1
|
Chakraborty S, Yadav SK, Subramanian M, Priyadarsini KI, Iwaoka M, Chattopadhyay S. DL-trans-3,4-Dihydroxy-1-selenolane (DHSred) accelerates healing of indomethacin-induced stomach ulceration in mice. Free Radic Res 2012; 46:1378-86. [DOI: 10.3109/10715762.2012.718766] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
2
|
Adhikary B, Yadav SK, Bandyopadhyay SK, Chattopadhyay S. Epigallocatechin gallate accelerates healing of indomethacin-induced stomach ulcers in mice. Pharmacol Rep 2011; 63:527-36. [PMID: 21602608 DOI: 10.1016/s1734-1140(11)70519-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 08/04/2010] [Indexed: 12/13/2022]
Abstract
Management of the gastric toxicity of non-steroidal anti-inflammatory drugs (NSAIDs) remains a crucial problem because the commercially available drugs have side effects and are often expensive. Therefore, we examined the potential of the green tea-derived polyphenol epigallocatechin gallate (EGCG) to treat indomethacin-induced stomach ulcers in mice. Administration of indomethacin (18 mg/kg, po) to mice induced ulceration in the glandular portion of the gastric mucosa, accompanied by increased lipid peroxidation (LPO) and protein oxidation and reductions in thiol defense, mucin, cyclooxygenase (COX) expression and prostaglandin (PG) synthesis in the gastric tissues. Daily oral administration of EGCG (2 mg/kg) or omeprazole (3 mg/kg) for 3 days produced similar (≈ 72-75%, p < 0.001) beneficial effects on the acute gastric ulceration. Treatment with the test samples partially reversed all the adverse oxidative effects of indomethacin. In addition, EGCG, but not omeprazole, enhanced expression of the COX isoforms and PG synthesis. The results suggest that the non-toxic and inexpensive tea polyphenol EGCG may be an excellent candidate for further evaluation as a potent anti-ulcer drug.
Collapse
Affiliation(s)
- Biplab Adhikary
- Department of Biochemistry, Dr. B.C. Roy Post Graduate Institute of Basic Medical Sciences & IPGME&R, 244B, Acharya Jagadish Chandra Bose Road, Kolkata - 700 020, India
| | | | | | | |
Collapse
|
3
|
Chiou SK, Hoa N, Hodges A. Sulindac sulfide induces autophagic death in gastric epithelial cells via Survivin down-regulation: A mechanism of NSAIDs-induced gastric injury. Biochem Pharmacol 2011; 81:1317-23. [DOI: 10.1016/j.bcp.2011.03.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 03/18/2011] [Accepted: 03/22/2011] [Indexed: 11/17/2022]
|
4
|
Black tea and theaflavins assist healing of indomethacin-induced gastric ulceration in mice by antioxidative action. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2010; 2011. [PMID: 20953434 PMCID: PMC2952312 DOI: 10.1155/2011/546560] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 05/06/2010] [Accepted: 08/06/2010] [Indexed: 12/19/2022]
Abstract
The healing activities of black tea (BT) and the theaflavins (TF) against the indomethacin-induced stomach ulceration were studied in a mouse model. Indomethacin (18 mg/kg, p.o.) administration induced maximum ulceration in the glandular portion of the gastric mucosa on the 3rd day, accompanied by increased lipid peroxidation and protein oxidation, depletion of thiol-defense and mucin, as well as reduced expressions of cyclooxygenases (COX) and prostaglandin (PG) E synthesis in the gastric tissues, and plasma total antioxidant status of mice. Treatment with BT (40 mg/kg), TF (1 mg/kg), and omeprazole (3 mg/kg) produced similar (74%-76%) ulcer healing, as revealed from the histopathological studies. Treatment with all the above samples reversed the adverse oxidative effects of indomethacin significantly. BT and TF also enhanced the PGE synthesis by augmenting the expressions of COX 1 and 2, but did not modulate acid secretion.
Collapse
|
5
|
Banerjee D, Bhattacharya S, Bandyopadhyay SK, Chattopadhyay S. Biochemical mechanism of healing activity of the natural phenolic, allylpyrocatechol against indomethacin-induced gastric ulceration in mice. Dig Dis Sci 2008; 53:2868-77. [PMID: 18431645 DOI: 10.1007/s10620-008-0266-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Accepted: 03/26/2008] [Indexed: 12/24/2022]
Abstract
Indomethacin caused maximum stomach ulceration in mice on the 3rd day, which was associated with reduction of plasma total antioxidant status (TAS), COX-1, COX-2, mucosal PGE(2), VEGF, and vWF, along with an increase in endostatin levels. Treatment with the phytochemical allylpyrocatechol (5 mg/kg, p.o. for 3 days) provided significant ulcer healing by reversing these biochemical parameters, as well as increasing the EGF expression more than that observed due to ulceration. Omeprazole (3 mg/kg, p.o. for 3 days) provided a similar healing by improving TAS and mucin levels, without significantly altering the other parameters.
Collapse
Affiliation(s)
- Debashish Banerjee
- Department of Biochemistry, Dr. B.C. Roy Post Graduate Institute of Basic Medical Sciences, Kolkata, 700 020, India
| | | | | | | |
Collapse
|
6
|
Banerjee D, Maity B, Nag SK, Bandyopadhyay SK, Chattopadhyay S. Healing potential of Picrorhiza kurroa (Scrofulariaceae) rhizomes against indomethacin-induced gastric ulceration: a mechanistic exploration. Altern Ther Health Med 2008; 8:3. [PMID: 18237397 PMCID: PMC2266895 DOI: 10.1186/1472-6882-8-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Accepted: 01/31/2008] [Indexed: 12/31/2022]
Abstract
Background The present study was undertaken to evaluate the potential of the rhizomes of the Indian medicinal plant, Picrorhiza kurroa in healing indomethacin-induced acute stomach ulceration in mice and examine its capacity to modulate oxidative stress and the levels of prostaglandin (PGE2) and EGF during the process. Methods Male swiss albino mice, ulcerated with indomethacin (18 mg/kg, p. o., single dose) were treated up to 7 days with different doses of the methanol extract of P. kurroa rhizomes (designated as PK). The healing capacity of the most effective dose of PK (20 mg/kg, p. o. × 3 d) was compared with that of omeprazole (Omez) (3 mg/kg, p. o. × 3 d). The effects of the drug-treatment for one and three days on the biochemical parameters were assessed by comparing the results with that of untreated mice of the 1st and 3rd day of ulceration. The stomach tissues of the mice were used for the biochemical analysis. Results The macroscopic indices revealed maximum ulceration on the 3rd day after indomethacin administration, which was effectively healed by PK. Under the optimized treatment regime, PK and Omez reduced the ulcer indices by 45.1% (P < 0.01), and 76.3% respectively (P < 0.001), compared to the untreated ulcerated mice. Compared to the ulcerated untreated mice, those treated with PK for 3 days showed decreased the levels of thiobarbituric acid reactive substances (TBARS) (32.7%, P < 0.05) and protein carbonyl (37.7%, P < 0.001), and increased mucin (42.2%, P < 0.01), mucosal PGE2 (21.4%, P < 0.05), and expressions of COX-1 and 2 (26.9% and 18.5%, P < 0.05), EGF (149.0%, P < 0.001) and VEGF (56.9%, P < 0.01). Omez reduced the TBARS (29.4%, P < 0.05), and protein carbonyl (38.9%, P < 0.001), and increased mucin (38.3%, P < 0.01), without altering the other parameters significantly. Conclusion PK (20 mg/kg, p. o. × 3 days) could effectively heal indomethacin-induced stomach ulceration in mice by reducing oxidative stress, and promoting mucin secretion, prostaglandin synthesis and augmenting expressions of cyclooxygenase enzymes and growth factors.
Collapse
|
7
|
Hattori Y, Ohno T, Ae T, Saeki T, Arai K, Mizuguchi S, Saigenji K, Majima M. Gastric mucosal protection against ethanol by EP2 and EP4 signaling through the inhibition of leukotriene C4 production. Am J Physiol Gastrointest Liver Physiol 2008; 294:G80-7. [PMID: 17947453 DOI: 10.1152/ajpgi.00292.2007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prostaglandin (PG)E derivatives are widely used for treating gastric mucosal injury. PGE receptors are classified into four subtypes, EP(1), EP(2), EP(3), and EP(4). We have tested which EP receptor subtypes participate in gastric mucosal protection against ethanol-induced gastric mucosal injury and clarified the mechanisms of such protection. The gastric mucosa of anesthetized rats was perfused at 2 ml/min with physiological saline, agonists for EP(1), EP(2), EP(3), and EP(4), or 50% ethanol, using a constant-rate pump connected to a cannula placed in the esophagus. The gastric microcirculation of the mucosal base of anesthetized rats was observed by transillumination through a window made by removal of the adventitia and muscularis externa. PGE(2) and subtype-specific EP agonists were applied to the muscularis mucosae at the window. Application of 50% ethanol dilated the mucosal arterioles and constricted the collecting venules. Collecting venule constriction by ethanol was completely inhibited by PGE(2) and by EP(2) and EP(4) agonists (100 nM) but not by an EP(1) or an EP(3) agonist. Ethanol-induced mucosal injury was also inhibited by EP(2) and EP(4) agonists. When leukotriene (LT)C(4) levels in the perfusate of the gastric mucosa were determined by ELISA, intragastric ethanol administration elevated the LTC(4) levels sixfold from the basal levels. These elevated levels were significantly (60%) reduced by both EP(2) and EP(4) agonists but not by other EP agonists. Since LTC(4) application at the window constricted collecting venules strongly, and an LTC antagonist reduced ethanol-induced mucosal injury, reductions in LTC(4) generation in response to EP(2) and EP(4) receptor signaling may be relevant to the protective action of PGE(2). The present results indicate that EP(2) and EP(4) receptor signaling inhibits ethanol-induced gastric mucosal injury through cancellation of collecting venule constriction by reducing LTC(4) production.
Collapse
Affiliation(s)
- Youichiro Hattori
- Dept. of Pharmacology, Kitasato Univ. School of Medicine, Sagamihara, Kanagawa 228-8555, Japan
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Shimokawa O, Matsui H, Nagano Y, Kaneko T, Shibahara T, Nakahara A, Hyodo I, Yanaka A, Majima HJ, Nakamura Y, Matsuzaki Y. Neoplastic transformation and induction of H+,K+-adenosine triphosphatase by N-methyl-N′-nitro-N-nitrosoguanidine in the gastric epithelial RGM-1 cell line. In Vitro Cell Dev Biol Anim 2007; 44:26-30. [DOI: 10.1007/s11626-007-9067-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Accepted: 10/04/2007] [Indexed: 12/19/2022]
|
9
|
Healing properties of malabaricone B and malabaricone C, against indomethacin-induced gastric ulceration and mechanism of action. Eur J Pharmacol 2007; 578:300-12. [PMID: 17977527 DOI: 10.1016/j.ejphar.2007.09.041] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Revised: 09/20/2007] [Accepted: 09/24/2007] [Indexed: 12/13/2022]
Abstract
The healing activity of malabaricone B and malabaricone C, the major antioxidant constituents of the spice Myristica malabarica against the indomethacin-induced gastric ulceration in mice has been studied. The histological indices revealed maximum ulceration on the 3rd day after indomethacin administration, which was effectively healed by malabaricone B, malabaricone C (each 10 mg/kg body weight/day) and omeprazole (3 mg/kg body weight/day) for 3 days. Compared to the untreated ulcerated mice, treatment with malabaricone B, malabaricone C and omeprazole reduced the ulcer indices by 60.3% (P<0.01), 88.4% and 86.1% respectively (P<0.001). All the test samples accelerated ulcer healing than observed in natural recovery even after 7 days. Stomach ulceration reduced the total antioxidant status of plasma by 41% (P<0.05), which was significantly increased by malabaricone B (36%, P<0.01), malabaricone C (61%, P<0.001) and omeprazole (53%, P<0.001). Compared to the ulcerated untreated mice, those treated with malabaricone B reduced the levels of thiobarbituric acid reactive substances and protein carbonyls by 17% and approximately 34% respectively (P<0.05), while malabaricone C and omeprazole reduced the parameters almost equally (approximately 30%, P<0.01, and approximately 40%, P<0.01 respectively). Likewise, all the test samples reduced the oxidation of protein and non-protein thiols significantly (P<0.05). The antioxidant activity of the test samples could partly account their healing capacities. However, the differential potency of them was explainable by considering their relative abilities to modulate mucin secretion, PGE(2) synthesis and expression of EGF receptor and COX isoforms, malabaricone C being most effective in controlling all these factors.
