ArticlePDF Available

Roles of Cyclooxygenase-2 and Prostacyclin/IP Receptors in Mucosal Defense against Ischemia/Reperfusion Injury in Mouse Stomach

Authors:

Abstract and Figures

We examined the roles of cyclooxygenase (COX) isozymes, prostaglandins (PGs), and their receptors in the mucosal defense against ischemia/reperfusion (I/R)-induced gastric lesions in mice. Male C57BL/6 mice, including wild-type animals and those lacking prostaglandin E(2) (EP)1, EP3, or prostaglandin I(2) (IP) receptors, were used after 18 h of fasting. Under urethane anesthesia, the celiac artery was clamped (ischemia) for 30 min, and then reperfusion was achieved for 60 min through the removal of the clamp, and the stomach was examined for lesions. I/R produced hemorrhagic gastric lesions in wild-type mice. The severity of lesions was significantly increased by pretreatment with indomethacin (a nonselective COX inhibitor) and rofecoxib (a selective COX-2 inhibitor) but not 5-(4-chlorophenyl)-1-(4-methoxyphenyl)-3-(trifluoromethyl)-1H-pyrazole (SC-560; a selective COX-1 inhibitor). The expression of COX-2 mRNA was up-regulated in the stomach following I/R but not by sham operation or ischemia alone. The ulcerogenic response was markedly aggravated in IP receptor knockout mice but not those lacking EP1 or EP3 receptors. I/R increased the levels of 6-keto-PGF(1alpha) and PGE(2) in the stomach of wild-type mice, and this response was attenuated by indomethacin and rofecoxib but not SC-560. Pretreatment of wild-type mice with iloprost, a prostacyclin (PGI(2)) analog, significantly prevented the I/R-induced gastric lesions in the absence and presence of indomethacin or rofecoxib. PGE(2) also reduced the severity of I/R-induced gastric lesions, yet the effect was much less pronounced than that of iloprost. These results suggest that endogenous PGs derived from COX-2 play a crucial role in gastric mucosal defense during I/R, and this action is mainly mediated by PGI(2) through the activation of IP receptors.
Content may be subject to copyright.
Roles of Cyclooxygenase-2 and Prostacyclin/IP Receptors in
Mucosal Defense against Ischemia/Reperfusion Injury in
Mouse Stomach
Tohru Kotani, Atsushi Kobata, Eiji Nakamura, Kikuko Amagase, and Koji Takeuchi
Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto,
Japan
Received July 24, 2005; accepted October 17, 2005
ABSTRACT
We examined the roles of cyclooxygenase (COX) isozymes,
prostaglandins (PGs), and their receptors in the mucosal de-
fense against ischemia/reperfusion (I/R)-induced gastric lesions
in mice. Male C57BL/6 mice, including wild-type animals and
those lacking prostaglandin E
2
(EP)1, EP3, or prostaglandin I
2
(IP) receptors, were used after 18 h of fasting. Under urethane
anesthesia, the celiac artery was clamped (ischemia) for 30 min,
and then reperfusion was achieved for 60 min through the
removal of the clamp, and the stomach was examined for
lesions. I/R produced hemorrhagic gastric lesions in wild-type
mice. The severity of lesions was significantly increased by
pretreatment with indomethacin (a nonselective COX inhibitor)
and rofecoxib (a selective COX-2 inhibitor) but not 5-(4-
chlorophenyl)-1-(4-methoxyphenyl)-3-(trifluoromethyl)-1H-
pyrazole (SC-560; a selective COX-1 inhibitor). The expression
of COX-2 mRNA was up-regulated in the stomach following I/R
but not by sham operation or ischemia alone. The ulcerogenic
response was markedly aggravated in IP receptor knockout
mice but not those lacking EP1 or EP3 receptors. I/R increased
the levels of 6-keto-PGF
1
and PGE
2
in the stomach of wild-
type mice, and this response was attenuated by indomethacin
and rofecoxib but not SC-560. Pretreatment of wild-type mice
with iloprost, a prostacyclin (PGI
2
) analog, significantly pre
-
vented the I/R-induced gastric lesions in the absence and
presence of indomethacin or rofecoxib. PGE
2
also reduced the
severity of I/R-induced gastric lesions, yet the effect was much
less pronounced than that of iloprost. These results suggest
that endogenous PGs derived from COX-2 play a crucial role in
gastric mucosal defense during I/R, and this action is mainly
mediated by PGI
2
through the activation of IP receptors.
