesis and metabolism of MEL and the growing body of experimental evidence transferable to the practical medicine supporting a pleiotropic molecule beneficial effects on the health including protection against various organ abnormalities, including internal organs such as the liver. Although the beneficial effects of MEL in various types of liver damage have been well documented in experimental studies, there are relatively few studies on liver dysfunction in humans. Considering the worldwide obesity pandemic often associated with the occurrence of steatohepatitis and cirrhosis, the beneficial effects of MEL in liver pathology should be proven in randomized trials involving patients presenting with hepatic disorders.


Subject(s)
Digestive System Diseases , Melatonin , Pineal Gland , Animals , Humans , Melatonin/therapeutic use , Melatonin/pharmacology , Pineal Gland/physiology , Circadian Rhythm/physiology , Digestive System Diseases/drug therapy , Retinoids , Receptors, Melatonin
2.
J Physiol Pharmacol ; 67(5): 717-729, 2016 Oct.
Article in English | MEDLINE | ID: mdl-28011952

ABSTRACT

Carbon monoxide (CO) is a physiological gaseous mediator recently implicated in the mechanism of gastric mucosal defense due to its vasodilatory and antioxidative properties. Small quantities of endogenous CO are produced during heme degradation by heme oxygenase (HO-1), however, the involvement of the capsaicin-sensitive afferent neurons releasing calcitonin gene related peptide (CGRP) and anti-oxidative factors and mechanisms in the CO-induced gastroprotection against stress ulcerogenesis has been little studied. We investigated the possible role of CO released from the CO donor, tricarbonyldichlororuthenium (II) dimer (CORM-2) in the protection against water immersion and restraint stress (WRS)-induced lesions in rats with intact sensory nerves and those with capsaicin denervation and the accompanying changes in malondialdehyde (MDA) content considered as an index of lipid peroxidation, the activity of GSH and SOD-2 and gastric mucosal expression of antioxidative enzymes glutathione peroxidase (GPx) and SOD-2. Wistar rats with intact sensory nerves or those with capsaicin administered in total dose of 125 mg/kg s.c. within 3 days (capsaicin denervation) were pretreated either with 1) vehicle (saline) or 2) CORM-2 (0.1 - 0 mg/kg i.g.) with or without exogenous CGRP (10 µg/kg i.p.) and 30 min later exposed to 3.5 h of WRS. At the termination of WRS, the number of gastric lesions was counted and gastric blood flow (GBF) was assessed by H2-gas clearance technique. The mucosal content of MDA and reduced glutathione (GSH) and the activity of SOD-2 were determined and the expression of GPx-1 and SOD-2 mRNA in the gastric mucosa was analyzed by real-time PCR. The exposure of rats to 3.5 h of WRS resulted in numerous hemorrhagic gastric lesions and significantly decreased the GBF, raised MDA content and significantly decreased the mucosal SOD and GSH contents compared with intact gastric mucosa and these changes were exacerbated in rats with capsaicin denervation. Pretreatment with CORM-2 (1 mg/kg i.g.) which in our previous studies significantly reduced the ethanol and aspirin-induced gastric damage, significantly decreased the number of WRS-induced gastric lesions while raising the GBF and significantly increasing the activity of SOD and GSH (P < 0.05). The pretreatment with CORM-2 significantly decreased MDA content as compared with vehicle-pretreated rats exposed to WRS (P < 0.05). The reduction of WRS damage and the accompanying increase in the GBF as well as the significant decrease in MDA content and the increase in GSH content and SOD activity induced by CORM-2 (1 µg/kg i.g.) were all significantly altered in rats with capsaicin denervation (P < 0.05). The concurrent treatment of CORM-2 with exogenous CGRP in rats with or without sensory nerves tended to decrease the number of WRS lesions as compared with CORM-2 alone pretreated animals and significantly increased the GBF over the values measured in gastric mucosa of CORM-2 alone pretreated rats with or without capsaicin denervation. Such combined administration of CORM-2 and CGRP in rats with capsaicin denervation significantly inhibited an increase in MDA and 4-HNE content and evoked a significant increase in the GSH concentration (P < 0.05) remaining without significant effect on the increase in SOD activity observed with CORM-2 alone. The gastric mucosal expression of SOD-2- and GPx-1 mRNA was significantly increased as compared with those in intact gastric mucosa (P < 0.05). The pretreatment with CORM-2 applied with or without CGRP failed to significantly alter the mRNA expression for SOD-2 and GPx in the gastric mucosa of rats exposed to WRS. Both, the expression of SOD-2- and GPx-1 mRNA was significantly increased in capsaicin denervated rats exposed to WRS rats (P < 0.05) and this effect was abolished by the pretreatment with CORM-2. The expression of SOD-2 tended to decrease, though insignificantly, in rats pretreated with the combination of CORM-2 and CGRP as compared with that detected in CORM-2 alone in rats with capsaicin denervation. In contrast, the mRNA expression of GPx-1 was significantly decreased in gastric mucosa of capsaicin-denervated rats treated with the combination of CORM-2 and CGRP as compared with CORM-2 alone pretreated animals. We conclude that 1) CORM-2 releasing CO exerts gastroprotective activity against stress ulcerogenesis and this effect depends upon an increase in the gastric microcirculation and the vasodilatory activity of this gaseous mediator, and 2) the sensory nerve endings releasing CGRP can contribute, at least in part, to the CO-induced gastric hyperemia, the attenuation of gastric mucosal lipid peroxidation and prevention of oxidative stress as indicated by the CORM-2-induced normalization of the antioxidative enzyme expression enhanced in gastric mucosa of capsaicin-denervated rats.


Subject(s)
Carbon Monoxide/physiology , Gastric Mucosa/metabolism , Glutathione/metabolism , Peptic Ulcer/metabolism , Sensory Receptor Cells/physiology , Superoxide Dismutase/metabolism , Animals , Capsaicin , Denervation , Gastric Mucosa/innervation , Gastric Mucosa/pathology , Glutathione Peroxidase/genetics , Lipid Peroxidation , Male , Malondialdehyde/metabolism , Organometallic Compounds/pharmacology , Peptic Ulcer/pathology , Protective Agents/pharmacology , RNA, Messenger/metabolism , Rats, Wistar , Restraint, Physical , Stress, Psychological/metabolism , Superoxide Dismutase/genetics , Glutathione Peroxidase GPX1
3.
J Physiol Pharmacol ; 67(1): 75-91, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27010897

ABSTRACT

The inhibition of angiotensin-converting enzyme (ACE) or the blockade of angiotensin (Ang) AT-1 receptors affords protection against acute gastric mucosal injury, but whether the major metabolite of renin-angiotensin system (RAS), Ang-(1-7), accelerates the healing process of preexisting gastric ulcers remains unknown. Previous studies documented that Ang-(1-7) acting via its own Mas receptor exerts vascular responses opposing those of Ang II. We studied the effects of the Ang-(1-7)/Mas receptor axis on the healing rate of acetic-acid-induced gastric ulcers with or without the blockade of Mas receptors by A 779 and compared it with the effects of activation and blockade of the AT-1 receptor by the treatment with Ang II and losartan, respectively, the inhibition of ACE by lisinopril, the NO/cNOS inhibition by L-NAME and inhibition of prostaglandin/COX system by indomethacin in the presence of Ang-(1-7). Additionally, ex vivo metabolism of Ang I in gastric tissue was assessed by LC/MS method. At day 9 after ulcer induction, the area of these ulcers and the accompanying changes in total gastric blood flow (GBF) were determined as were gastric mucosal blood flow (GMBF) at ulcer margin and gastric oxygen uptake (GVO2). The gastric mucosal expression of mRNAs for constitutive nitric oxide synthase (cNOS), superoxide dismutase (SOD), and pro-inflammatory cytokines interleukin 1ß (IL-1ß) and tumor necrosis factor alpha (TNF-α) and plasma level of both cytokines were determined by RT-PCR and ELISA. The 9 days treatment with Ang II dose-dependently increased the area of gastric ulcers and this effect was accompanied by a significant fall in the GBF, GVO2 and GMBF at ulcer margin. In contrast, treatment with Ang-(1-7) which produced a significant rise in the luminal content of NO significantly reduced the area of gastric ulcer and significantly increased the GBF, GVO2 and the GMBF at ulcer margin. Similar GMBF changes and significant reduction the area of gastric ulcer was observed in rats with gastric ulcers treated with the agonist of Mas receptor, AVE 0991. These effects of Ang-(1-7) and AVE 0991 were eliminated by blockade of the Mas receptor with A779. Similarly to Ang-(1-7), treatment with losartan or lisinopril significantly reduced the area of gastric ulcers and the accompanying increase in the GMBF at ulcer margin and these effects were significantly attenuated by a concomitant administration of L-NAME and indomethacin. The rate of healing of ulcers was associated with a decrease in ex vivo Ang-(1-7) formation and this effect was attenuated by lisinopril. The treatment with Ang-(1- 7) or AVE 0991 increased the expression of mRNA for cNOS and SOD and downregulated that of IL-1ß and TNF-α followed by the decrease in the plasma IL-1ß and TNF-α levels. We conclude that the Ang-(1-7)/Mas receptor system accelerates the healing of preexisting gastric ulcers via an increase in the gastric macro- and microcirculations, and an increase in gastric tissue oxygenation. These effects are mediated by PG and NO derived from overexpression of cNOS, an increase in the expression of antioxidizing enzyme SOD 2 and an anti-inflammatory action involving the inhibition of expression and release of pro-inflammatory cytokines IL-1ß and TNF-α. Our results seem to underlie the importance of the Ang-(1-7), AT-1 and Mas receptors in the regulation of local vascular and metabolic effects associated with mechanism of gastric ulcer healing.