Collapse
|
10
|
Aihara E, Nomura Y, Sasaki Y, Ise F, Kita K, Takeuchi K. Involvement of prostaglandin E receptor EP3 subtype in duodenal bicarbonate secretion in rats. Life Sci 2007; 80:2446-53. [PMID: 17512019 DOI: 10.1016/j.lfs.2007.04.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Revised: 03/31/2007] [Accepted: 04/09/2007] [Indexed: 11/23/2022]
Abstract
We investigated the involvement of prostaglandin E (PGE) receptor subtype EP3 in the regulatory mechanism of duodenal HCO(3)(-) secretion in rats. A proximal duodenal loop or a chambered stomach was perfused with saline, and HCO(3)(-) secretion was measured using a pH-stat method and by adding 2 mM HCl. Mucosal acidification was achieved through 10 min of exposure to 10 mM HCl in the duodenum or 100 mM HCl in the stomach. Various EP agonists or the EP4 antagonist were given i.v., while the EP1 or EP3 antagonist was given s.c. or i.d., respectively. Sulprostone (EP1/EP3 agonists) stimulated duodenal HCO(3)(-) secretion in a dose-dependent manner, and this response was inhibited by AE5-599 (EP3 antagonist) but not AE3-208 (EP4 antagonist). AE1-329 (EP4 agonist) also increased duodenal HCO(3)(-) secretion, and this action was inhibited by AE3-208 but not AE5-599. The response to PGE(2) or acidification in the duodenum was partially attenuated by AE5-599 or AE3-208 alone but completely abolished by the combined administration. Duodenal damage caused by mucosal perfusion with 150 mM HCl for 4 h was worsened by pretreatment with AE5-599 and AE3-208 as well as indomethacin and further aggravated by co-administration of these antagonists. Neither the EP3 nor EP4 antagonist had any effect on the gastric response induced by PGE(2) or acidification. These results clearly demonstrate the involvement of EP3 receptors, in addition to EP4 receptors, in the regulation of duodenal HCO(3)(-) secretion as well as the maintenance of the mucosal integrity of the duodenum against acid injury.
Collapse
Affiliation(s)
- Eitaro Aihara
- Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
11
|
Ushida Y, Shimokawa Y, Toida T, Matsui H, Takase M. Bovine α-Lactalbumin Stimulates Mucus Metabolism in Gastric Mucosa. J Dairy Sci 2007; 90:541-6. [PMID: 17235130 DOI: 10.3168/jds.s0022-0302(07)71537-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Bovine alpha-lactalbumin (alpha-LA), a major milk protein, exerts strong gastroprotective activity against rat experimental gastric ulcers induced by ethanol or stress. To elucidate the mechanisms underlying this activity, the influence of alpha-LA on gastric mucus metabolism was investigated in vitro and in vivo. For the in vitro study, RGM1 cells (a rat gastric epithelial cell line) were selected for observation of the direct activity of alpha-LA on gastric mucosal cells and cultured in the presence of either alpha-LA or ovalbumin (OVA), a reference protein showing no gastroprotective activity. Amounts of synthesized and secreted mucin, a major component of mucus, were determined using [3H]glucosamine as a tracer, and prostaglandin E2 (PGE2) levels in the culture medium were determined by RIA. For the in vivo study, the thickness of the mucus gel layer, a protective barrier for gastric mucosa, was evaluated histochemically in rat gastric mucosa. alpha-Lactalbumin (3 mg/mL) significantly stimulated mucin synthesis and secretion in RGM1 cells and also increased PGE2 levels in the culture medium. In contrast, OVA showed no enhancing effects under identical conditions. Neither indomethacin, a cyclo-oxygenase inhibitor, nor AH23848, a prostaglandin EP4 receptor antagonist, affected alpha-LA-induced enhancement of mucin synthesis and secretion. In vivo, oral administration of alpha-LA (300 mg/kg x 3 times/d x 7 d) increased the thickness of the mucus gel layer in rats. These results indicate that alpha-LA fortifies the mucus gel layer by stimulating mucin production and secretion in gastric mucus-producing cells, and that this enhancing effect is independent of endogenous PGE2. Comparison of the efficacy of alpha-LA with OVA suggests that the activities observed in RGM1 cells are closely related to the gastroprotective effects in rat gastric ulcer models. In conclusion, alpha-LA stimulates mucus metabolism, and this action may be responsible for its gastroprotective activity.
Collapse
Affiliation(s)
- Y Ushida
- Biochemical Research Laboratory, Morinaga Milk Industry Co. Ltd., Zama, Kanagawa 228-8583, Japan.
| | | | | | | | | |
Collapse
|
12
|
Becker JC, Grosser N, Waltke C, Schulz S, Erdmann K, Domschke W, Schröder H, Pohle T. Beyond gastric acid reduction: Proton pump inhibitors induce heme oxygenase-1 in gastric and endothelial cells. Biochem Biophys Res Commun 2006; 345:1014-21. [PMID: 16712795 DOI: 10.1016/j.bbrc.2006.04.170] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Accepted: 04/28/2006] [Indexed: 10/24/2022]
Abstract
Proton pump inhibitors (PPIs) have been demonstrated to prevent gastric mucosal injury by mechanisms independent of acid inhibition. Here we demonstrate that both omeprazole and lansoprazole protect human gastric epithelial and endothelial cells against oxidative stress. This effect was abrogated in the presence of the heme oxygenase-1 (HO-1) inhibitor ZnBG. Exposure to either PPI resulted in a strong induction of HO-1 expression on mRNA and protein level, and led to an increased activity of this enzyme. Expression of cyclooxygenase isoforms 1 and 2 remained unaffected, and COX-inhibitors did not antagonize HO-1 induction by PPIs. Our results suggest that the antioxidant defense protein HO-1 is a target of PPIs in both endothelial and gastric epithelial cells. HO-1 induction might account for the gastroprotective effects of PPIs independently of acid inhibition, especially in NSAID gastropathy. Moreover, our findings provide additional perspectives for a possible but yet unexplored use of PPIs in vasoprotection.
Collapse
Affiliation(s)
- Jan C Becker
- Department of Medicine B, University of Muenster, 48149 Muenster, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Gower WR, Carter GM, McAfee Q, Solivan SM. Identification, regulation and anti-proliferative role of the NPR-C receptor in gastric epithelial cells. Mol Cell Biochem 2006; 293:103-18. [PMID: 16786190 DOI: 10.1007/s11010-006-9234-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Accepted: 05/11/2006] [Indexed: 11/30/2022]
Abstract
Evidence suggests that functional atrial natriuretic peptide (ANP) receptors occur in surface gastric mucosal epithelial cells. To evaluate functional aspects of ANP in a model of these cells we examined the expression of natriuretic peptide receptors (NPR) subtypes A and C in the non-transformed rat gastric mucosal epithelial cell line RGM1. Transcripts for NPR-A and NPR-C were detected in RGM1 cells by RT-PCR. However, only NPR-C protein was detected by Western blot and immunohistochemical analyses. Specific saturable binding of (125)I-ANP to RGM1 cells revealed a single class of high affinity binding sites (K (d) = 208 +/- 71pM, B (max) = 110,000 +/- 14,000 sites/cell, Hill coefficient = 0.97 +/- 0.05). ANP (IC(50) 130 +/- 47pM), BNP (IC(50) 716 +/- 26 pM), CNP (IC(50) 356 +/- 85pM) and C-ANP (IC(50) 134 +/- 13pM), a specific ligand for NPR-C, effectively displaced (125)I-ANP binding. Cross-linking of (125)I-ANP to cells labeled predominantly a protein of 66,000 Da. These data suggest that (125)I-ANP binding was primarily to NPR-C. ANP and C-ANP inhibited forskolin- and prostaglandin E(2) (PGE(2))-stimulated cAMP in a PTx-sensitive fashion. PGE(2), transforming growth factor-+/-1 (TGF-+/-1), forskolin, 8-bromo-cyclic AMP, and phorbol-12-myristate-13-acetate (PMA) caused a dose-dependent decrease in specific (125)I-ANP binding, whereas epidermal growth factor (EGF), 8-bromo-cyclic GMP and 4+/--phorbol didecanoate had no effect. PGE(2), forskolin, TGF-+/-1 and PMA significantly decreased (125)I-ANP B (max) values, NPR-C protein and steady-state NPR-C transcript levels. H89, a protein kinase A inhibitor, blocked the reduction of NPR-C mRNA produced by both forskolin and PGE(2.) GF109203X, a protein kinase C inhibitor, abolished the PMA-induced decrease in NPR-C transcripts but only partially blocked that produced by TGF-+/-1. RGM1 cells exhibited a dose-dependent decrease in both DNA synthesis and cell proliferation when cultured in the presence of ANP or C-ANP. These findings indicate that RGM1 cells express functional NPR-C receptors that can influence RGM1 cell proliferation and are down-regulated by PGE(2) and TGF-+/-1.
Collapse
Affiliation(s)
- William R Gower
- Surgery and Research Services, James A. Haley Veterans Administration Hospital, 13000 Bruce B. Downs Boulevard, Tampa, FL 33612-4745, USA.
| | | | | | | |
Collapse
|
14
|
Naito Y, Kajikawa H, Mizushima K, Shimozawa M, Kuroda M, Katada K, Takagi T, Handa O, Kokura S, Ichikawa H, Yoshida N, Matsui H, Yoshikawa T. Rebamipide, a gastro-protective drug, inhibits indomethacin-induced apoptosis in cultured rat gastric mucosal cells: association with the inhibition of growth arrest and DNA damage-induced 45 alpha expression. Dig Dis Sci 2005; 50 Suppl 1:S104-12. [PMID: 16184411 DOI: 10.1007/s10620-005-2814-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Accepted: 06/06/2005] [Indexed: 12/09/2022]
Abstract
Rebamipide, a gastromucosal protective drug, suppresses indomethacin-induced gastropathy in humans and rodents. Effects of rebamipide on gene expression in indomethacin-treated gastric mucosal cells (RGM1) were investigated using high-density oligonucleotide arrays. Indomethacin induced apoptosis in RGM1 cells in a dose-dependent manner. Rebamipide pretreatment significantly reduced indomethacin-induced apoptosis. We used gene expression profiling on high-density oligonucleotide probe arrays to characterize the transcriptional response of RGM1 cells to indomethacin treatment for 6 hr. Of the 8,799 probes examined, 717 (8.1%) were induced (400 probes) or repressed (317 probes) at least 1.5-fold. Among the 158 genes that were induced by indomethacin at least 2.0-fold, four genes that were down-regulated by rebamipide at least 2.0-fold are listed: growth arrest and DNA-damage-inducible 45 alpha (GADD 45 alpha), golgi SNAP receptor complex member 1, iodothyronine deiodinases, and transcription factor 8. Real time-PCR confirmed GADD 45 alpha expression and its inhibition by rebamipide. Inhibition of apoptosis-related genes is possibly important for the cytoprotective effect of rebamipide against indomethacin-induced gastric mucosal cell injury.