The damage caused by an interruption of blood supply to
an organ or tissue followed by the reintroduction of blood into
the affected area is called ischemia/reperfusion (I/R) injury.
The phenomenon of I/R injury is a major clinical problem
after stroke, infarction, shock, and organ transplantation.
The depletion of adenosine triphosphate (ATP) and distur-
bance of intracellular calcium homeostasis have been sug-
gested as the major pathophysiological mechanisms during
ischemia, leading to loss of cell viability (Farber et al., 1981;
Cheung et al., 1986). Reperfusion of ischemic tissues para-
doxically exacerbates the injury process and leads to the
release of reactive oxygen species and proinflammatory me-
diators and the attraction of inflammatory cells infiltrating
the tissues (Chamoun et al., 2000; Piper et al., 2003). In the
gastrointestinal tract, I/R injuries are known to be associated
with significant morbidity and mortality during the course of
hemorrhagic shock, abdominal aortic aneurysm repair, isch-
emic bowel disease, and necrotizing enterocolitis (Yasue et
al., 1992; Riaz et al., 2002; Dimmitt et al., 2003).
Nonselective cyclooxygenase (COX) inhibitors damage the
gastrointestinal mucosa in patients as an adverse reaction
(Soll et al., 1991). By contrast, selective COX-2 inhibitors
such as rofecoxib and celecoxib do not induce gastric lesions
in rats (Vane and Botting, 1995; Laudanno et al., 2001).
Hence, these COX-2 inhibitors were expected to be anti-
inflammatory and chemopreventive drugs devoid of gastro-
intestinal toxicity, although chronic treatment with a COX-2
inhibitor delayed the healing of gastric ulcers (Mizuno et al.,
1997; Miura et al., 2004). Recently, it was reported that a
selective COX-2 inhibitor aggravated gastric lesions induced
This research was supported in part by the Kyoto Pharmaceutical Univer-
sity’s “21st Century COE” program and the “Open Research” Program from the
Ministry of Education, Science and Culture of Japan.
Article, publication date, and citation information can be found at
http://jpet.aspetjournals.org.
doi:10.1124/jpet.105.093195.
ABBREVIATIONS: I/R, ischemia/reperfusion; COX, cyclooxygenase; PG, prostaglandin; EP, prostaglandin E
2
receptor; PGI
2
, prostacyclin; IP,
prostaglandin I
2
receptor; MPO, myeloperoxidase; PCR, polymerase chain reaction; SC-560, 5-(4-chlorophenyl)-1-(4-methoxyphenyl)-3-(triflu
-
oromethyl)-1H-pyrazole; NS-398, N-[2-(cyclohexyloxy)-4-nitrophenyl]methanesulfonamide; DFU, 5,5-dimethyl-3-(3-fluorophenyl)-4-(4-methyl sul-
fonyl)phenyl-2(5II)-furanone.
0022-3565/06/3162-547–555$20.00
T
HE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS Vol. 316, No. 2
Copyright © 2006 by The American Society for Pharmacology and Experimental Therapeutics 93195/3073009
JPET 316:547–555, 2006 Printed in U.S.A.
547
at ASPET Journals on December 31, 2015jpet.aspetjournals.orgDownloaded from
by I/R in rats, suggesting the involvement of endogenous
prostaglandins (PGs) in the mucosal defense during I/R
(Maricic et al., 1999).