Subject(s)
Angiotensin I/metabolism , Cytokines/metabolism , Nitric Oxide/metabolism , Peptide Fragments/metabolism , Prostaglandins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Renin-Angiotensin System/drug effects , Stomach Ulcer/metabolism , Angiotensin II/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Enzyme Inhibitors/pharmacology , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Imidazoles/pharmacology , Indomethacin/pharmacology , Interleukin-1beta/metabolism , Lisinopril/pharmacology , Losartan/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/metabolism , Proto-Oncogene Mas , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
J Physiol Pharmacol ; 65(5): 613-22, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25371520

ABSTRACT

The gastric mucosa plays an important role in the physiological function of the stomach. This mucosa acts as gastric barrier, which protects deeper located cells against the detrimental action of the gastric secretory components, such as acid and pepsin. Integrity of the gastric mucosa depends upon a variety of factors, such as maintenance of microcirculation, mucus-alkaline secretion and activity of the antioxidizing factors. The pathogenesis of gastric mucosal damage includes reactive oxygen species (ROS), because of their high chemical reactivity, due to the presence of uncoupled electron within their molecules. Therefore they cause tissue damage, mainly due to enhanced lipid peroxidation. Lipid peroxides are metabolized to malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE). The local increase of MDA and 4-HNE concentration indicates ROS-dependent tissue damage. Superoxide dismutase (SOD) is the main enzyme, which neutralizes ROS into less noxious hydrogen peroxide. A decrease of SOD activity is an indicator of impairment of the protective mechanisms and significantly contributes to cell damage. Hydrogen peroxide is further metabolized to water in the presence of reduced glutathione (GSH). GSH can also work synergetically with SOD to neutralize ROS. The reactions between GSH and ROS yields glutathione free radical (GS(•)), which further reacts with GSH leading to free radical of glutathione disulphide (GSSG(•)). This free radical of GSSG can then donate an electron to the oxygen molecule, producing O2 (•-) Subsequently, O2 (•-) is eliminated by SOD. Adecrease of the GSH level has detrimental consequences for antioxidative defense cellular properties. Gastric mucosa, exposed to stress conditions, exhibits an enhancement of lipid peroxidation (increase of MDA and 4-HNE), as well as a decrease of SOD activity and GSH concentration. This chain reaction of ROS formation triggered by stress, appears to be an essential mechanism for understanding the pathogenesis of stress - induced functional disturbances in the gastric mucosa leading to ulcerogenesis.


Subject(s)
Gastric Mucosa/injuries , Gastric Mucosa/metabolism , Oxidative Stress , Animals , Glutathione/metabolism , Humans , Lipid Peroxidation , Reactive Oxygen Species , Superoxide Dismutase/metabolism
5.
J Physiol Pharmacol ; 65(6): 809-22, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25554985

ABSTRACT

Gastroesophageal reflux disease (GERD) is a global disease rapidly increasing among world population. The pathogenesis of reflux esophagitis which is considered as the early stage of GERD is complex, resulting from an imbalance between aggressive factors damaging the esophagus and a number of the natural defense mechanisms. The esophageal mucosa is in a state of continuous exposure to potentially damaging endogenous and exogenous factors. Important aggressive components of gastric refluxate include acid and pepsin and also pancreatic enzymes and bile. Among aggressive factors of exogenous origin, cigarette smoking, non-steroidal anti-inflammatory drugs (NSAID), and steroids are of the utmost importance. The basic level of esophageal defense against acid-pepsin damage consists of the anti-reflux mechanisms such as the luminal acid clearance and removal of the esophageal contents and neutralization of luminal acidity. In addition the esophageal mucosal protection includes the presence of pre-epithelial, epithelial and post-epithelial cellular and functional components. Recently, the progress have been made in the understanding of role of the heptapeptide member of the renin-angiotensin system (RAS), angiotensin-(1-7) (Ang-(1-7)) in the control of gastrointestinal functions. It has been shown that all components of local RAS including Ang-(1-7) are detectable in the gastrointestinal wall including not only the stomach but also the esophagus. Previous studies revealed that Ang-(1-7), which is an important component of the RAS, exerts vasodilatory, anti-inflammatory and antioxidant activities in the stomach. Ang-(1-7) was recently implicated in gastroprotection, but its effects on esophageal mucosa in a rodent model of reflux esophagitis and in human subjects presenting GERD symptoms have not been explored. The present study was aimed to evaluate the possible protective effects of Ang-(1-7) and Mas-receptors upon esophageal mucosal damage in acute reflux esophagitis (RE) induced in anesthetized rats by ligating the pylorus and the limiting ridge (a transitional region between the forestomach and the corpus of stomach). Consequently, the total gastric reservoir to store gastric juice was greatly diminished, resulting in the reflux of this juice into the esophagus. Because Mas receptors are functionally linked to nitric oxide (NO) formation, we also studied involvement of endogenous NO in the mediation of protective and circulatory effects of exogenous Ang-(1-7). Moreover, an attempt was made to assess the possible role of sensory neurons in the modulation of the protective effects exerted by Ang-(1-7)/Mas receptor system. Six series of rats were pretreated 30 min before induction of RE with 1) vehicle (saline), 2) Ang-(1-7) (5-50 µg/kg i.p.), 3) A779 (50 µg/kg i.p.), the selective Mas receptor antagonist applied alone, 4) Ang-(1-7) (50 µg/kg i.p.) combined with A779, 5) L-NNA (20 mg/kg i.p.) administered alone, and 6) Ang-(1-7) (50 µg/kg i.p.) combined with L-NNA. In separate group of rats, capsaicin (total dosage of 125 mg/kg within three days) was administered s.c. 2 weeks before the induction of RE to induce functional ablation of sensory nerves. Rats with intact sensory nerves and those with capsaicin-induced sensory denervation received vehicle (saline) or Ang-(1-7) (50 µg/kg i.p.) to determine whether this vasoactive metabolite of angiotensin I could be also effective in rats with capsaicin-induced impairment of the synthesis and release of sensory neuropeptides such as CGRP. Four hours after induction of RE, the mucosal damage was graded with mucosal lesion index (LI) from 0 to 6, the esophageal microcirculatory blood flow (EBF) was determined by H2-gas clearance technique and plasma level of pro-inflammatory cytokines interleukin-1b (IL-1ß), and tumor necrosis factor-α (TNF-α) was determined by ELISA. The expression of proinflammatory factors including COX-2, cytokine IL-1ß and hypoxia inducible factor 1alpha (Hif1α) was analyzed in the esophageal mucosal biopsies. In rats with RE, the esophageal LI was significantly elevated comparing its value observed in intact rats, and the EBF was significantly decreased as compared with intact mucosa. Pretreatment with Ang-(1-7) of control rats without esophagitis induced increase in EBF by about 25% without any macroscopic changes in the esophageal mucosa or in the plasma level of cytokines. In animals with RE, pretreatment with Ang-(1-7) significantly reduced gross and histological esophageal mucosal injury and significantly increased EBF in comparison to vehicle-pretreated animals. The observed gross and histologic esophagoprotective effect of Ang-(1-7) was totally abolished by A779 so in rats with combined treatment of A779 with Ang-(1-7), the LI was identical with this observed in control RE and the EBF was decreased in these animals by about 39%. Inhibition of NO synthase by L-NNA significantly reduced the LI and the rise in EBF caused by Ang-(1-7). Similarly, the capsaicin denervation also significantly attenuated the vasodilatory and the esophagoprotective effects of Ang-(1-7). The expression of proinflammatory factors COX-2, Hif1α and IL-1ß which was negligible in intact esophageal mucosa, was upregulated in esophageal mucosa of rats with RE. In contrast, the administration of Ang-(1-7) resulted in a downregulation of mRNA for COX-2, Hif1 and IL-1ß in esophageal mucosa an this effect was abolished in A779-dependent manner. The Ang-(1-7) significantly decreased the RE-induced elevation of plasma levels of IL-1ß and TNF-α, and this effect was also reversed by pretreatment with A779, and significantly attenuated by pretreatment with L-NNA and capsaicin-induced sensory denervation. The present study indicates that the protective effect of Ang-(1-7) observed in the esophageal mucosa during early acute stage of gastroesophageal reflux depends upon the enhancement of esophageal microcirculatory blood flow via the activation of Mas receptor possibly due to NO synthase/NO system activation, stimulation of sensory nerves, the inhibition of expression of pro-inflammatory factors including COX-2, Hif1α and IL-1ß and release of proinflammatory cytokines IL-1ß and TNF-α.