Collapse
Affiliation(s)
- Yuji Naito
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Katada K, Naito Y, Shimozawa M, Mizushima K, Kuroda M, Takagi T, Kokura S, Ichikawa H, Yoshida N, Matsui H, Yoshikawa T. Gene expression analysis following hypoxia-reoxygenation in rat gastric epithelial cells using a high-density oligonucleotide array. Redox Rep 2005; 9:337-42. [PMID: 15720829 DOI: 10.1179/135100004225006849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Recent investigations have demonstrated that the signaling of hypoxia-re-oxygenation is a major contributing pathway leading to gastric mucosal injury induced by stress, non-steroidal anti-inflammatory drugs, and Helicobacter pylori. The aim of the present study was to perform a gene expression analysis on the gastric mucosal cellular response to hypoxia-reoxygenation using a high-density oligonucleotide array. Cells were subjected to hypoxia with 95% N(2) and 5% CO(2) at 37 degrees C for 2 h. Reoxygenation was initiated by placing the cells in an environment of normoxia for 2 h. Total RNA was extracted, and differences in gene expression profiles between the normoxia and hypoxia-reoxygenation groups were investigated using a GeneChip of Rat Toxicology U34 array (Affymetrix). Hypoxia-reoxygenation up-regulated the stress-related genes (heat shock protein-70 [HSP-70], catalase). The enhanced expression of HSP-70 was confirmed by Western blot analysis. In conclusion, these results suggest that up-regulation of the HSP-70 gene after reoxygenation may play a role in maintaining cell survival and supporting cell function as a molecular chaperone.
Collapse
Affiliation(s)
- Kazuhiro Katada
- Department of Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Handa O, Naito Y, Takagi T, Shimozawa M, Kokura S, Yoshida N, Matsui H, Cepinskas G, Kvietys PR, Yoshikawa T. Tumor necrosis factor-alpha-induced cytokine-induced neutrophil chemoattractant-1 (CINC-1) production by rat gastric epithelial cells: role of reactive oxygen species and nuclear factor-kappaB. J Pharmacol Exp Ther 2004; 309:670-6. [PMID: 14747614 DOI: 10.1124/jpet.103.062216] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Rat cytokine-induced neutrophil chemoattractant-1 (CINC-1), a counterpart of the human growth-regulated oncogene product (GRO), has been suggested to participate in neutrophil recruitment in an experimental model of gastritis in rat. However, the mechanism(s) involved in regulation of CINC-1 production by the gastric mucosa remains unclear. The aim of this study was to investigate the mechanism(s) of CINC-1 production by rat gastric mucosa in vitro. All experiments were performed using rat normal gastric mucosal cell line (RGM-1). RGM-1s were stimulated with tumor necrosis factor (TNF)-alpha, and CINC-1 mRNA levels (reverse transcription-polymerase chain reaction) and protein secretion (enzyme-linked immunosorbent assay) were assessed. The production of reactive oxygen species (ROS) and nuclear factor (NF)-kappaB activation (translocation to the nuclei) in response to TNF-alpha stimulation was evaluated using fluorescence microscopy in the presence or absence of the inhibitors of mitochondrial electron flow and NF-kappaB activation. Stimulation of RGM-1 cells with TNF-alpha resulted in an increase in intracellular oxidative stress, NF-kappaB translocation to the nuclei, and up-regulation of CINC-1 mRNA and protein, which was prevented by interfering with mitochondria-dependent ROS production and NF-kappaB activation. Taken together, these findings indicate that CINC-1, a counterpart of the human GRO, production by rat gastric epithelial cells in response to TNF-alpha stimulation is an oxidant stress-mediated and NF-kappaB-dependent event.
Collapse
Affiliation(s)
- Osamu Handa
- Vascular Cell Biology/Inflammation Program, Lawson Health Research Institute, London, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Takezono Y, Joh T, Oshima T, Suzuki H, Seno K, Yokoyama Y, Alexander JS, Itoh M. Role of prostaglandins in maintaining gastric mucus-cell permeability against acid exposure. ACTA ACUST UNITED AC 2004; 143:52-8. [PMID: 14749685 DOI: 10.1016/j.lab.2003.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Regulation of gastric epithelial permeability is important in the protection of the gastric mucosa from secreted acid. However, the mechanism(s) for this regulation in gastric mucus cells remains unknown. In this study, we evaluated gastric epithelial-cell permeability in response to acid exposure by monitoring trans-epithelial electrical resistance (TEER) and paracellular permeability with carbon 14-labeled mannitol. We also examined the role of prostaglandins on gastric epithelial permeability. Rat gastric epithelial cells (RGM-1) were plated on 8-microm-pore tissue-culture inserts. Cells were exposed to solutions of differing pH (3-7.4), with and without the nonsteroidal antiinflammatory drug (NSAID) indomethacin (10(-7) mol/L), for 60 to 120 minutes. Transepithelial permeability was measured on the basis of TEER and the diffusion rate of [(14)C]mannitol. Prostaglandin E(2) (PGE(2)) was administered in some experiments with NSAIDs. After acid exposure (pH 3.0-5.0), TEER rapidly and significantly increased, peaking in 5 minutes. Diffusion of [(14)C]mannitol was blocked during the period when TEER increased. Pretreatment with the cyclooxygenase (COX) inhibitor indomethacin blocked the rapid acid-induced increase in TEER. A specific COX-2 inhibitor had no effect on this rapid increase in TEER. The blockade by indomethacin was eliminated by the addition of PGE(2). These findings suggest that when gastric-surface mucus cells are exposed to acid, gastric epithelial permeability decreases rapidly to inhibit acid back-diffusion. Prostaglandins play an important role in this protective response to acid exposure. COX inhibitors such as indomethacin may inhibit the regulation of epithelial permeability by reducing the concentration of PGE(2).
Collapse
Affiliation(s)
- Yasuhide Takezono
- Department of Internal Medicine and Bioregulation, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Kato S, Aihara E, Nakamura A, Xin H, Matsui H, Kohama K, Takeuchi K. Expression of vanilloid receptors in rat gastric epithelial cells: role in cellular protection. Biochem Pharmacol 2003; 66:1115-21. [PMID: 12963499 DOI: 10.1016/s0006-2952(03)00461-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Vanilloid receptors subtype 1 (VR1), a nonselective cation channel responsive to capsaicin, protons, and noxious heat, has been recently identified in not only neural but also non-neural cells. In the present study, we demonstrated the peripheral expression of VR1 in gastric mucosal epithelial cells and investigated the role of the receptor in cellular protection. The rat gastric mucosal epithelial cell line was used. The expression of VR1 was examined by Western blotting and RT-PCR. Cell damage was induced by immersion in 10% ethanol or acid (pH 4.0) for 30 min, and cell viability was determined by MTT assay. Capsaicin or resiniferatoxin was added 30 min before the challenge with ethanol or acid, while capsazepine or ruthenium red (a VR1 antagonist) was added simultaneously with capsaicin. The distinct expression of VR1 protein and mRNA was detected in rat gastric mucosal epithelial cell line as well as in the rat stomach and spinal cord by Western blotting and RT-PCR, respectively. The cDNA sequence of the PCR product was found to be almost identical to that of the authentic VR1 (99.8%) when the product was subcloned and sequenced. On the other hand, the cell damage induced by ethanol or acid was dose-dependently prevented by pretreatment with capsaicin. The protective effect of capsaicin was mimicked by resiniferatoxin and almost totally abolished by co-addition of capsazepine or ruthenium red. These findings suggest that VR1 is expressed peripherally in gastric mucosal epithelial cells and plays a cellular protective role.
Collapse
Affiliation(s)
- Shinichi Kato
- Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto 607-8414, Japan.
| | | | | | | | | | | | | |
Collapse
|
19
|
Yuki T, Ishihara S, Rumi M, Ortega-Cava Cesar F, Kadowaki Y, Kazumori H, Yuki M, Wada T, Miyaoka Y, Yoshino N, Kinoshita Y. Expression of midkine and receptor-like protein tyrosine phosphatase (RPTP)-beta genes in the rat stomach and the influence of rebamipide. Aliment Pharmacol Ther 2003; 18 Suppl 1:106-12. [PMID: 12925147 DOI: 10.1046/j.1365-2036.18.s1.12.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
BACKGROUND Midkine has been reported to bind to receptor-like protein tyrosine phosphatase (RPTP)-beta and to play important roles in growth and differentiation of various cells. Midkine is expressed in rat stomach during experimental ulcer healing, suggesting that the midkine-RPTP-beta system has some physiological functions in the stomach. Rebamipide is a mucoprotective drug used for the treatment of gastric ulcers. We have tested the hypothesis that the ulcer healing mechanism stimulated by rebamipide is linked physiologically to the gastric midkine-RPTP-beta system. MATERIALS AND METHODS Seven-week-old-male Wistar rats were used. Midkine and RPTP-beta gene expression in rat stomach was investigated by laser capture microdissection coupled with the reverse transcription-polymerase chain reaction (RT-PCR). The effects of rebamipide on midkine and RPTP-beta expression in rat stomach and the gastric epithelial cell line RGM1 were evaluated by RT-PCR and Northern blot analyses. RESULTS Midkine and RPTP-beta expression was detected in the gastric mucosal, submucosal and muscle layers. Rebamipide stimulated both midkine and RPTP-beta expression in rat stomach and RGM1 cells. CONCLUSION Rebamipide may protect the gastric mucosa by regulating midkine and RPTP-beta expression.
Collapse
Affiliation(s)
- T Yuki
- Department of Internal Medicine II, Shimane Medical University, Shimane, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Joh T, Takezono Y, Oshima T, Sasaki M, Seno K, Yokoyama Y, Ohara H, Nomura T, Alexander JS, Itoh M. The protective effect of rebamipide on paracellular permeability of rat gastric epithelial cells. Aliment Pharmacol Ther 2003; 18 Suppl 1:133-8. [PMID: 12925151 DOI: 10.1046/j.1365-2036.18.s1.15.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Barrier function in gastric epithelial cells is essential for the gastric defence mechanism against acid back-diffusion into the mucosal layer. Our previous study indicated that trans-epithelial resistance (TER) of rat gastric epithelial cells was rapidly increased when the cells were exposed to acid. This response to acid was diminished by indometacin. AIM Evaluate the effects of a mucoprotective agent, rebamipide, on the nonsteroidal anti-inflammatory drug (NSAID)-induced increase of gastric epithelial permeability. METHODS Rat gastric epithelial cells were plated on tissue culture inserts. Cells were exposed to a NSAID (indometacin, 10-7 M). Trans-epithelial permeability was measured by TER and diffusion rate of 14C-mannitol. The effect of rebamipide was evaluated by measuring TER. Endogenous prostaglandin E2 (PGE2) production in culture medium was also measured. RESULTS Indometacin gradually and significantly decreased TER and increased 14C-manitol permeability. Rebamipide reversed the indometacin-induced changes in epithelial permeability and induced PGE2 synthesis. This induction was blocked by either indometacin or a Cyclooxygenase (COX)-2 specific inhibitor. CONCLUSIONS COX inhibitors such as indometacin inhibit regulation of epithelial permeability by reducing PGE2. COX-1 has an important role in the gastric defense mechanism. Rebamipide suppressed an indometacin-induced increase in gastric epithelial permeability by increasing PGE2 levels in a COX-2 dependent manner.