PGs, especially PGE
2
, have been shown to exert a protec
-
tive action in the stomach through the activation of EP1
receptors, although the effects on various functions are me-
diated by different EP receptor subtypes; cf., acid inhibition
by EP3 receptors, an increase of mucus secretion by EP4
receptors, an increase of mucosal blood flow by EP2/EP4
receptors, and an inhibition of gastric motility by EP1 recep-
tors (Takeuchi et al., 2002b). Likewise, prostacyclin (PGI
2
), a
prostanoid mainly synthesized in the endothelium, exerts
various physiological actions at the interface between blood
and tissue (Konturek and Robert, 1982; Whittle et al., 1984).
Since PGI
2
enhances the gastric mucosal microcirculation
through vasodilation and inhibition of platelet aggregation, it
is possible that this prostanoid contributes to the mainte-
nance of the mucosal integrity of the stomach during I/R
(Granger and Kubes, 1994; Saika et al., 1999). However, it
remains unknown which type of prostanoid plays a role in
mucosal defense of the stomach under I/R conditions. In the
present study, we examined the effects of various COX in-
hibitors on I/R-induced gastric lesions in mice and further
investigated which type of prostanoid receptor is involved in
mucosal defense under I/R-induced conditions using animals
lacking EP1, EP3, or IP receptors.
Materials and Methods
Animals. Male C57BL/6 mice (3 months old; SLC, Shizuoka,
Japan) were used. Mice lacking EP1, EP3, or IP receptors were
generated as described previously (Sugimoto et al., 1992; Ushikubi et
al., 1998). In brief, the genes encoding the EP1, EP3, and IP recep-
tors were individually disrupted, and chimeric mice were generated.
These animals were then backcrossed with C57BL/6 mice, and the
resulting heterozygous littermates [EP1 (/), EP3 (/), or IP
(/)] were bred to produce homozygous EP1 (/), EP3 (/), or IP
(/) mice. Homozygous mice were born at the predicted Mendelian
frequency, grew normally, lived longer than 1 year, and were fertile.
Distribution of the EP1, EP3, and IP receptor genes was verified by
Northern blot hybridization, which failed to detect messenger RNAs
encoding the respective receptors in EP1 (/), EP3 (/), and IP
(/) mice. These knockout mice were deprived of food but allowed
free access to tap water for 18 h before the experiments. All experi-
ments were carried out using four to eight mice under urethane
anesthesia unless otherwise specified. The experimental procedures
described here were approved by the Experimental Animal Research
Committee of Kyoto Pharmaceutical University.
Induction of Gastric Mucosal Lesion by Ischemia and
Reperfusion. Acute gastric mucosal lesions were produced by I/R
(Wada et al., 1996). Briefly, under urethane anesthesia (1.25 g/kg
i.p.), the celiac artery was clamped with a small clamp (disposable
vascular clip, holding force of 40 g; BEAR Medical Corporation,
Chiba, Japan), and 30 min later, reperfusion was achieved through
removal of the clamp. After reperfusion for 60 min, the stomach was
excised, inflated by injecting 0.4 ml of 2% formalin for 10 min to fix
the tissue walls, and opened along the greater curvature. In wild-
type mice, the effects of COX inhibitors on the I/R-induced gastric
lesions were examined. Indomethacin (5 mg/kg), rofecoxib (a selec-
tive COX-2 inhibitor, 5 mg/kg), or SC-560 (a selective COX-1 inhib-
itor, 5 mg/kg) was administered p.o. 60 min before ischemia. The
doses of these COX inhibitors were selected to show nonselective
inhibition of both COX-1 and COX-2 or selective inhibition of COX-1
or COX-2, respectively (Takeeda et al., 2003). In addition, the effects
of iloprost, an analog of PGI
2
, and PGE
2
on the I/R-induced gastric
lesions were examined also in both wild-type and IP receptor knock-
out mice. Iloprost (0.33
g/kg) or PGE
2
(0.11 mg/kg) was given i.v.
5 min before reperfusion. The area (millimeters
2
10). The person
measuring the lesions did not know the treatments given to the
animals.