Subject(s)
Angiotensin I/therapeutic use , Esophagitis, Peptic/drug therapy , Peptide Fragments/therapeutic use , Protective Agents/therapeutic use , Angiotensin I/pharmacology , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Animals , Cyclooxygenase 2/genetics , Disease Models, Animal , Esophagitis, Peptic/metabolism , Esophagitis, Peptic/pathology , Esophagitis, Peptic/physiopathology , Esophagus/blood supply , Esophagus/metabolism , Esophagus/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Interleukin-1beta/blood , Interleukin-1beta/genetics , Male , Mucous Membrane/drug effects , Mucous Membrane/metabolism , Nitric Oxide/metabolism , Peptide Fragments/pharmacology , Protective Agents/pharmacology , Proto-Oncogene Mas , Proto-Oncogene Proteins/antagonists & inhibitors , RNA, Messenger/metabolism , Rats, Wistar , Receptors, G-Protein-Coupled/antagonists & inhibitors , Regional Blood Flow/drug effects , Sensory Receptor Cells/metabolism , Tumor Necrosis Factor-alpha/blood
6.
J Physiol Pharmacol ; 64(5): 625-37, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24304576

ABSTRACT

Hormonal peptides like ghrelin, orexin A (OXA) or nesfatin-1 not only regulate appetite, which is their basic biological function, but also contribute to mechanisms responsible for maintaining integrity of the gastric mucosa. Previous studies including those from our laboratory have revealed that their gastroprotective effect results from cooperation with other factors responsible for protection of the gastric mucosa, including prostaglandin (PG) synthesis pathway, nitric oxide (NO) and the sensory afferent fibres releasing the vasoactive neurotransmitters. The aim of the present study was to determine whether ghrelin, orexin-A (OX-A) or nesfatin-1 with their protective effect on the gastric mucosa, also can modify the healing of chronic gastric ulcers. Furthermore, an attempt was made to explain participation of these peptides in healing processes of chronic gastric ulcers with comorbid conditions for the human beings resulted from diabetes mellitus. In our study, a model of gastric ulcers caused by concentrated acetic acid to induce the chronic gastric ulcers was used, while the clinical condition corresponding to diabetes was induced by single injection of streptozotocin (STZ). We found that ghrelin, OX-A and nesfatin-1 accelerate dynamics of the acetic acid ulcers healing, confirmed by a reduction in the ulcer area and this effect was accompanied by an increase in gastric blood flow at the ulcer margin. Destruction of sensory afferent fibres with capsaicin or blocking of vanilloid receptors with capsazepine resulted in a significant reduction of ghrelin, OX-A and nesfatin-1-induced acceleration of ulcer healing. Similar results were obtained when an NO-synthase blocker, L-NNA was used in a combination with these peptides. Moreover, it was found that OX-A and nesfatin-1 failed to accelerate the healing process under diabetic condition because both these hormones induced reduction in the ulcer area and the increase in blood flow in normal, non-diabetic rats were completely lost in the group of animals with diabetes. Treatment with OX-A and nesfatin-1 increased superoxide dismutase (SOD) mRNA expression even in acetic acid ulcers concurrent with diabetes. However, the treatment with OX-A and nesfatin-1 failed to alter the increase in gastric mucosal mRNA expression for ghrelin and hypoxia-inducible factor 1-alpha (HIF-1α), this latter effect that had been strongly pronounced in diabetic animals. We conclude that the hormonal peptides involved in the regulation of satiety and hunger such as ghrelin, OX-A and nesfatin-1 contribute to the process of chronic gastric ulcers healing cooperating with NO and sensory afferent nerve endings releasing vasoactive neuropeptide CGRP. Furthermore, OX-A and nesfatin-1, the two relatively unrecognized peptides, play an essential role in healing process of chronic gastric ulcers activating the gastric blood flow at ulcer margin and the mucosal regeneration and both ulcer healing and accompanying hyperemia at ulcer margin are greatly impaired during diabetes. Possibly, loss of the healing effect of these peptides during diabetes results from an interaction with radical generation processes as reflected by an increase of mRNA expression for SOD as well as the failure of their attenuating activity on proinflammatory factors such as HIF-1α.


Subject(s)
Anti-Ulcer Agents/therapeutic use , Calcium-Binding Proteins/therapeutic use , DNA-Binding Proteins/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Ghrelin/therapeutic use , Intracellular Signaling Peptides and Proteins/therapeutic use , Nerve Tissue Proteins/therapeutic use , Neuropeptides/therapeutic use , Stomach Ulcer/drug therapy , Acetic Acid , Animals , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Eating , Gastric Mucosa/blood supply , Gastric Mucosa/pathology , Nucleobindins , Orexins , Rats , Rats, Wistar , Regional Blood Flow/drug effects , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology , Stomach Ulcer/physiopathology
7.
Curr Med Chem ; 19(1): 55-62, 2012.
Article in English | MEDLINE | ID: mdl-22300076

ABSTRACT

The term cytoprotection pioneered by Robert and colleagues has been introduced to describe the remarkable ability of endogenous and exogenous prostaglandins (PGs) to prevent acute gastric hemorrhagic lesions induced by noxious stimuli such as ethanol, bile acids, hiperosmolar solutions and nonsteroidal anti-inflammatory agents such as aspirin. Since that time many factors were implicated to possess gastroprotective properties such as growth factors including epidermal growth factor (EGF) and transforming factor alpha (TGFα), vasodilatory mediators such as nitric oxide (NO) and calcitonin gene related peptide (CGRP) as well as appetite gut hormones including gastrin and cholecystokinin (CCK), leptin and recently ghrelin. This protective action of gut peptides has been attributed to the release of PG but question remains whether another peptide angiotensin, the classic component of the systemic and local renin-angiotensin system (RAS) could be involved in the mechanism of gastric integrity and gastroprotection. After renin stimulation, the circulating angiotensin I is converted to angiotensin II (ANG II) by the activity of the Angiotensin Converting Enzyme (ACE). The ANG II acting via its binding to two major receptor subtypes the ANG type 1 (AT1) and type 2 (AT2) has been shown be activated during stress and to contribute to the pathogenesis of cold stress- and ischemia-reperfusion-induced gastric lesions. All bioactive angiotensin peptides can be generated not only in systemic circulation, but also locally in several tissues and organs. Recently the new functional components of RAS, such as Ang-(1-7), Ang IV, Ang-(1-12) and novel pathways ACE2 have been described suggesting the gastroprotective role for the novel ANG II metabolite, Ang-(1-7). The fact that Ang-(1-7) is produced in excessive amounts in the gastric mucosa of rodents and that pretreatment by Ang-(1-7) exhibits a potent gastroprotective activity against the gastric lesions induced by cold-restraint stress suggests that this and possibly other vasoactive metabolites of ANG II pathway could be involved in the mechanism of gastric integrity and gastroprotection. This review summarizes the novel gastroprotective factors and mechanisms associated with metabolic fate of systemic and local RAS activation with major focus to recent advancement in the angiotensin pathways in the gut integrity.