Collapse
Affiliation(s)
- T Joh
- Department of Internal Medicine & Bioregulation, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Hoshino T, Tsutsumi S, Tomisato W, Hwang HJ, Tsuchiya T, Mizushima T. Prostaglandin E2 protects gastric mucosal cells from apoptosis via EP2 and EP4 receptor activation. J Biol Chem 2003; 278:12752-8. [PMID: 12556459 DOI: 10.1074/jbc.m212097200] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Prostaglandin E(2) (PGE(2)) has a strong protective effect on the gastric mucosa in vivo; however, the molecular mechanism of a direct cytoprotective effect of PGE(2) on gastric mucosal cells has yet to be elucidated. Although we reported previously that PGE(2) inhibited gastric irritant-induced apoptotic DNA fragmentation in primary cultures of guinea pig gastric mucosal cells, we show here that PGE(2) inhibits the ethanol-dependent release of cytochrome c from mitochondria. Of the four main subtypes of PGE(2) receptors, we also demonstrated, using subtype-specific agonists, that EP(2) and EP(4) receptors are involved in the PGE(2)-mediated protection of gastric mucosal cells from ethanol-induced apoptosis. Activation of EP(2) and EP(4) receptors is coupled with an increase in cAMP, for which a cAMP analogue was found here to inhibit the ethanol-induced apoptosis. The increase in cAMP is known to activate both protein kinase A (PKA) and phosphatidylinositol 3-kinase pathways. An inhibitor of PKA but not of phosphatidylinositol 3-kinase blocked the PGE(2)-mediated protection of cells from ethanol-induced apoptosis, suggesting that a PKA pathway is mainly responsible for the PGE(2)-mediated inhibition of apoptosis. Based on these results, we considered that PGE(2) inhibited gastric irritant-induced apoptosis in gastric mucosal cells via induction of an increase in cAMP and activation of PKA, and that this effect was involved in the PGE(2)-mediated protection of the gastric mucosa from gastric irritants in vivo.
Collapse
Affiliation(s)
- Tatsuya Hoshino
- Faculty of Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | | | | | | | | | | |
Collapse
|
22
|
Becker JC, Hertel M, Markmann A, Shahin M, Werner S, Domschke W, Pohle T. Dynamics and localization of activin A expression in rat gastric ulcers. Scand J Gastroenterol 2003; 38:260-7. [PMID: 12737440 DOI: 10.1080/00365520310000636a] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Activin A, the homodimer of the activin/inhibin betaA subunit, has been shown to participate in cutaneous wound healing. In this study we intended to determine its part in gastric ulceration. METHODS Activin A expression was studied by immunohistochemistry and in situ hybridization in acetic-acid-induced chronic gastric ulcers in rat. The dynamics of this process were also assessed by quantitative real time RT-PCR and RNase protection assays (RPA). The effects of different doses of this cytokine on epithelial and mesenchymal cell proliferation were quantitated in vitro. RESULTS Low amounts of activin A and its mRNA were expressed by epithelia, endothelia and fibroblasts in intact gastric tissue. Granulation tissue of gastric ulcers and gastric glands adjacent to the ulcer rim expressed markedly increased amounts of activin protein as well as activin/inhibin betaA mRNA. RPA and RT-PCR studies revealed a more than 3-fold increase in the relative abundance of this mRNA. Activin A did not affect the proliferation rate of fibroblasts and epithelial cells in vitro. CONCLUSIONS Activin A participates in gastric ulcer healing in a similar fashion as in cutaneous wounding. Its expression on protein and mRNA level is markedly increased in ulcer base and rim.
Collapse
Affiliation(s)
- J C Becker
- Dept. of Medicine B, Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | | | | | | | | | | | | |
Collapse
|
23
|
Naito Y, Handa O, Takagi T, Ishikawa T, Imamoto E, Nakagawa S, Yamaguchi T, Yoshida N, Matsui H, Yoshikawa T. Ubiquitin-proteasome inhibitor enhances tumour necrosis factor-alpha-induced apoptosis in rat gastric epithelial cells. Aliment Pharmacol Ther 2002; 16 Suppl 2:59-66. [PMID: 11966525 DOI: 10.1046/j.1365-2036.16.s2.30.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
BACKGROUND Tumour necrosis factor (TNF-alpha) is a candidate factor for involvement in inflammation-mediated gastric mucosal injury. However, the effect of this cytokine on gastric epithelial cells has been poorly investigated. In the present study, we examined whether gastric epithelial cells are resistant to TNF-alpha-induced apoptosis, and whether this resistance is related to ubiquitin-proteasome-associated nuclear factor-kappaB (NF-kappaB) activation. METHODS The rat gastric mucosal cell line RGM-1 was grown in DMEM/F12 medium supplemented with 10% FCS. Confluent monolayers of cells were pretreated or not for 60 min with PSI, a peptide aldehyde known to specifically inhibit the chymotrypsin-like activity of 26S proteasome. Cells were subsequently stimulated with recombinant rat TNF-alpha and their viability was determined by WST-1 assay. Apoptosis was confirmed by fluorescence microscopy after staining with Hoechst 33342 and propidium iodide, and DNA fragmentation was determined by flow cytometry using an APO-BRDU kit. IkappaB-alpha and the p65 binding subunit of NF-kappaB were detected by Western blots. RESULTS Twenty-four-hour incubation with TNF-alpha alone or PSI alone did not affect the cell viability of RGM-1 cells. Pretreatment with PSI significantly enhanced the level of apoptosis induced by TNF-alpha. In RGM-1 cells treated with TNF-alpha, cytoplasmic IkappaB-alpha decreased and p65 in nuclear extracts increased markedly 30 min after cytokine stimulation. Pretreatment with PSI at 12.5 micromol/L blocked these TNF-alpha-induced changes. CONCLUSION PSI enhances TNF-alpha-induced apoptosis through inhibition of NF-kappaB activation in RGM-1 cells.
Collapse
Affiliation(s)
- Y Naito
- First Department of Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Nakaya A, Wakabayashi H, Imamura L, Fukuta K, Makimoto S, Naganuma K, Orihara T, Minemura M, Shimizu Y, Nagasawa T, Hamazaki T, Watanabe A. Helicobacter pylori alters n-6 fatty acid metabolism and prostaglandin E2 synthesis in rat gastric mucosal cells. J Gastroenterol Hepatol 2001; 16:1197-1205. [PMID: 11903735 DOI: 10.1046/j.1440-1746.2001.02541.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Little is known about whether Helicobacter pylori infection alters fatty acid metabolism in gastric mucosal cells. By using cultured rat gastric mucosal cells (RGM-1), we investigated the effect of H. pylori broth culture filtrates on this point. Furthermore, our study aimed to find out whether n-6 long chain polyunsaturated fatty acids from linoleic acid are formed in RGM-1 cells. METHODS Rat gastric mucosal cells were incubated with 10, 20 and 40 microg/mL of linoleic acid or medium alone. Phosphatidylcholine content extracted from whole RGM-1 cells was quantitated by using a densitometer, and its fatty acid composition was analyzed by using gas chromatography. Prostaglandin E2 concentration in the culture medium was measured by using radioimmunoassay. The expression of cyclooxygenase (COX)-1 and COX-2 was examined by using reverse transcription-polymerase chain reaction. In addition, after incubation with [1-14C] linoleic acid, radioactivities of both linoleic acid and arachidonic acid components of the PC fraction were counted. The effects of H. pylori broth culture filtrates on PC content, its fatty acid composition and prostaglandin (PG)E2 synthesis were also assessed. RESULTS Linoleic acid addition caused an increase in the composition of arachidonic acid, as well as linoleic acid, and also in PGE2 concentration. Cyclo-oxygenase-2 expression was induced in RGM-1 cells by the addition of linoleic acid. In addition, [1-14C] linoleic acid added to the culture medium was converted to [1-14C] arachidonic acid in RGM-1 cells. Helicobacter pylori broth culture filtrates decreased linoleic acid composition and increased arachidonic acid composition. Moreover, after incubation with H. pylori broth culture filtrates, PGE2 concentrations were higher than that of the controls. CONCLUSIONS These findings suggest the presence of fatty acid elongase and Delta5- and Delta6-desaturases synthesize arachidonic acid from linoleic acid in RGM-1 cells. Thus, H. pylori infection may enhance PGE2 synthesis and accelerate n-6 fatty acid metabolism in gastric mucosal cells, which could make the gastric mucosal barrier more fragile.
Collapse
Affiliation(s)
- A Nakaya
- Third Department of Internal Medicine, Faculty of Medicine, Toyama Medical and Pharmaceutical University, Toyama, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Moriyama N, Ishihara S, Hirose M, Watanabe S, Sato N, Kinoshita Y. E-cadherin is essential for gastric epithelial restitution in vitro: a study using the normal rat gastric mucosal cell line RGM1. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 2001; 138:236-42. [PMID: 11574817 DOI: 10.1067/mlc.2001.118177] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The proliferation and migration of epithelial cells appear to have important roles in intercellular adhesion and the regeneration of gastric mucosal lesions. However, the role of E-cadherin, an important intercellular adhesion molecule, in restitution after gastric mucosal damage is unknown. This study was designed to investigate the possible role of E-cadherin in the regeneration of gastric mucosal lesions. Artificial small wounds were made in an RGM1 confluent monolayer sheet, and the healing process was monitored with or without the presence of different concentrations of anti-E-cadherin antibodies. E-cadherin mRNA and protein expression were determined by Northern blot analysis and immunostaining, respectively. Epithelial restitution in anti-E-cadherin antibody-treated monolayers was inhibited as compared with that in the controls without antibody. E-cadherin mRNA and protein expression were up-regulated transiently during the healing process. E-cadherin plays an important role during wound healing of gastric epithelial lesions and is crucial for sheet migration.
Collapse
Affiliation(s)
- N Moriyama
- Second Department of Internal Medicine, Shimane Medical University, Shimane, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Hiraoka S, Miyazaki Y, Kitamura S, Toyota M, Kiyohara T, Shinomura Y, Mukaida N, Matsuzawa Y. Gastrin induces CXC chemokine expression in gastric epithelial cells through activation of NF-kappaB. Am J Physiol Gastrointest Liver Physiol 2001; 281:G735-G742. [PMID: 11518686 DOI: 10.1152/ajpgi.2001.281.3.g735] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Although hypergastrinemia is frequently observed in individuals with a chronic Helicobacter pylori infection, its pathophysiological significance in gastric mucosal inflammation is unclear. The present study was designed to determine if gastrin induces the expression of CXC chemokines in gastric epithelial cells. Human and rat gastric epithelial cells, transfected with gastrin receptor, were stimulated with gastrin. The expression of mRNAs for human interleukin-8 (IL-8) and rat cytokine-induced neutrophil chemoattractant-1 and release of human IL-8 protein were then determined by Northern blot analysis and ELISA, respectively. Gastrin not only induced the expression of mRNAs for these chemokines but also stimulated IL-8 protein release. A luciferase assay using IL-8 promoter genes showed that nuclear factor (NF)-kappaB is absolutely required and activator protein-1 (AP-1) is partly required for the maximum induction of IL-8 by gastrin. An electrophoretic mobility shift assay revealed that gastrin is capable of activating both NF-kappaB and AP-1. In addition, the inhibition of NF-kappaB abrogated gastrin-induced chemokine expression. These results suggest that gastrin is capable of upregulating CXC chemokines in gastric epithelial cells and therefore may contribute to the progression of the inflammatory process in the stomach.
Collapse
Affiliation(s)
- S Hiraoka
- Department of Internal Medicine and Molecular Science, Graduate School of Medicine, Osaka University, 565-0871 Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Pohle T, Becker JC, Markmann A, Lügering N, Pauels HG, Konturek JW, Domschke W. Aspirin effects on gastric epithelial cell proliferation and cytokine expression. Microsc Res Tech 2001; 53:354-9. [PMID: 11376496 DOI: 10.1002/jemt.1103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Aspirin is known to cause gastric injury and to delay ulcer healing. The effects of aspirin on gastric epithelial cell function are heterogeneous; in contrast to injuring the mucosa, aspirin may also act beneficially by inducing adaptation; a mechanism that is poorly understood. We aimed to document the effects of different doses of aspirin on gastric epithelial cell function defined as proliferation, and secretion as well as mRNA expression of cytokines. Furthermore, we studied the effects of aspirin pretreatment on cytokine secretion as a potential element of gastric adaptation. The proliferative activity of three different gastric epithelial cell lines (AGS, KATO III, RGM-1) was assessed by (3)H-thymidine incorporation; secretion of growth factors PDGF-AB and VEGF into culture supernatant was documented by ELISA. mRNA transcripts of both cytokines were quantified by real time RT-PCR. Low doses of aspirin did not alter the proliferative dynamics in two of the three studied cell lines; high doses abolished proliferation. Secretion of PDGF-AB and VEGF increased during the first days of low dose aspirin exposition; higher concentrations led to a depletion of cytokines after an initial liberation in the case of VEGF, mRNA of which was also dose-dependently increased by aspirin. Seven-day pretreatment with low amounts of aspirin did not alter the secretory response of the epithelia caused by higher doses of this drug. The secretion of cytokines and proliferation of gastric epithelial cells are adversely effected by aspirin in a similarly dose-dependent fashion as the intended effects of this drug on platelet function and pain relief.
Collapse
Affiliation(s)
- T Pohle
- Department of Medicine B, University of Münster, Münster, Germany.
| | | | | | | | | | | | | |
Collapse
|
28
|
Hollande F, Blanc EM, Bali JP, Whitehead RH, Pelegrin A, Baldwin GS, Choquet A. HGF regulates tight junctions in new nontumorigenic gastric epithelial cell line. Am J Physiol Gastrointest Liver Physiol 2001; 280:G910-21. [PMID: 11292600 DOI: 10.1152/ajpgi.2001.280.5.g910] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The regulation of intercellular adhesion by hepatocyte growth factor (HGF) was examined on a novel nontumorigenic gastric epithelial cell line (IMGE-5) derived from H-2Kb-tsA58 transgenic mice. IMGE-5 cells constitutively expressed cytokeratin 18 and HGF receptors. Under permissive conditions (33 degrees C + interferon-gamma), IMGE-5 cells proliferated rapidly but did not display membrane expression of adherens and tight junction proteins. Under nonpermissive conditions, their proliferation was decreased and they displayed a strong, localized membrane expression of E-cadherin/beta-catenin and occludin/ZO-1. HGF treatment largely prevented the targeting of ZO-1 to the tight junction and induced a significant decrease of the transepithelial resistance measured across a confluent IMGE-5 cell monolayer. HGF rapidly increased the tyrosine phosphorylation of ZO-1 and decreased its association with occludin in a phosphatidylinositol 3-kinase (PI 3-kinase)-dependent manner. PI 3-kinase was also involved in HGF-induced migration of IMGE-5 cells. Our results demonstrate that 1) HGF prevents the appearance of ZO-1 in the membrane during epithelial cell differentiation; 2) HGF causes partial relocalization of ZO-1 to the cytoplasm and nucleus and concomitantly stimulates cell dissociation and migration; and 3) IMGE-5 cells offer a useful model for the study of gastric epithelial cell differentiation.
Collapse
Affiliation(s)
- F Hollande
- Laboratoire de Biochimie des Membranes, Faculté de Pharmacie, 15 av. C. Flahault, 34060 Montpellier cedex, France.
| | | | | | | | | | | | | |
Collapse
|
29
|
Takeuchi K, Kato S, Tanaka A. [Gastrointestinal cytoprotection by prostaglandin E and EP receptor subtypes]. Nihon Yakurigaku Zasshi 2001; 117:274-82. [PMID: 11338377 DOI: 10.1254/fpj.117.274] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Endogenous prostaglandins (PGs) play important roles in modulating the mucosal integrity and various functions of the gastrointestinal tract. Among them, E-type PGs are most effective in these actions. This article reviews recent studies dealing with the relationship of the cytoprotective action of PGE2- and EP-receptor subtypes in the gastrointestinal mucosa. PGE2 exerts gastric cytoprotection against HCl/ethanol and indomethacin. These effects were mimicked by only EP1 agonists and attenuated by EP1 antagonists. Likewise, the adaptive cytoprotection induced by a mild irritant was attenuated by EP1 antagonists as well as indomethacin. On the other hand, the protective effect of dmPGE2 against indomethacin-induced small intestinal lesions was mimicked by only EP3 and EP4 agonists. Similar results were obtained in EP-receptor knockout mice; i.e., PGE2 failed to exhibit both direct and adaptive cytoprotection in EP1-receptor knockout mice, while the protective action in both the duodenum and small intestine was hampered in EP3-receptor knockout mice. The underlying mechanism related to these actions of PGE2 in the stomach, duodenum or small intestine may be related to inhibition of stomach contraction, stimulation of duodenal alkaline secretion, or suppression of bacterial translocation due to inhibition of intestinal contraction as well as stimulation of mucus secretion, respectively.
Collapse
Affiliation(s)
- K Takeuchi
- Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto 607-8414, Japan.
| | | | | |
Collapse
|
30
|
Kunikata T, Araki H, Takeeda M, Kato S, Takeuchi K. Prostaglandin E prevents indomethacin-induced gastric and intestinal damage through different EP receptor subtypes. JOURNAL OF PHYSIOLOGY, PARIS 2001; 95:157-63. [PMID: 11595431 DOI: 10.1016/s0928-4257(01)00021-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Gastrointestinal ulcerogenic effect of indomethacin is causally related with an endogenous prostaglandin (PG) deficiency, yet the detailed mechanism remains unknown. We examined the effect of various PGE analogues specific to EP receptor subtypes on these lesions in rats and mice, and investigated which EP receptor subtype is involved in the protective action of PGE(2). Fasted or non-fasted animals were given indomethacin s.c. at 35 mg/kg for induction of gastric lesions or 10-30 mg/kg for intestinal lesions, and they were killed 4 or 24 h later, respectively. Various EP agonists were given i.v. 10 min before indomethacin. Indomethacin caused hemorrhagic lesions in both the stomach and intestine. Prior administration of 16,16-dimethyl PGE(2) (dmPGE(2)) prevented the development of damage in both tissues, and the effect in the stomach was mimicked by 17-phenyl PGE2 (EP1), while that in the small intestine was reproduced by ONO-NT-012 (EP3) and ONO-AE-329 (EP4). Butaprost (EP2) did not have any effect on either gastric or intestinal lesions induced by indomethacin. Similar to the findings in rats, indomethacin caused gastric and intestinal lesions in both wild-type and knockout mice lacking EP1 or EP3 receptors. However, the protective action of dmPGE(2) in the stomach was observed in wild-type and EP3 receptor knockout mice but not in mice lacking EP1 receptors, while that in the intestine was observed in EP1 knockout as well as wild-type mice but not in the animals lacking EP3 receptors. These results suggest that indomethacin produced damage in the stomach and intestine in a PGE(2)-sensitive manner, and exogenous PGE(2) prevents gastric and intestinal ulcerogenic response to indomethacin through different EP receptor subtypes; the protection in the stomach is mediated by EP1 receptors, while that in the intestine mediated by EP3/EP4 receptors.
Collapse
MESH Headings
- 16,16-Dimethylprostaglandin E2/pharmacology
- Animals
- Anti-Ulcer Agents/pharmacology
- Cyclooxygenase Inhibitors/pharmacology
- Dinoprostone/metabolism
- Dinoprostone/physiology
- Indomethacin/pharmacology
- Intestines/drug effects
- Intestines/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout/genetics
- Rats
- Rats, Sprague-Dawley
- Receptors, Prostaglandin E/agonists
- Receptors, Prostaglandin E/deficiency
- Receptors, Prostaglandin E/genetics
- Receptors, Prostaglandin E/physiology
- Receptors, Prostaglandin E, EP1 Subtype
- Receptors, Prostaglandin E, EP3 Subtype
- Stomach/drug effects
- Stomach/pathology
Collapse
Affiliation(s)
- T Kunikata
- Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto 607-8414, Japan
| | | | | | | | | |
Collapse
|
31
|
Suetsugu H, Ishihara S, Moriyama N, Kazumori H, Adachi K, Fukuda R, Watanabe M, Kinoshita Y. Effect of rebamipide on prostaglandin EP4 receptor gene expression in rat gastric mucosa. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 2000; 136:50-7. [PMID: 10882227 DOI: 10.1067/mlc.2000.107303] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Prostaglandin E2 (PGE2) plays an important role in the regulation of gastric mucus secretion. We have previously shown that the prostaglandin EP4 receptor (EP4) gene is abundantly expressed in gastric mucus-producing cells. Furthermore, we have shown that EP4 is present in a rat normal gastric mucosal cell line (RGM1) and that PGE2 increases mucus secretion from these cells via EP4. Rebamipide, an anti-gastric ulcer agent, has been reported to promote gastric PGE2 production and mucus secretion. However, it is unclear whether rebamipide influences mucus secretion by altering expression of the EP4 gene. Therefore, we tested the effect of rebamipide on EP4 gene expression in the gastric mucosa. Seven-week-old Wistar rats received oral rebamipide (100 mg/kg) with and without water-immersion restraint stress (WRS). All rats were killed, and their gastric tissues were used to investigate the expression of mRNA for EP4 and cyclooxygenase types 1 and 2. The thickness of the gastric mucus layer was also measured. The effect of rebamipide on EP4 gene expression and PGE2 production in RGM1 cells was also investigated in vitro. Furthermore, the effect of PGE2 on cyclic adenosine monophosphate (cAMP) production by RGM1 cells with or without rebamipide was studied. Oral rebami-pide significantly increased EP4 gene expression in the gastric antrum but not in the corpus after WRS. Furthermore, it increased surface mucus thickness and suppressed ulcer formation in the gastric mucosa after WRS. In vitro, rebamipide significantly augmented EP4 gene expression in RGM1 cells, and PGE2 significantly increased the cAMP production by RGM1 cells incubated with rebamipide. Rebamipide promotes EP4 gene expression and may consequently increase the gastric mucus secretion via EP4 receptors in the rat antral mucosa.