Subject(s)
Gastric Mucosa/physiology , Renin-Angiotensin System/physiology , Angiotensins/physiology , Animals , Humans
8.
J Physiol Pharmacol ; 62(4): 429-39, 2011 Aug.
Article in English | MEDLINE | ID: mdl-22100844

ABSTRACT

Ghrelin is an important hormone involved in the control of the human appetite center. Recently, protective properties of this hormone have been recognized in various models of impairment of the gastric mucosa, including stress, ischemia and reperfusion (I/R). Ghrelin is predominantly secreted by the gastric mucosa of stomach, but there are other sources of ghrelin, for example in the hypothalamus and various parts of the central nervous system (CNS) that should be taken into consideration. This hormone exerts biological effects via the activation of growth hormone secretagogue receptor (GHSR), the presence of which was confirmed in different parts of the gastrointestinal (GI) tract and midbrain structures. Although substantial evidence of the divergent biological effects of ghrelin and the mechanism of its action has been emphasized, the precise mechanisms of ghrelin which affords GI protection is still unclear. Particularly, there is a sparse amount of evidence concerning its action on the GI system. The major aim of the present study was to evaluate the importance of peripherally and centrally administered ghrelin at different times of the ischemia and reperfusion (I/R period in the modulation of resistance of the intestinal mucosa to the injury induced by ischemia and subsequent reperfusion. Secondly, we wanted to evaluate the possible mechanism of the action of ghrelin with a particular focus on its influence on the intestinal blood flow. Male Wistar rats were divided into 4 series (A-D) of the experimental groups (n=7). In series A the importance of peripherally administered ghrelin at different time of I/R period was studied. In series B the importance of centrally administered ghrelin at different time of I/R period was evaluated. In series C and D, the mechanisms of peripherally and centrally administered hormone were examined, respectively. Two models of the I/R period were selected: short lasting (30/60 min) and long lasting (60/120 min). The following drugs were used: ghrelin (50 µg/kg i.p. or 1 nmol in 10 µl i.c.v.), 6 hydroxy dopamine (50 mg/kg i.p.), nadolol (0.5 mg/kg i.p.), calcitonin gene related peptide fragment (CGRP(8-37), 100 µg /kg i.p.), capsaicin (5-10 mg/100 ml solution s.c.). The mesenteric blood flow (MBF-ml/min), the intestinal microcirculatory blood flow (LDBF-PU), the arterio-venous oxygen difference (AVO(2)-ml/O(2)/100 ml blood), and the intestinal oxygen uptake (VO(2)) in ml O(2)/min were measured. Mucosal impairment was assessed planimetrically with the use of a digital photo analyzer (LA) and histologically with the use of the six-point Park/Chiu scale. Peripheral administration of ghrelin evoked marked increase of MBF and LDBF by 42% and 48%, respectively, with significant reduction of LA by 38%. When ghrelin was administered at the beginning of the reperfusion period during the short I/R period or prior to the long lasting I/R period, the vascular reactions and protective effects were reduced, but not completely abolished. The central administration of ghrelin before the short I/R period significantly increased the MBF and LDBF by about 32% and 35%, respectively, as well as LA reduction by about 20% in comparison to the control group. However, when ghrelin was administered prior to the long I/R period or after the onset of completed ischemia, neither vascular nor protective effects were noticed. Sensory denervation and the blockade of the CGRP1 receptors totally blocked the protective and hyperemic effects of the peripherally administered ghrelin. Selective blockade of the adrenergic system or blunting of the vagal nerves (vagotomy) significantly but not totally eliminated the effects of centrally applied ghrelin, which were abolished when both adrenergic and parasympathetic pathways were ablated. These results indicate that ghrelin applied centrally or peripherally markedly increases resistance of the intestinal tissue during the I/R period induced mucosal and hyperemic impairment evoked by I/R. Ghrelin is an important mediator of the increase in the intestinal microcirculation and elevation of the intestinal metabolism, which seems to be, at least in part, responsible for the observed protection of the intestine subjected to I/R. Impairment of this microvasculature response due to I/R seems to be responsible for a markedly observed weaker effect of ghrelin when this hormone was administered after the ischemic period. The lack of a protective effect observed after central administration of this peptide against a long lasting I/R period is probably due to damage of neural pathways caused by I/R. Finally, the peripheral activity of ghrelin in the intestine is mediated by the sensory neurons with a prominent role of CGRP released from their endings. However, this peripheral action of ghrelin depends upon the proper functioning of both the sympathetic and parasympathetic system.


Subject(s)
Ghrelin/administration & dosage , Ghrelin/therapeutic use , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Mesenteric Vascular Occlusion/prevention & control , Reperfusion Injury/prevention & control , Afferent Pathways/drug effects , Animals , Capsaicin/pharmacology , Disease Models, Animal , Drug Administration Schedule , Ghrelin/metabolism , Ghrelin/pharmacology , Injections, Intraperitoneal , Injections, Intraventricular , Intestinal Mucosa/blood supply , Intestinal Mucosa/innervation , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/blood supply , Intestine, Small/innervation , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mesenteric Vascular Occlusion/etiology , Mesenteric Vascular Occlusion/metabolism , Mesenteric Vascular Occlusion/pathology , Microcirculation/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oxygen Consumption/drug effects , Rats , Rats, Wistar , Receptors, Ghrelin/metabolism , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Splanchnic Circulation/drug effects , Sympathectomy, Chemical , Time Factors , Vagotomy
9.
J Physiol Pharmacol ; 62(3): 357-68, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21893697

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used for their anti-inflammatory, analgesic and antipyretic effects, however their use is associated with the broad spectrum of side effects observed in human as well as the experimental animals. Despite damaging activity of NSAIDs in upper gastrointestinal (GI) tract, these drugs exert deleterious influence in lower GI tract, including colon. The role of GI microflora in the pathogenesis of NSAIDs-induced experimental colonic damage is not completely understood. The aim of this study was 1) to evaluate the relative importance of the GI microflora on the experimental colonic damage in the presence of caused by NSAID, and 2) to assess the efficacy of antibiotic treatment with ampicillin on the process of healing of colitis. We compared the effect of vehicle, ASA applied 40 mg/kg intragastrically (i.g.) or the selective cyclooxygenase (COX)-2 inhibitor, celecoxib (25 mg/kg i.g.) without or with ampicillin treatment (800 mg/kg i.g.) administered throughout the period of 10 days, on the intensity of TNBS-induced colitis in rats. The severity of colonic damage, the alterations in the colonic blood flow (CBF) and myeloperoxidase (MPO) activity, the mucosal expression of TNF-α, IL-1ß, COX-2, VEGF and iNOS and the plasma concentration of TNF-α and IL-1ß were assessed. In all rats, the faeces samples as well as those from the colonic mucosa, blood, liver and spleen underwent microbiological evaluation for intestinal bacterial species including Escherichia coli and Enterococcus spp. The administration of TNBS resulted in macroscopic and microscopic lesions accompanied by the significant fall in the CBF, an increase in tissue weight and 4-5-fold rise in the MPO activity and a significant increase in the plasma IL-1ß and TNF-α levels. ASA or celecoxib significantly increased the area of colonic lesions, enhanced MPO activity and caused the marked increase in colonic tissue weight and plasma IL-1ß and TNF-α levels, as well as an overexpression of mRNA for IL-1ß and TNF-α, COX-2, VEGF and iNOS in the colonic tissue. ASA and coxib also resulted also in a significant increase of E. coli counts in the stool at day 3 and day 10 day of the observation compared with the intact rats. Moreover, E. coli translocation from the colon to the blood and extraintestinal organs such as liver and spleen in the group of rats treated without or with ASA and coxib. E. coli was the most common bacteria isolated from these organs. Treatment with ampicillin significantly attenuated the ASA- or celecoxib-induced increase in plasma levels of IL-1ß and TNF-α and suppressed the mucosal mRNA expression for IL-1ß and TNF-ß, COX-2, iNOS and VEGF in the colonic mucosa. Ampicillin administration caused a significant fall in the number of E. coli in the faeces at day 3 and day 10 of observation in ASA- and coxib-treated rats with colitis. Antibiotic therapy markedly reduced bacterial translocation to the colonic tissue and the extraintestinal organs such as the liver and spleen. We conclude that administration of ASA and to lesser extent of celecoxib, delays the healing of experimental colitis and enhances the alterations in colonic blood flow, proinflammatory markers such as IL-1ß, TNF-α, COX-2, iNOS and VEGF and increased intestinal mucosal permeability resulting in the intestinal bacterial translocation to the blood, spleen and liver. Antibiotic treatment with ampicillin is effective in the diminishing of the severity of colonic damage, counteracts both the NSAID-induced fall in colonic microcirculation and bacterial E.coli translocation to the extraintestinal organs.