Collapse
Affiliation(s)
- H Suetsugu
- Department of Internal Medicine II, Shimane Medical University, Izumo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Northey A, Denis D, Cirino M, Metters KM, Nantel F. Cellular distribution of prostanoid EP receptors mRNA in the rat gastrointestinal tract. Prostaglandins Other Lipid Mediat 2000; 62:145-56. [PMID: 10938408 DOI: 10.1016/s0090-6980(00)00058-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The inhibition of PGE(2) synthesis resulting from sustained NSAIDs therapy has been linked to gastrointestinal irritations and ulceration. The multiple physiological effects of PGE(2) in the gut are mediated through the activation of four receptors termed EP(1-4). The aim of the study was to determine the precise distribution of the four prostaglandin E(2) receptors in the rat stomach, small intestine, and colon. We used non-radioactive in situ hybridization techniques on paraffin-embedded tissue. Mucous cells of the stomach and goblet cells of the small intestine and colon were found to express mRNA for all four EP subtypes. A positive hybridization signal for EP(1), EP(3), and EP(4) was detected in the parietal cells of the stomach whereas the chief cells expressed low levels of EP(1) and EP(3). The EP(1) and EP(3) receptor mRNA could also be detected in the muscularis mucosa, longitudinal muscle and enteric ganglias of the stomach and small intestine. However, close examination of the enteric ganglias indicated that most of the positive labeling was localized to the glial cells, although some neurons did express EP(3). In conclusion, we have detailed the distribution of prostanoid EP receptors in the gut at the cellular level, giving new insights to the role of prostaglandins in gastrointestinal functions.
Collapse
MESH Headings
- Animals
- In Situ Hybridization
- Intestinal Mucosa/chemistry
- Intestinal Mucosa/cytology
- Intestine, Large/chemistry
- Intestine, Large/cytology
- Intestine, Large/innervation
- Intestine, Small/chemistry
- Intestine, Small/cytology
- Intestine, Small/innervation
- Male
- Muscle, Smooth/chemistry
- Muscle, Smooth/cytology
- RNA, Complementary
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- Rats
- Rats, Sprague-Dawley
- Receptors, Prostaglandin E/analysis
- Receptors, Prostaglandin E/genetics
- Stomach/chemistry
- Stomach/cytology
- Stomach/innervation
Collapse
Affiliation(s)
- A Northey
- Department of Biochemistry and Molecular Biology, Merck Frosst Center for Therapeutic Research, P.O. Box 1005, Dorval-Pointe-Claire, H9R 4P8, Québec, Canada
| | | | | | | | | |
Collapse
|
33
|
Naito Y, Yoshikawa T, Boku Y, Fujii T, Masui Y, Tanaka Y, Fujita N, Yoshida N, Kondo M. Protective role of intracellular glutathione against nitric oxide-induced necrosis in rat gastric mucosal cells. Aliment Pharmacol Ther 2000; 14 Suppl 1:145-52. [PMID: 10807416 DOI: 10.1046/j.1365-2036.2000.014s1145.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Nitric oxide synthase activity is increased in the stomach in association with Helicobacter pylori infection and portal hypertension, but the mechanism by which nitric oxide contributes to mucosal damage remains unclear. AIM To examine whether nitric oxide injures gastric mucosal cells and whether cellular glutathione affects nitric oxide-induced cytotoxicity. METHODS A confluent monolayer of RGM-1 gastric mucosal cells was exposed to nitric oxide donors (NOC5 or NOC12). Cell viability was determined by trypan blue dye exclusion, lactate dehydrogenase release and supravital staining with Hoechst 33342 and propidium iodide. The kinetics of the reduced/oxidized forms of glutathione were also measured, as well as the effect of glutathione-depletion or glutathione-precursor treatment on nitric oxide-induced cytotoxicity. RESULTS Excess exogenous nitric oxide produced by NOC5 or NOC12 induced necrosis in RGM-1 cells in a time- and concentration-dependent manner. The level of reduced glutathione drastically decreased prior to the loss of cell viability and remained low, but oxidized glutathione was not affected. Glutathione depletion increased necrosis of both NOCs in an NOC-concentration-related fashion, while pre-treatment with gamma-glutamylcysteine ethyl ester reduced their necrotic susceptibility. CONCLUSION Exogenous nitric oxide induced necrosis in gastric mucosal cells, and intracellular reduced glutathione protects gastric mucosal cells from damage by nitric oxide.
Collapse
Affiliation(s)
- Y Naito
- First Department of Medicine, Kyoto Prefectural University of Medicine, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Araki H, Ukawa H, Sugawa Y, Yagi K, Suzuki K, Takeuchi K. The roles of prostaglandin E receptor subtypes in the cytoprotective action of prostaglandin E2 in rat stomach. Aliment Pharmacol Ther 2000; 14 Suppl 1:116-24. [PMID: 10807413 DOI: 10.1046/j.1365-2036.2000.014s1116.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
AIM To investigate the EP receptor subtype involved in the gastroprotective action of prostaglandin (PG) E2 using various EP receptor agonists in rats, and using knockout mice lacking EP1 or EP3 receptors. METHODS Male SD rats and C57BL/6 mice were used after an 18-h fast. Gastric lesions were induced by oral administration of HCl/ethanol (150 mM HCl in 60% ethanol). Rats were given various EP agonists i.v. 10 min before HCl/ethanol: PGE2, sulprostone (EP1/EP3 agonist), butaprost (EP2 agonist), 17-phenyl-omega-trinorPGE2 (17-phenylPGE2: EP1 agonist), ONO-NT012 (EP3 agonist) and 11-deoxyPGE1 (EP3/EP4 agonist). In a separate study, the effect of PGE2 on HCl/ethanol lesions was examined in EP1- and EP3-receptor knockout mice. RESULTS Gastric lesions induced by HCl/ethanol were dose dependently prevented by PGE2: this effect was mimicked by sulprostone and 17-phenylPGE2 and was significantly antagonized by ONO-AE-829, an EP1 antagonist. Neither butaprost, ONO-NT012 nor 11-deoxyPGE1 exhibited any protective activity against HCl/ethanol-induced gastric lesions. PGE2 caused an inhibition of gastric motility as well as an increase of mucosal blood flow and mucus secretion, the effects being mimicked by prostanoids activating EP1 receptors, EP2/EP3/EP4 receptors and EP4 receptors, respectively. On the other hand, although HCl/ethanol caused similar damage in both wild-type mice and knockout mice lacking EP1 or EP3 receptors, the cytoprotective action of PGE2 observed in wild-type and EP3-receptor knockout mice totally disappeared in mice lacking EP1 receptors. CONCLUSION The gastric cytoprotective action of PGE2 is mediated by activation of EP1 receptors. This effect may be functionally associated with inhibition of gastric motility but not with increased mucosal blood flow or mucus secretion.
Collapse
Affiliation(s)
- H Araki
- Department of Pharmacology & Experimental Therapeutics, Kyoto Pharmaceutical University, Yamashina, Japan
| | | | | | | | | | | |
Collapse
|
35
|
Suzuki H, Mori M, Seto K, Shibata F, Nagahashi S, Kawaguchi C, Suzuki M, Matsui H, Watanabe K, Miura S, Ishii H. Rat CXC chemokine GRO/CINC-1 paradoxically stimulates the growth of gastric epithelial cells. Aliment Pharmacol Ther 2000; 14 Suppl 1:94-100. [PMID: 10807410 DOI: 10.1046/j.1365-2036.2000.014s1094.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND CXC chemokines such as interleukin (IL)-8 are neutrophil chemoattractants, the levels of which increase in Helicobacter pylori-infected gastric mucosa. Many investigators have focused on the chemotactic aspects of IL-8: however, CXC chemokines are also reported to have angiogenic activity and to serve as remodelling factors. Rat GRO/CINC-1 is a rodent counterpart of human GROalpha, a member of the family of CXC chemokines. Gastric mucosa infected with H. pylori is in a state of hyperproliferation, with increases in the amounts of growth factors such as hepatocyte growth factor (HGF). AIM To investigate whether rat GRO/CINC-1 had growth-stimulating activity for gastric epithelial cells. METHODS The rat gastric epithelial cell line RGM-1 was incubated in serum-free medium for 12 h to adjust the cell cycle to the G0 phase, and GRO/CINC-1 was then added for 24 h. The total cell number was determined by fluorogenic analysis after propidium iodide staining, and cell proliferation was assessed by measuring 5-bromo-2'-deoxyuridine (BrdU) incorporation. The activity of p42/p44 mitogen-activated protein kinase (MAPK) was measured 5-20 min after the start of GRO/CINC-1 exposure. RESULTS Cultures treated with GRO/CINC-1 showed a significant increase in cell number and BrdU incorporation in a concentration-dependent fashion. The MAPK activity increased within 5 min after GRO/CINC-1 application and returned to the control level at 20 min. CONCLUSION The growth-stimulatory effect of GRO/CINC-1 on rat gastric epithelial cells suggests a dual function of this chemokine: proinflammatory action and induction of epithelial proliferation.
Collapse
Affiliation(s)
- H Suzuki
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Takahashi S, Takeuchi K, Okabe S. EP4 receptor mediation of prostaglandin E2-stimulated mucus secretion by rabbit gastric epithelial cells. Biochem Pharmacol 1999; 58:1997-2002. [PMID: 10591156 DOI: 10.1016/s0006-2952(99)00286-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Prostaglandin (PG) E receptors are divided into four subtypes (EP1-EP4). We investigated the EP receptor subtype involved in PGE2-stimulated mucus secretion by rabbit gastric epithelial cells. Northern blot analysis revealed that epithelial cells express EP3 and EP4 receptor mRNAs, but neither EP1 nor EP2 receptor mRNAs were detected. PGE2, 11-deoxy-PGE1 (an EP3/EP4/EP2 agonist) and 16,16-dimethyl-PGE2 (an EP3/EP2/EP4 agonist) concentration-dependently promoted mucus secretion. In contrast, 17-phenyl-PGE2 (an EP3/EP1 agonist), sulprostone (an EP3/EP1 agonist), and butaprost (an EP2 agonist) failed to stimulate secretion. The effective concentrations of PGE2, 11-deoxy-PGE1, and 16,16-dimethyl-PGE2 were associated with their affinities for the EP4 receptor. In addition, PGE2, 11-deoxy-PGE1, and 16,16-dimethyl-PGE2 increased cyclic AMP (cAMP) production, but the other prostanoids had no effect. SQ22536 [9-(tetrahydro-2'-furyl)adenine; an adenylate cyclase inhibitor] inhibited both the increased cAMP production and mucus secretion induced by PGE2, 11-deoxy-PGE1, and 16,16-dimethyl-PGE2. H-89 (N-[2-((p-bromocinnamyl)amino)ethyl]-5-isoquinoline sulfonamide; a protein kinase A inhibitor) also abolished the stimulatory effects of the prostanoids on mucus secretion, but calphostin C (a protein kinase C inhibitor) did not. These results indicate that PGE2 promotes mucus secretion by rabbit gastric epithelial cells, mediated through EP4 receptor stimulation and the subsequent activation of protein kinase A.
Collapse
Affiliation(s)
- S Takahashi
- Department of Applied Pharmacology, Kyoto Pharmaceutical University, Japan.
| | | | | |
Collapse
|
37
|
Nakamura E, Takahashi S, Ishikawa M, Okabe S. Inhibitory effect of macrophage-derived factors on the recovery of wounds induced in rat gastric epithelial monolayers. Biochem Pharmacol 1999; 58:1221-7. [PMID: 10484080 DOI: 10.1016/s0006-2952(99)00202-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The effect of macrophage supernatant on the recovery of wounds induced in rat gastric epithelial RGM1 monolayers was investigated. The repair of wounds induced in the monolayers of RGM1 cells was accelerated time-dependently by 10 ng/mL of transforming growth factor-alpha (TGF-alpha). TGF-alpha also significantly stimulated DNA synthesis in RGM1 cells for 24 hr. Upon treatment of the cells with the macrophage supernatant, spontaneous and TGF-alpha-stimulated restoration was inhibited in a time- and concentration-dependent manner. After 24 hr, TGF-alpha-enhanced restoration was eliminated completely by the supernatant at 10(6) cells/mL. Similarly, the macrophage supernatant suppressed the spontaneous and TGF-alpha-stimulated DNA syntheses in a concentration-dependent manner. The macrophage supernatant at 10(6) cells/mL contained 0.4 ng/mL of interleukin-1beta (IL-1beta). Interleukin-1 receptor antagonist (IL-1RA) reversed the inhibition induced by the macrophage supernatant in a concentration-dependent manner. Nonetheless, pretreatment with IL-1RA had no effects on the spontaneous and TGF-alpha-stimulated DNA syntheses. Reverse transcription-polymerase chain reaction analysis revealed that RGM1 cells express mRNA for IL-1 receptor type 1, but not for type 2. These results indicate that macrophages can inhibit the spontaneous and TGF-alpha-stimulated recovery of wounds induced in gastric epithelial monolayers. The inhibitory effects of the supernatant are suggested to be partially mediated through a IL-1beta/IL-1 receptor type 1 pathway.
Collapse
Affiliation(s)
- E Nakamura
- Department of Applied Pharmacology, Kyoto Pharmaceutical University, Japan.
| | | | | | | |
Collapse
|
38
|
Ohno T, Katori M, Majima M, Saeki T, Boku K, Nishiyama K, Hayashi H, Saigenji K. Dilatation and constriction of rat gastric mucosal microvessels through prostaglandin EP2 and EP3 receptors. Aliment Pharmacol Ther 1999; 13:1243-50. [PMID: 10468708 DOI: 10.1046/j.1365-2036.1999.00577.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
BACKGROUND Prostaglandin (PG)E2 has both a vasodilating action and a protective function in the gastric mucosa. There are four subtypes of PGE2-sensitive, or EP, receptors. AIM To identify the subtype of EP receptors in the microvessels of the rat gastric mucosa using EP2 and EP3 receptor agonists. METHODS The posterior wall of the anaesthetized rat stomach was secured in a chamber and superfused with Tyrode's solution, and the gastric microcirculation of the mucosal base was observed through a window with transillumination. PGE2 and its derivatives (20 microL) were applied topically in the window. RESULTS PGE2 (0.001-10 micromol/L), misoprostol (EP2/EP3 receptor agonist; 0.01-100 micromol/L) and butaprost (EP2 receptor agonist; 1-1000 micromol/L) dilated the arterioles dose-dependently, but M&B 28 767 (EP3 receptor agonist; 0.001-10 micromol/L) did not alter their diameters. M&B 28 767 constricted the venules and collecting venules dose-dependently whereas butaprost dilated them. PGE2 and misoprostol had bell-shaped dose-response curves: constriction by low doses of PGE2 and misoprostol (0.001-0.1 micromol/L and 0.01-1 micromol/L) and dilation by high doses of PGE2 and misoprostol (0.1-100 micromol/L and 1-100 micromol/L). CONCLUSIONS These results suggest that PGE2 dilated both arterioles and venules in the rat gastric mucosa through the EP2 receptors and constricted the venules through the EP3 receptors.
Collapse
Affiliation(s)
- T Ohno
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Maekawa T, Waki S, Okada A, Fukui H, Kinoshita Y, Chiba T. Midkine gene expression in the healing process of gastric ulcer. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 1999; 133:349-52. [PMID: 10218765 DOI: 10.1016/s0022-2143(99)90065-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Midkine is a newly cloned growth-promoting factor for fibroblastic cells. This study was performed to investigate the possible role of midkine in the stomach. Rats with acute or chronic gastric mucosal lesions were used. Histologically, acute mucosal lesions are not accompanied by the formation of granulation tissue; conversely, chronic mucosal lesions are accompanied by the formation of granulation tissue. The expression of the midkine gene was found in the normal intact gastrointestinal tract, especially the submucosal and muscle layers. Midkine mRNA increased during the healing stage of chronic gastric ulcer accompanied by a fibroblastic reaction. Furthermore, the fibroblast cell line MRC-5 expressed midkine mRNA strongly. Therefore, midkine may have some role in the healing of gastric deep ulcers that is accompanied by fibroblast proliferation.
Collapse
Affiliation(s)
- T Maekawa
- Department of Internal Medicine, Kanebo Memorial Hospital, Kobe, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Ishihara S, Hassan S, Kinoshita Y, Moriyama N, Fukuda R, Maekawa T, Okada A, Chiba T. Growth inhibitory effects of somatostatin on human leukemia cell lines mediated by somatostatin receptor subtype 1. Peptides 1999; 20:313-8. [PMID: 10447088 DOI: 10.1016/s0196-9781(99)00037-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Reverse transcription polymerase chain reaction analysis revealed that only somatostatin receptor (SSTR) 1 mRNA was expressed in Ball-1 B-, Jurkat T-, and HL60 leukemia cell lines. In contrast, human normal mononuclear cells expressed the mRNA of all five subtypes of SSTR, although the expression level of SSTR1 was the highest. A binding study, revealed that [125I]-somatostatin bound specifically to HL60 cells and this binding was inhibited concentration-dependently by unlabeled somatostatin (SS). A [3H]thymidine incorporation study showed that SS significantly and concentration-dependently inhibited HL60 and BALL-1 leukemia cell growth. Furthermore, this inhibition of leukemia cell growth was associated with reduces c-fos gene expression. These data indicate that leukemia cells express SSTR1 and SS reduce c-fos gene expression with resultant suppression of leukemia cell growth, possibly mediated by the SSTRI.
Collapse
Affiliation(s)
- S Ishihara
- 2nd Division of Internal Medicine, Shimane Medical University, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Takahashi S, Shigeta J, Ishikawa M, Kobayashi N, Okabe S. Role of thromboxane A2 in healing of gastric ulcers in rats. JAPANESE JOURNAL OF PHARMACOLOGY 1999; 79:101-7. [PMID: 10082323 DOI: 10.1254/jjp.79.101] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We investigated the role of thromboxane (TX) A2 in gastric ulcer healing in rats. Acetic acid ulcers were produced in male Donryu rats. TXA2 synthesis in the stomachs with ulcers was significantly elevated in ulcerated tissue, but not in intact tissue, compared with that in the gastric mucosa of normal rats. Indomethacin inhibited both TXA2 and prostaglandin E2 (PGE2) synthesis in ulcerated tissue, while NS-398 (selective cyclooxygenase-2 inhibitor) reduced only PGE2 synthesis. OKY-046 (TXA2 synthase inhibitor) dose-relatedly inhibited only TXA2 synthesis. The maximal effect of OKY-046 (80% inhibition) was found at more than 30 mg/kg. When OKY-046 was administered for 14 days, the drug at more than 30 mg/kg significantly accelerated ulcer healing without affecting acid secretion. The maximal reduction of ulcerated area by OKY-046 was about 30%, compared with the area in the control. Histological studies revealed that regeneration of the mucosa was significantly promoted by OKY-046, but neither maturation of the ulcer base nor angiogenesis in the base were affected. OKY-046 and TXB2 had no effect on proliferation of cultured rat gastric epithelial cells, but U-46619 (TXA2 mimetic) dose-relatedly prevented the proliferation without reducing cell viability. These results indicate that the increased TXA2, probably derived from cyclooxygenase-1 in ulcerated tissue, exerts a weak inhibitory effect on ulcer healing in rats. The effect of TXA2 might be due partly to prevention of gastric epithelial cell proliferation at the ulcer margin.
Collapse
Affiliation(s)
- S Takahashi
- Department of Applied Pharmacology, Kyoto Pharmaceutical University, Yamashina, Japan
| | | | | | | | | |
Collapse
|
42
|
Takeuchi K, Yagi K, Kitamura M, Kubomi M, Tashima KI. Prostaglandin E-type receptor subtypes and gastroduodenal bicarbonate secretion in rats. J Gastroenterol Hepatol 1998; 13:S221-S226. [PMID: 28976658 DOI: 10.1111/j.1440-1746.1998.tb01881.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
We investigated the relationship between prostaglandin E-type receptor (EP receptor) subtypes and gastroduodenal HCO3- secretion in rats. Under urethane anaesthesia, a stomach mounted in an ex vivo chamber or a proximal duodenal loop was perfused with saline and the HCO3- secretion was measured at pH 7.0 using a pH-stat method and by adding 10 mmol/L HCl. Prostaglandin E2 (PGE2 , i.V.) increased HCO3- secretion in both the stomach and duodenum; this action was verapamil sensitive and only in the duodenum was potentiated by isobutylmethyl xanthine (IBMX). Duodenal HCO3- secretion was also stimulated by both sulprostone (EP1 /EP3 agonist), enprostil (EP1 /EP3 agonist), misoprostol (EP2 /EP3 agonist), 11-deoxy PGE1 (EP3 /EP4 agonist) and ONO-NT-012 (EP3 agonist), but was not affected by either butaprost (EP2 agonist) or 17-phenyl-ω-trinor-PGE2 (EP1 agonist). In contrast, gastric HCO3- secretion was stimulated by sulprostone, enprostil and 17-phenyl-ω-trinor-PGE2 , but not by misoprostol, butaprost, 11-deoxy PGE1 or ONO-NT-012. The EP1 antagonist SC-51089 inhibited the HCO3- stimulatory action of sulprostone in the stomach but not in the duodenum. Isobutylmethyl xanthine potentiated the HCO3- response to sulprostone in the duodenum, while verapamil reduced the response in both the stomach and duodenum. These results suggest that PGE2 stimulates HCO3- secretion via different EP receptor subtypes in the stomach and duodenum: in the former the EP1 receptors linked to Ca2+ and in the latter, the EP3 receptors coupled with both cAMP and Ca2+ .
Collapse
Affiliation(s)
- Koji Takeuchi
- Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto, Japan
| | - Koji Yagi
- Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto, Japan
| | - Motohiro Kitamura
- Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto, Japan
| | - Masafumi Kubomi
- Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto, Japan
| | - Kim-Ihito Tashima
- Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto, Japan
| |
Collapse
|
43
|
Okada A, Kinoshita Y, Waki S, Fukui H, Maekawa T, Matsushima Y, Kawanami C, Kishi K, Nakata H, Wang HY, Hassan S, Chiba T. Rat gastric mucosal cells express ICAM-1 and proinflammatory cytokines during indomethacin-induced mucosal injury. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 1998; 131:538-47. [PMID: 9626989 DOI: 10.1016/s0022-2143(98)90062-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adhesion molecules and cytokines are known to be involved in the formation of acute gastric mucosal injury. However, it is not clear whether the gastric mucosal cells express these molecules and modulate the inflammation. To clarify whether gastric mucosal cells express intercellular adhesion molecule-1 (ICAM-1) and proinflammatory cytokines (tumor necrosis factor-alpha (TNF-alpha), interleukin-1-alpha (IL-1-alpha), and cytokine-induced neutrophil chemoattractant-2-beta (CINC-2-beta)) in the formation of gastric mucosal injury, we have used rat indomethacin-induced gastric mucosal lesions as an in vivo model. The gene expression of all cytokines and ICAM-1 increases at the early stages of indomethacin-induced gastritis (TNF-alpha and IL-1-alpha gene expression began to increase earlier than that of ICAM-1 and CINC-2-beta) and can mainly be detected in the gastric epithelial layer. To further identify the source of those molecules, the epithelial cells were separated into seven fractions according to their sizes by a counterflow elution. ICAM-1 and CINC-2-beta gene expressions are particularly enhanced in the middle-sized cell fractions that are rich in gastric mucous-producing cells. The effect of TNF-alpha or IL-1-alpha on the gene expression of ICAM-1 and cytokines was examined by using RGM-1 cells as a model for gastric mucosal cells. RGM-1 cells show an augmented ICAM-1 and proinflammatory cytokine expression in response to TNF-alpha or IL-1-alpha stimulation. Moreover, immunohistochemical staining also reveals an increase in ICAM-1 and CINC protein production in RGM-1 cells in response to TNF-alpha stimulation. We conclude that gastric mucosal cells express various cytokines and an adhesion molecule during the formation of acute gastric mucosal injury and that they may modulate the inflammation.