Subject(s)
Ampicillin/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Colitis/drug therapy , Colitis/pathology , Cyclooxygenase 2 Inhibitors/toxicity , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Aspirin/toxicity , Bacterial Load , Bacterial Translocation , Celecoxib , Chemokines/blood , Colitis/chemically induced , Colon/blood supply , Colon/microbiology , Colon/pathology , Cyclooxygenase 2 Inhibitors/pharmacology , Enterococcus/growth & development , Enterococcus/isolation & purification , Enterococcus/metabolism , Escherichia coli/growth & development , Escherichia coli/isolation & purification , Escherichia coli/metabolism , Feces/microbiology , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestines/microbiology , Male , Microcirculation , Peroxidase/metabolism , Pyrazoles/pharmacology , Pyrazoles/toxicity , Rats , Rats, Wistar , Sulfonamides/pharmacology , Sulfonamides/toxicity , Trinitrobenzenesulfonic Acid/toxicity
10.
J Physiol Pharmacol ; 62(1): 75-86, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21451212

ABSTRACT

The purpose of this study was to develop an acute animal model of reflux esophagitis, which would be suitable to induce the esophageal damage caused by gastric acid reflux, thus mimicking the esophageal injury of human gastroesophageal reflux disease (GERD). Global research indicates that GERD is rapidly increasing among the world's population. NSAIDs are known to induce gastrointestinal damage and low doses of aspirin (ASA) have been shown to increase the incidences of GERD in humans. Gastric acid and pepsin secretion and enhanced COX-2 expression were implicated in the pathogenesis of reflux esophagitis, but the effect of selective COX-2 inhibitors against lesions induced by the reflux of gastric acid content into esophagus has not been thoroughly studied. Here, we compared the effect of aspirin (ASA) and so called "safe" nitric oxide (NO) derivative of ASA with those of non-selective and selective cyclooxygenase (COX)-1 and COX-2 in rat model of reflux esophagitis. Reflux esophagitis was induced in anesthetized rats by ligating the pylorus and limiting ridge transitional region between the forestomach and the corpus of stomach. Subsequently, the total gastric reservoir to store gastric juice was greatly diminished, resulting in the reflux of this juice into the esophagus. Rats with esophagitis received intragastric (i.g.) pretreatment either with: 1) vehicle (saline), 2) ASA or NO-ASA (100 mg/kg); 3) the non-selective COX inhibitor, indomethacin (5 mg/kg); 4) the selective COX-1 inhibitor, SC-560 (10 mg/kg), and 5) the selective COX-2 inhibitor, celecoxib (5 mg/kg). In a separate series of rats with reflux oesophagitis, the efficacy of ASA combined with a donor of NO, glyceryl trinitrate (GTN; 10 mg/kg i.g.) to prevent esophageal mucosal injury was investigated. Four hours after induction of esophagitis the gross mucosal damage was graded with a macroscopic lesion index (LI) from 0-6. The esophageal blood flow (EBF) was determined by H2-gas clearance technique, the oesophageal mucosal and blood samples were collected for histology and analysis of the RT-PCR expression and release of proinflammatory cytokines IL-1ß, TNF-α and IL-6 using specific ELISA. The exposure of the esophagus to reflux of gastric acid time-dependently increased the esophageal LI and morphologic damage, and decreased EBF with the most significant changes observed at 4 hrs after the ligation procedure. The pretreatment with native ASA in the dose that suppressed the generation of mucosal PGE2, enhanced gross and histologic esophageal damage and produced a significant fall in EBF. NO-ASA or ASA coupled with GTN counteracted the aggravation of the damage and accompanying fall in EBF when compared with native ASA applied alone to rats with esophagitis. The proinflammatory cytokines IL-1ß and TNF-α were overexpressed in rats with esophagitis and those pretreated with ASA but this effect was significantly attenuated by NO-ASA. Plasma IL-1ß, TNF-α and IL-6 were negligible in the intact rats but significantly increased in those with esophagitis, with this effect being further enhanced by non-selective (indomethacin) and selective (SC-560, celecoxib) COX-1 and COX-2 inhibitors. We conclude that conventional NSAID such as aspirin augments esophagitis, while NO-ASA exerts the beneficial protective effect against reflux esophagitis via the enhancement of esophageal microcirculation due to NO release and an inhibitory effect on expression and release of pro-inflammatory cytokines.


Subject(s)
Aspirin/analogs & derivatives , Cytokines/biosynthesis , Esophagitis, Peptic/drug therapy , Esophagitis, Peptic/metabolism , Esophagus/drug effects , Esophagus/pathology , Nitric Oxide Donors/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Celecoxib , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Cytokines/blood , Cytokines/genetics , Dinoprostone/metabolism , Esophagitis, Peptic/chemically induced , Esophagitis, Peptic/pathology , Esophagus/blood supply , Esophagus/metabolism , Gastric Acid/metabolism , Humans , Indomethacin/pharmacology , Male , Nitric Oxide/pharmacology , Nitroglycerin/pharmacology , Pyrazoles/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Sulfonamides/pharmacology
11.
J Physiol Pharmacol ; 60(2): 41-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19617644

ABSTRACT

Ghrelin is a novel growth hormone (GH)-releasing and orexigenic peptide with anti-inflammatory activities. However, the role of ghrelin in the colonic inflammation is still controversial. The aim of the present study was: 1) to examine the expression of ghrelin and TNF-alpha mRNA in the inflamed colonic mucosa of patients with ulcerative colitis (UC), 2) to analyze the effect of treatment with exogenous ghrelin on the healing of trinitrobenze sulphonic acid (TNBS)-induced colitis in rats, and 3) to assess the effects of ghrelin treatment on mRNA expression for iNOS and protein expression for COX-2 and PPARalpha in intact colonic mucosa and in that with TNBS-induced colitis. Fifteen patients with UC and fifteen healthy controls were enrolled in this study. Expression of ghrelin and TNF-alpha was assessed by semi-quantitative RT-PCR in the colonic mucosal biopsies from UC patients and healthy controls. In addition, the effect of exogenous ghrelin on healing of TNBS colitis was tested in rats without or with capsaicin-induced functional ablation of sensory nerves. Patients with UC showed a significant upregulation of mRNA for ghrelin and TNF-alpha in colonic mucosa as compared to that observed in healthy controls. The expression of ghrelin correlated with the grade of inflammation and expression of TNF-alpha. In rats the exogenous ghrelin administered daily at a dose of 20 microg/kg i.p. significantly accelerated the healing of TNBS colitis and this effect was accompanied by an increase in mRNA expression for iNOS and protein expression for COX-2 in the colonic mucosa. The protein expression for PPARgamma, which was down-regulated in rat colonic mucosa after exposure to TNBS as compared to that in intact colonic mucosa, was not significantly influenced by ghrelin treatment. We conclude that 1) patients with UC show an increased mucosal expression of mRNA for ghrelin in the colonic mucosa which could trigger protective response in inflamed colon; and 2) exogenous ghrelin accelerates healing of colonic lesions in animal model of ulcerative colitis via increased release of NO and PGE(2) due to an increase in iNOS and COX-2 expression and stimulation of sensory neuropeptides such as CGRP released from sensory afferent endings.