Collapse
Affiliation(s)
- A Okada
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Nakamura E, Takahashi S, Matsui H, Okabe S. Interleukin-1beta inhibits growth factor-stimulated restoration of wounded rat gastric epithelial cell monolayers. Dig Dis Sci 1998; 43:476-84. [PMID: 9539640 DOI: 10.1023/a:1018890419648] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We examined the effect of interleukin-1beta (IL-1beta) on spontaneous and enhanced restoration (cell migration and proliferation) using an in vitro wound model comprising a confluent monolayer of rat gastric epithelial RGM1 cells. Repair of an artificial wound in a cell monolayer was found to be time- and concentration-dependent when the cells were incubated with epidermal growth factor (EGF) or transforming growth factor (TGF) -alpha alone for up to 24 hr. The growth factors also stimulated DNA synthesis significantly for 24 hr in a concentration-related manner. IL-1beta had no effect on wound restoration in the absence of the growth factors. However, it markedly inhibited the restoration enhanced by EGF and TGF-alpha, the inhibition being about 60% and 70%, respectively. In addition, IL-1beta significantly reduced the DNA synthesis stimulated by the growth factors. The EGF- and TGF-alpha-enhanced restoration was reduced by about 30% by mitomycin C, which potently inhibited the stimulated DNA synthesis. Mitomycin C had no effect on the spontaneous restoration. Even when treated with mitomycin C, the inhibitory effect of IL-1beta on the enhanced wound repair was still observed; however, the extent of the inhibition was decreased. These results indicate that IL-1beta inhibits the migration as well as the proliferation of gastric epithelial cells enhanced by EGF and TGF-alpha, resulting in a failure of wound healing.
Collapse
Affiliation(s)
- E Nakamura
- Department of Applied Pharmacology, Kyoto Pharmaceutical University, Yamashina, Japan
| | | | | | | |
Collapse
|
45
|
Naito Y, Yoshikawa T, Fujii T, Boku Y, Yagi N, Dao S, Yoshida N, Kondo M, Matsui H, Ohtani-Fujita N, Sakai T. Monochloramine-induced cell growth inhibition and apoptosis in a rat gastric mucosal cell line. J Clin Gastroenterol 1998; 25 Suppl 1:S179-85. [PMID: 9479646 DOI: 10.1097/00004836-199700001-00029] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent studies have indicated that monochloramine (NH2Cl), a reaction product of NH3 and hypochlorous acid, is involved in the pathogenesis of Helicobacter pylori-associated gastric mucosal damage, but how NH2Cl contributes to lesions is unclear. In the present study, the effects of NH2Cl on mucosal cell growth and the cell cycle were evaluated in vitro using a normal rat gastric mucosal cell line RGM-1. Cell viability was assessed by the Trypan Blue dye exclusion test and cell cycle patterns were determined by DNA labeling with propidium iodide and flow cytometric quantification. NH2Cl inhibited the growth of RGM-1 cells in a concentration-dependent manner. Exposure of cells to NH2Cl caused a time- and dose-dependent loss of G1-phase cells with accumulation of G2/M-phase cells, and produced a fraction of subdiploid cells with oligonucleosomal DNA degradation characteristic of apoptosis. NH2Cl-induced apoptosis was confirmed by fluorescent microscopy with Hoechst 33342 and propidium iodide. These results suggest that NH2Cl inhibits gastric mucosal cell growth and induces apoptosis in RGM-1 cells, events that may be important in gastric mucosal damage or atrophy induced by H. pylori infection.
Collapse
Affiliation(s)
- Y Naito
- First Department of Medicine, Kyoto Prefectural University of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Waki S, Kinoshita Y, Fukui H, Maekawa T, Okada A, Matsushima Y, Hassan S, Sakatani T, Kawanami C, Kishi K, Chiba T. Intragastric distribution of nonsteroidal anti-inflammatory drug-related ulcers in patients without collagen diseases. J Clin Gastroenterol 1997; 25:592-4. [PMID: 9451669 DOI: 10.1097/00004836-199712000-00008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
During the long-term administration of nonsteroidal anti-inflammatory drugs (NSAIDs), approximately 3% of patients have gastric ulcers develop in each year. Although much is known about the endoscopic characteristics of NSAID-induced gastric ulcers in patients with rheumatoid arthritis (RA), it is not clear where in the stomach NSAIDs induce ulcers in patients without RA. We looked at that question. During the 1-year study period, 29 patients with gastric ulcer, who had been taking NSAIDs regularly for more than 4 weeks mainly for osteoarthritis, were identified. Seventy-five patients with gastric ulcers who had not taken NSAID also were found. The sites of gastric ulcers of these two groups were quite different. The NSAID-induced ulcers mainly were found in the gastric antrum, whereas the majority of NSAID-unrelated ulcers were in the gastric corpus. We conclude that NSAID-induced ulcers in non-RA patients mainly are formed in the gastric antrum.
Collapse
Affiliation(s)
- S Waki
- Division of Gerontology, Kobe University School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kishi K, Kinoshita Y, Matsushima Y, Okada A, Maekawa T, Kawanami C, Watanabe N, Chiba T. Pepsinogen C gene product is a possible growth factor during gastric mucosal healing. Biochem Biophys Res Commun 1997; 238:17-20. [PMID: 9299443 DOI: 10.1006/bbrc.1997.7231] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We isolated, by the subtraction cloning method, a pepsinogen C (PGC) gene fragment (the sequence between the 968th and 1179th base pairs) from a rat gastric mucosal cDNA library as a cDNA clone encoding a substance that promotes growth of the normal rat gastric mucosal cell line RGM1. Northern blot analysis revealed that PGC gene expression was enhanced not only in acetic acid-induced chronic gastric ulcers but also in indomethacin-induced gastric mucosal lesions. PGC gene expression was also increased in the Helicobacter felis-infected stomachs. Thus, the PGC gene may play a role in gastric epithelial cell growth during gastric mucosal healing.
Collapse
Affiliation(s)
- K Kishi
- Department of Medicine, Kyoto University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Ding M, Kinoshita Y, Kishi K, Nakata H, Hassan S, Kawanami C, Sugimoto Y, Katsuyama M, Negishi M, Narumiya S, Ichikawa A, Chiba T. Distribution of prostaglandin E receptors in the rat gastrointestinal tract. PROSTAGLANDINS 1997; 53:199-216. [PMID: 9206801 DOI: 10.1016/s0090-6980(97)00015-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
AIMS In order to study the role of prostaglandin in the regulation of the gastrointestinal functions, gene expression of prostaglandin receptors along the rat gastrointestinal tracts were investigated. METHODS Rats were used for the study. The combination of counterflow elutriation separation of mucosal cells and Northern blot analysis was used to detect the gene expression of prostaglandin receptors in gastrointestinal tracts. RESULTS In small intestine and colon, prostaglandin E2 EP1 and EP3 receptor mRNAs were mainly localized in the deeper intestinal wall containing muscle layers. EP4 receptor gene expression, on the other hand, was detected in the intestinal mucosal layer. In the stomach, EP1 mRNA was detected in gastric muscle layers, whereas EP3 and EP4 receptor gene expression was mainly present in the gastric mucosal layer containing epithelial cells. In gastric epithelial cells, parietal cells were found to have both EP3 and EP4 receptors. At lower concentrations, prostaglandin E2 inhibited gastric acid secretion by parietal cells probably through EP4 receptors. At higher concentrations, however, it stimulated it. On the other hand, mucous cells possessed only EP4 receptor mRNA. CONCLUSIONS Thus, it is suggested that prostaglandin E2 modulates gastrointestinal functions through at least three different prostaglandin receptors (EP1, EP3, and EP4), each of which has a distinct contribution in the gastrointestinal tract.
Collapse
MESH Headings
- Aminopyrine/metabolism
- Aminopyrine/pharmacokinetics
- Animals
- Blotting, Northern
- Carbon Radioisotopes
- Colon/chemistry
- Colon/metabolism
- Cyclic AMP/metabolism
- DNA, Complementary/metabolism
- Digestive System/chemistry
- Digestive System/metabolism
- Dinoprostone/pharmacology
- Enprostil/pharmacology
- Gastric Mucosa/metabolism
- Intestinal Mucosa/chemistry
- Intestinal Mucosa/metabolism
- Intestines/chemistry
- Male
- Parietal Cells, Gastric/drug effects
- Parietal Cells, Gastric/metabolism
- RNA/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Prostaglandin E/chemistry
- Receptors, Prostaglandin E/genetics
- Receptors, Prostaglandin E/metabolism
- Receptors, Prostaglandin E, EP1 Subtype
- Receptors, Prostaglandin E, EP3 Subtype
- Receptors, Prostaglandin E, EP4 Subtype
- Stomach/chemistry
Collapse
Affiliation(s)
- M Ding
- Department of Medicine, Kobe University School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Furukawa O, Nakamura E, Okabe S. Characterization of a novel cell damage model induced by acid and pepsin using rat gastric epithelial cells: protective effect of sucralfate. J Gastroenterol Hepatol 1997; 12:115-21. [PMID: 9083912 DOI: 10.1111/j.1440-1746.1997.tb00394.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We have established a new model for rat gastric epithelial cell (RGM1) damage caused by both acid and pepsin. Exposure of RGM1 to an acidified medium (pH 3.5-5.0) for 10-50 min decreased cell viability in a time- and pH-dependent manner. Pepsin (0.5-1.0 mg/mL) at pH 4.5 potentiated cell damage in a concentration-dependent manner. Based on these results, two types of cell damage models caused by incubation of cells at pH 4.0 and with pepsin (0.75 mg/mL) at pH 4.5 for 30 min, respectively, were established. The intracellular pH (pHi) gradually decreased with a decrease in medium pH and an increase in exposure time. At pH < or = 4.0, pHi reached approximately pH 6.3. Pepsin at pH 4.5 caused a further reduction in pHi compared with the acidified medium alone. Pepsin pre-incubated with pepstatin did not induce any cell damage. Pretreatment with sucralfate (0.1-3 mg/ mL) for 2 h significantly prevented cell damage caused at both pH 4.0 and with pepsin at pH 4.5 in a concentration-dependent manner. Sucralfate (3 mg/mL) significantly prevented the reduction in pHi at pH 4.0 or with pepsin at pH 4.5. 16,16-Dimethyl prostaglandin E2 (30 micrograms/mL) had no effect on either cell damage or pHi. These cell damage models involving RGM1 are useful for studying the mechanism underlying cell damage and for the screening of cytoprotective drugs.
Collapse
Affiliation(s)
- O Furukawa
- Department of Applied Pharmacology, Kyoto Pharmaceutical University, Japan
| | | | | |
Collapse
|