Subject(s)
Colitis, Ulcerative/metabolism , Colon/metabolism , Ghrelin/biosynthesis , Ghrelin/pharmacology , Neurons, Afferent/metabolism , Nitric Oxide/metabolism , Animals , Capsaicin , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Colon/pathology , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Dinoprostone/metabolism , Ghrelin/therapeutic use , Humans , Neurons, Afferent/drug effects , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , PPAR gamma/biosynthesis , PPAR gamma/genetics , RNA, Messenger/biosynthesis , Rats , Trinitrobenzenesulfonic Acid , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Up-Regulation
12.
J Physiol Pharmacol ; 60 Suppl 7: 47-56, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20388945

ABSTRACT

This review summarizes the involvement of centrally and peripherally applied melatonin, a major hormone of pineal gland, in the mechanism of gastric mucosal integrity, gastroprotection and ulcer healing. Melatonin was originally shown to attenuate gastric mucosal lesions but the controversy exists in the literature as to whether melatonin derived from the pineal gland, considered as the major source of this indole or rather that locally generated from L-tryptophan within gastric mucosa, plays predominant role in the mechanism of gastrointestinal integrity. Both, intragastric (i.g.) and intracerebroventricular (i.c.v.) administration of melatonin and its precursor, L-tryptophan to rats without or with removed pineal gland by pinealectomy attenuates in the dose-dependent manner the formation of on gastric lesions induced by topical irritants and water immersion restraint stress (WRS). Melatonin accelerated the gastric ulcer healing and this was accompanied by the rise in gastric blood flow (GBF), the plasma melatonin and gastrin levels, the mucosal generation of PGE(2) and luminal NO content. Pinealectomy, which suppresses the plasma melatonin levels, markedly aggravated the gastric lesions induced by WRS. Concurrent supplementation of pinealectomized animals with melatonin or L-tryptophan, the melatonin precursor, attenuated the lesions induced by WRS. Treatment with luzindole, an antagonist of Mel(2) receptors, or with L-NNA, the NO-synthase inhibitor, significantly attenuated melatonin- and L-tryptophan-induced protection and the acceleration of ulcer healing and the accompanying increase in the GBF and luminal content of NO. We conclude that 1) exogenous melatonin and that released from the L-tryptophan attenuate lesions induced by topical irritant such as ethanol and WRS via interaction with MT(2) receptors and due to an enhancement of gastric microcirculation, probably mediated by NO and PG derived from cNOS, iNOS and COX-2 overexpression and activity, and 2) the pineal gland plays an important role in the limitation of WRS-induced gastric lesions and acceleration of ulcer healing via releasing melatonin predominately at night time, that exerts gastroprotective and ulcer healing actions.


Subject(s)
Gastric Mucosa/physiology , Gastric Mucosa/physiopathology , Gastrointestinal Tract/physiology , Gastrointestinal Tract/physiopathology , Melatonin/physiology , Stomach Diseases/prevention & control , Animals , Gastric Mucosa/blood supply , Gastric Mucosa/metabolism , Gastrointestinal Tract/blood supply , Humans , Lipid Peroxidation , Melatonin/biosynthesis , Melatonin/blood , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Organ Specificity , Pineal Gland/physiology , Prostaglandin-Endoperoxide Synthases/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/physiology , Receptors, Melatonin/antagonists & inhibitors , Receptors, Melatonin/physiology , Stomach Diseases/physiopathology , Tryptophan/metabolism , Wound Healing
13.
J Physiol Pharmacol ; 59 Suppl 2: 89-102, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18812631

ABSTRACT

Prostaglandins mediate various physiological aspects of mucosal defense and the suppression of prostaglandin synthesis in the stomach is a critical event in terms of the development of mucosal injury after NSAID administration. However, it has become clear that other mediators besides prostaglandins can similarly act to protect the stomach from injury. 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. Thus, administration of NO in a form of NO-donors exert protective influence on the stomach from the injury that usually occurs when mucosal prostaglandin levels are suppressed. The new class of NO releasing NSAIDs, including NO-aspirin, represent a very promising approach to reducing the toxicity of anti-inflammatory drugs. Lipoxins are another group of lipid mediators that can protect the stomach. Aspirin-triggered lipoxin synthesis, via COX-2, acts to reduce the severity of damage induced by this drug. Lipoxin analogues may prove to be useful for preventing mucosal injury and for modulating mucosal inflammation. Aspirin-triggered lipoxin also seems to play in important role in gastric adaptation during chronic aspirin administration. Suppression of COX-2 activity by selective COX-2 inhibitors abolishes the production of this endogenous gastroprotective substance and diminishes the gastric tolerability of NSAIDS and gastric adaptation to these drugs. This review was designed to give an updated overview on the physiological factors and experimental and clinical attempts that were used or may be used in the future as the therapeutic approach to counteract adverse effects in the stomach associated with NSAID ingestion.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Aspirin/adverse effects , Gastric Mucosa/drug effects , Lipoxins/pharmacology , Nitric Oxide/metabolism , Animals , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/adverse effects , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Humans , Lipoxins/biosynthesis , Nitric Oxide Donors/pharmacology
14.
Inflammopharmacology ; 16(1): 40-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18046513

ABSTRACT

Hormonal fluctuations are known to predispose ulceration of the upper gastrointestinal tract, but to date no comparative study of their effects on the healing of pre-existing ulcers in the oral cavity and stomach has been made. We studied the effects of depletion of testosterone and of EGF on the healing of acetic acid-induced ulcers using rats having undergone bilateral orchidectomy and/or salivectomy respectively. We measured alterations in gastric acid secretion and blood flow at ulcer margins, as well as plasma levels of testosterone, gastrin and the proinflammatory cytokines IL-1 beta and TNF-alpha. Testosterone (0.01-10 mg/kg/day i. m.) dose-dependently delayed oral and gastric ulcer healing. When applied in an optimal dose of 1 mg/kg/day, this hormone significantly raised gastric acid secretion and plasma IL-1 beta and TNF-alpha levels. Attenuation of plasma testosterone levels via bilateral orchidectomy inhibited gastric acid secretion and accelerated the healing of oral and gastric ulcers, while increasing plasma gastrin levels and these effects were reversed by testosterone. Salivectomy raised plasma testosterone levels, and delayed oral and gastric ulcer healing. Treatment of salivectomised animals with testosterone further inhibited ulcer healing, and this effect was counteracted by EGF. We propose that testosterone delays ulcer healing via a fall in blood flow at the ulcer margin, a rise in plasma levels of IL-1 beta and TNF-alpha and, in the case of gastric ulcers, an increase in gastric acid secretion. EGF released from the salivary glands plays an important role in limitation of the deleterious effects of testosterone on ulcer healing.


Subject(s)
Chemokines/pharmacology , Epidermal Growth Factor/pharmacology , Gastric Juice/metabolism , Stomach Ulcer/drug therapy , Testosterone/pharmacology , Tongue Diseases/drug therapy , Animals , Chemokines/administration & dosage , Chemokines/metabolism , Dose-Response Relationship, Drug , Epidermal Growth Factor/administration & dosage , Epidermal Growth Factor/metabolism , Gastric Mucosa/blood supply , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastrins/blood , Injections, Intramuscular , Interleukin-1beta/blood , Male , Orchiectomy , Photomicrography/methods , Rats , Rats, Wistar , Regional Blood Flow/drug effects , Stomach Ulcer/blood , Testosterone/administration & dosage , Testosterone/blood , Time Factors , Tongue/blood supply , Tongue/drug effects , Tongue/pathology , Tongue Diseases/blood , Treatment Outcome , Tumor Necrosis Factor-alpha/blood , Wound Healing/drug effects
15.
J Physiol Pharmacol ; 57 Suppl 6: 63-76, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17228088

ABSTRACT

Ghrelin, identified in oxyntic mucosa has been recently implicated in the control of food intake and growth hormone (GH) release but whether this hormone can influence the gastric secretion and gastric mucosal integrity have been little studied. We compared the effects of intraperitoneal (i.p.) and intracerebroventricular (i.c.v.) administration of ghrelin on gastric secretion in rats equipped with gastric fistula (GF) and gastric lesions induced in rats by 75% ethanol and ischemia-reperfusion (I/R) with or without vagotomy or functional ablation of afferent sensory nerves by capsaicin. The number and the area of gastric lesions was measured by planimetry, the GBF was assessed by H(2)-gas clearance method and blood was withdrawn for the determination of the plasma ghrelin and gastrin levels. Ghrelin (5-80 microg/kg i.p. or 600-5000 ng/rat i.c.v.) increased gastric acid secretion and attenuated gastric lesions induced by ethanol and I/R. These protective effects of ghrelin were accompanied by the significant rise in the gastric mucosal blood flow (GBF) and plasma ghrelin and gastrin levels. Ghrelin given i.p. or injected i.c.v. in standard doses 20 microg/kg or 5000 ng/kg, respectively, significantly attenuated the gastric mucosal damage and significantly raised the GBF. Ethanol applied i.g. in smaller concentrations (12.5% and 25%) produced a significant increase in plasma immunorective ghrelin levels and this effect was inhibited in rats receiving ethanol in higher concentrations (75% and 100%). Ghrelin-induced protection after its i.p. or i.c.v. administration and accompanying increase in the GBF were completely abolished by vagotomy and capsaicin-deactivation of sensory nerves. Concurrent treatment with CGRP added to ghrelin restored the gastroprotective and hyperemic effects of ghrelin applied i.p. or i.c.v. in rats with capsaicin denervation. We conclude that central and peripheral ghrelin exerts a potent protective and gastric secretory effects in rats exposed to ethanol and I/R, and that these actions involve vagal nerve integrity, partially depending upon afferent nerves and hyperemia mediated by sensory neuropeptides such as CGRP released from these nerves.


Subject(s)
Gastric Mucosa/drug effects , Ghrelin/pharmacology , Neurons, Afferent/physiology , Protective Agents/pharmacology , Stomach Ulcer/prevention & control , Vagus Nerve/physiology , Animals , Capsaicin/pharmacology , Dose-Response Relationship, Drug , Ethanol , Gastric Acid/metabolism , Gastric Mucosa/metabolism , Gastrins/blood , Ghrelin/blood , Injections, Intraperitoneal , Injections, Intraventricular , Ischemia/complications , Male , Neurons, Afferent/drug effects , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Stomach/blood supply , Stomach Ulcer/chemically induced , Stomach Ulcer/etiology , Vagus Nerve/drug effects , Vagus Nerve/surgery
16.
J Physiol Pharmacol ; 57 Suppl 9: 35-49, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17242486

ABSTRACT

UNLABELLED: The influence of fungal colonization and probiotic treatment on the course of gastric ulcer (GU) and ulcerative colitis (UC) was not explored. Our studies included: 1) clinical investigation of 293 patients with dyspeptic and ulcer complaints and 72 patients with lower gastrointestinal (GI) tract: 60 patients with UC, 12 with irritable bowel syndrome (IBS) - the control group. Significant fungal colonization (SFC), over 10(5) CFU/ml was evaluated. Mycological investigation was performed, including qualitative and quantitative examination, according to Muller method, 2) experimental studies in rats included estimation of the influence of inoculation of Candida isolated from human GI tract on the healing process of GU, induced by acetic acid with or without probiotic Lactobacillus acidophilus (10(6) CFU/ml) introduced intragastrically (i.g.). At 0, 4, 15 and 25 day after ulcer induction. Weight, damage area, gastric blood flow (GBF) (H2 clearance), expression of mRNA for cytokines IL-beta, TNF-alpha (ELISA) were evaluated. Mycology: qualitative and quantitative examination was performed. MPO serum activity was measured. Results of clinical studies: 1) SFC was more frequent in patients with GU: 54.2% of cases and patients with over 5 years history of UC: 33.3% cases. 2) SFC delayed GU healing and influenced the maintenance of clinical symptoms in both diseases. Results of animal studies: 3) In Candida inoculated rats, the GBF was significantly lower than in the vehicle controls (saline administered group). Upregulation of TNF-alpha, IL-1 beta was recorded. The GUs were still present till 25 day in all rats inoculated with Candida, in contrast to vehicle group (reduction of ulcer in 92% at day 25). CONCLUSIONS: 1) Fungal colonization delays process of ulcer and inflammation healing of GI tract mucosa. That effect was attenuated by probiotic therapy. 2) Probiotic therapy seems to be effective in treatment of fungal colonization of GI tract. 3) Lactobacillus acidophilus therapy shortens the duration of fungal colonization of mucosa (enhanced Candida clearance is associated with IL-4, INF-gamma response).


Subject(s)
Candida/isolation & purification , Colitis, Ulcerative/microbiology , Gastrointestinal Tract/microbiology , Lactobacillus acidophilus , Probiotics/therapeutic use , Stomach Ulcer/therapy , Acetic Acid , Adult , Aged , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/therapeutic use , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Candida/classification , Case-Control Studies , Chronic Disease , Colitis, Ulcerative/drug therapy , Colony Count, Microbial , Cytokines/metabolism , Disease Models, Animal , Female , Gastritis/microbiology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/pathology , Humans , Irritable Bowel Syndrome/microbiology , Male , Middle Aged , Rats , Rats, Wistar , Severity of Illness Index , Stomach Ulcer/chemically induced , Stomach Ulcer/metabolism , Stomach Ulcer/microbiology , Time Factors , Treatment Outcome
17.
J Physiol Pharmacol ; 55(4): 773-90, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15613743

ABSTRACT

Previous studies have demonstrated that the gastric mucosa of diabetic rats is highly vulnerable to acute injury but the influence of nonsteroidal anti-inflammatory drugs (NSAID) and their new nitric oxide (NO) releasing derivatives of aspirin (NO-ASA) on the ulcer healing under diabetic conditions has been little studied. In this study streptozocin (STZ, 70 mg/kg injected intraperitoneally) was used to induce diabetes mellitus in rats. Four weeks after STZ injection, gastric ulcers were induced using the acetic acid method and rats with gastric ulcers received the treatment with 1) aspirin (ASA, 30 mg/kg-d i.g.), 2) NO-ASA applied in equimolar dose of 50 mg/kg-d i.g., 3) rofecoxib (5 mg/kg-d i.g.), the selective cyclooxygenase-(COX)-2 inhibitor and 4) SNAP (5 mg/kg-d i.g.), a donor of NO, combined with ASA (30 mg/kg-d i.g.). Ten days after the induction of the ulcers, the healing rate and the gastric blood flow (GBF) were measured by planimetry and hydrogen (H(2))-gas clearance method, respectively and the plasma cytokine such as IL-1beta, TNF-alpha and IL-10 were determined. In addition, the effect of insulin (4 IU/day/rat i.p.) with or without the blockade of NO-synthase by L-NNA (20 mg/kg-d i.p.) on the ulcer healing and the GBF in non-diabetic and diabetic rats was determined. In the diabetic rats, a significant delay in ulcer healing (approximately by 300%) was observed with an accompanied decrease in the GBF at ulcer margin. The prolongation of the healing in diabetic animals was associated with an increase in the plasma cytokine (IL-1beta, TNF-alpha and IL-10) levels. ASA and rofecoxib, that significantly suppressed the mucosal prostaglandin (PG) E(2) generation in ulcer area, delayed significantly the rate of ulcer healing and decreased the GBF at ulcer margin, while elevating plasma IL-1beta, TNF-alpha and IL-10 concentrations in non-diabetic rats and these alterations were significantly augmented in diabetic animals. In contrast to ASA, the treatment with NO-ASA failed to influence both, the ulcer healing and GBF at ulcer margin and significantly attenuated the plasma levels of IL-1beta, TNF-alpha and IL-10 as compared to those recorded in ASA- or rofecoxib-treated animals. Co-treatment of SNAP with native ASA abolished the deleterious effect of ASA on ulcer healing, GBF at ulcer margin and luminal NO release in diabetic rats. Administration of insulin in rats with diabetes, opposed the delay in ulcer healing, and the fall in the GBF at ulcer margin and these effects were counteracted by the concurrent treatment with L-NNA. We conclude that: 1) ulcer healing is dramatically impaired in experimental diabetes and this effect involves the fall in the gastric microcirculation at the ulcer margin and increased release of proinflammatory cytokines; 2) classic NSAID such as ASA and selective COX-2 inhibitors such as rofecoxib, prolong ulcer healing under diabetic conditions probably due to suppression of endogenous PG and the fall in the GBF at the ulcer margin suggesting that both COX isoforms, namely, COX-1 and COX-2, are important sources of PG during ulcer healing in diabetes; and 3) NO-ASA counteracts the impairment of ulcer healing in diabetic rats induced by ASA, mainly due to the release of NO that compensates for PG deficiency resulting in enhancement in the GBF at ulcer margin and suppression of cytokine release in the ulcer area.


Subject(s)
Aspirin/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Nitric Oxide/therapeutic use , Stomach Ulcer/drug therapy , Animals , Aspirin/analogs & derivatives , Chronic Disease , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Male , Nitric Oxide/analogs & derivatives , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Wistar , Stomach Ulcer/complications , Stomach Ulcer/metabolism
18.
Regul Pept ; 120(1-3): 39-51, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15177919

ABSTRACT

Ghrelin, identified in the gastric mucosa has been involved in control of food intake and growth hormone (GH) release but little is known about its influence on gastric secretion and mucosal integrity. The effects of ghrelin on gastric secretion, plasma gastrin and gastric lesions induced in rats by 75% ethanol or 3.5 h of water immersion and restraint stress (WRS) were determined. Exogenous ghrelin (5, 10, 20, 40 and 80 microg/kg i.p.) increased gastric acid secretion and attenuated gastric lesions induced by ethanol and WRS and this was accompanied by the significant rise in plasma ghrelin level, gastric mucosal blood flow (GBF) and luminal NO concentrations. Ghrelin-induced protection was abolished by vagotomy and attenuated by suppression of COX, deactivation of afferent nerves with neurotoxic dose of capsaicin or CGRP(8-37) and by inhibition of NOS with L-NNA but not influenced by medullectomy and administration of 6-hydroxydopamine. We conclude that ghrelin exerts a potent protective action on the stomach of rats exposed to ethanol and WRS, and these effects depend upon vagal activity, sensory nerves and hyperemia mediated by NOS-NO and COX-PG systems.


Subject(s)
Central Nervous System Depressants/toxicity , Ethanol/toxicity , Gastric Mucosa/blood supply , Peptide Hormones/therapeutic use , Stomach Diseases/prevention & control , Adrenergic Agents/administration & dosage , Animals , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Gene-Related Peptide Receptor Antagonists , Capsaicin/pharmacology , Cyclooxygenase 1 , Gastric Acid/metabolism , Gastric Mucosa/pathology , Gastrins/blood , Ghrelin , Growth Hormone/metabolism , Isoenzymes/metabolism , Male , Membrane Proteins , Miotics/pharmacology , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Oxidopamine/administration & dosage , Peptide Fragments/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Wistar , Stomach Diseases/etiology , Stomach Diseases/pathology , Vagotomy , Vagus Nerve/drug effects , Vagus Nerve/metabolism
19.
J Physiol Pharmacol ; 55(2): 325-36, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15213356

ABSTRACT

Ghrelin, a novel peptide expressed in the gastrointestinal tract, especially in the gastric mucosa, exerts several biological activities including the stimulation of appetite and food intake, the stimulation of intestinal motility and the release of growth hormone. The aim of this study was to examine the expression of ghrelin in gastric mucosa after its exposure to ethanol and its effects on gastric lesions induced by ethanol with and without pretreatment with indomethacin. Acute gastric lesions were induced by intragastric administration of 75% ethanol in rats pretreated with saline-vehicle or ghrelin injected intraperitoneally (i.p.) without or with i.p. pretreatment with indomethacin. At the end of experiments, the rats were anesthetized, the stomach was exposed to measure gastric blood flow (GBF), to determine the area of gastric lesions and to take biopsy samples from the oxyntic mucosa for determination of transcripts of ghrelin, tumor necrosis alpha (TNF-alpha) and transforming growth factor alpha (TGFalpha) using RT-PCR and to assess the generation of PGE(2) by RIA. Exposure of gastric mucosa to 75% ethanol resulted in numerous mucosal lesions of an area of about 115 mm(2) and in the increase of mucosal expression of TNF-alpha, PGE(2), TGFalpha and ghrelin with concomitant decrease in GBF. Exogenous ghrelin reduced dose-dependently acute gastric lesions with simultaneous attenuation of GBF and a decrease in the expression of TNF-alpha but not TGFalpha. Pretreatment with indometahcin, which suppressed the generation of PGE(2) by about 85%, augmented ethanol-induced gastric lesions and eliminated the ghrelin-induced protection of mucosa against ethanol. We conclude that ghrelin, whose mucosal expression is enhanced after exposure to ethanol, exhibits a strong gastroprotection, at least in part, due to its anti-inflammatory action mediated by prostaglandins.


Subject(s)
Gastric Mucosa/metabolism , Peptide Hormones/biosynthesis , Stomach Diseases/metabolism , Animals , Blotting, Western , Dinoprostone/biosynthesis , Disease Models, Animal , Dose-Response Relationship, Drug , Ethanol , Gastric Mucosa/drug effects , Ghrelin , Indomethacin , Male , Peptide Hormones/pharmacology , Rats , Rats, Wistar , Regional Blood Flow/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Stomach/blood supply , Stomach/drug effects , Stomach Diseases/chemically induced , Stomach Diseases/prevention & control , Transforming Growth Factor alpha/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
20.
J Physiol Pharmacol ; 55(1 Pt 2): 165-77, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15082876

ABSTRACT

Limitation of the damage to the organs such as heart, liver, intestine, stomach and brain by an earlier brief complete occlusion of their arteries is defined as ischemic preconditioning (IP). No study so for has been undertaken to check whether brain-gut axis is involved in the gastroprotection exhibited by gastric IP or in that induced by repeated brief episodes of ischemia of remote organs such as heart and liver. This study was designed to determine the possible involvement of vagal and sensory afferent nerves, in the mechanism of gastric and remote organ IP on the gastric mucosa in rats exposed to prolonged ischemia-reperfusion with or without functional ablation of sensory nerves by capsaicin or in those with removed vagal innervation by vagotomy. This gastric IP was induced by short ischemia episodes (occlusion of celiac artery 1-5 times for 5 min) applied 30 min before subsequent ischemia followed by 3 h of reperfusion (I/R) and compared with remote IP induced by occlusion of left descending coronary artery or hepatic artery plus portal vein. The area of gastric lesions was determined by planimetry, gastric blood flow (GBF) was measured by H(2)-gas clearance method and mucosal biopsy samples were taken for the assessment of calcitonin gene-related peptide (CGRP) by RIA. Exposure of gastric mucosa to standard 3 h of I/R produced numerous gastric lesions and significant fall in the GBF and mucosal CGRP content. Two 5 min short ischemic episodes by occlusion of coronary or hepatic arteries, significantly reduced gastric damage induced by I/R with the extent similar to that exhibited by two short (5 min) episodes of gastric ischemia. These protective effects of gastric and remote IPs were accompanied by a restoration of the fall in the CGRP content caused by I/R alone. Protection and hyperemia induced by gastric IP were significantly attenuated in capsaicin-denervated or vagotomized animals and completely removed in those exposed to the combination of vagotomy and capsaicin-denervation. The IP-induced protection and hyperemia were restored by the administration of exogenous CGRP to gastric IP in capsaicin-treated animals. Gastroprotective and hyperemic actions of remote IP were markedly diminished in capsaicin-denervated rats and in those subjected to vagotomy. We conclude that brief ischemia in remote organs such as heart and liver protects gastric mucosa against gastric injury induced by I/R as effectively as gastric IP via mechanism involving both vagal and sensory nerves releasing vasodilatatory mediators such as CGRP.


Subject(s)
Brain/physiology , Gastric Mucosa/blood supply , Gastric Mucosa/physiopathology , Gastrointestinal Tract/physiology , Ischemic Preconditioning/methods , Reperfusion Injury/prevention & control , Animals , Calcitonin Gene-Related Peptide/administration & dosage , Calcitonin Gene-Related Peptide/pharmacokinetics , Capsaicin/administration & dosage , Capsaicin/adverse effects , Celiac Artery/injuries , Coronary Vessels/anatomy & histology , Coronary Vessels/injuries , Heart Injuries/etiology , Heart Injuries/physiopathology , Injections, Subcutaneous , Liver/blood supply , Liver/injuries , Liver/innervation , Male , Neurons, Afferent/drug effects , Neurons, Efferent/drug effects , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacokinetics , Rats , Rats, Wistar , Reperfusion Injury/etiology , Stomach Diseases/drug therapy , Stomach Diseases/physiopathology , Stomach Diseases/prevention & control , Vagotomy/methods , Vagus Nerve/drug effects , Vagus Nerve/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...