In some cases, the gastric mucosa was examined with a light
microscope following I/R. The animals were killed after I/R treat-
ment, and the stomachs were excised. The tissue samples were then
immersed in 2% formalin-saline, embedded in paraffin, sectioned at
4
m, and stained with hematoxylin and eosin (H&E).
Determination of Myeloperoxidase Activity. Myeloperoxi-
dase (MPO) activity in the gastric mucosa was measured after I/R
treatment in wild-type and IP receptor knockout mice, according to a
modified version of the method of Krawisz et al. (1984). After 60 min
after I/R treatment, the animals were sacrificed by withdrawal of
blood from the heart by perfusing with saline, and the stomach was
excised. After rinsing of the tissue with ice-cold saline, the mucosa
was scraped with glass slides, weighed, and homogenized in a 50 mM
phosphate buffer containing 0.5% hexadecyltrimethylammonium
bromide (pH 6.0; Sigma-Aldrich, St. Louis, MO). The homogenized
samples were subjected to freezing and thawing three times and
centrifuged at 2000 rpm for 10 min at 4°C. MPO activity in the
supernatant was determined by adding 100
l of the supernatant to
1.9 ml of 10 mM phosphate buffer (pH 6.0) and 1 ml of 1.5 M
o-dianisidine hydrochloride (Sigma-Aldrich) containing 0.0005%
(w/v) hydrogen peroxide. The changes in absorbance at 450 nm of
each sample were recorded on a Hitachi spectrophotometer (U-2000;
Hitachi, Ibaraki, Japan). Sample protein content was estimated by
spectrophotometric assay (Pierce protein assay kit; Rockford, IL),
and the MPO activity was obtained from the slope of the reaction
curve, based on the following equation: specific activity (
mol H
2
O
2
/
min/mg of protein) (OD/min)/OD/
mol H
2
O
2
mg of protein).
Measurement of Mucosal PGE
2
and 6-Keto-PGF
1
Levels.
Levels of PGE
2
and 6-keto-PGF
1
, the stable metabolite of PGI
2
,in
the gastric mucosa were measured after I/R treatment in wild-type
mice. The animals were killed under deep ether anesthesia after the
60-min reperfusion period, and the gastric mucosa was isolated,
weighed, and placed in a tube containing 100% methanol plus 0.1
mM indomethacin (Futaki et al., 1994). Then the tissues were ho-
mogenized by a Polytron homogenizer (IKA, Tokyo, Japan) and cen-
trifuged at 12,000 rpm for 10 min at 4°C. After the supernatant of
each sample had been evaporated with N
2
gas, the residue was
resolved in assay buffer and used for determination of PGE
2
and
6-keto-PGF
1
. The concentrations of PGE
2
and 6-keto-PGF
1
were
measured using a PGE
2
or 6-keto-PGF
1
enzyme immunoassay kit
(GE Healthcare, Little Chalfont, Buckinghamshire, UK).
Analysis of COX-1, -2, EP1, EP3, and IP mRNA Expression
by Reverse Transcription-PCR. Wild-type animals were killed
under deep ether anesthesia after I/R treatment, and the stomachs
were removed, frozen in liquid nitrogen, and stored at 80°C prior to
use. Tissue samples were pooled from two to three rats for extraction
of total RNA, which was prepared by a single-step acid phenol-
chloroform extraction procedure by use of TRIzol (Invitrogen, Carls-
bad, CA). Total RNA primed by random hexadeoxy ribonucleotide
was reverse-transcribed with the SuperScript preamplification sys-
tem (Invitrogen). The sequences of sense and antisense primers for
the mouse COX-1, COX-2, EP1, EP3, and IP are shown in Table 1. An
aliquot of the reverse transcription reaction product served as a
template in 35 cycles of PCR with 1 min of denaturation at 94°C, 0.5
min of annealing at 56°C, and 1 min of extension at 72°C on a
thermal cycler. A portion of the PCR mixture was electrophoresed in
1.5% agarose gel in Tris-EDTA-acetic acid buffer, and the gel was
stained with ethidium bromide and photographed.
Determination of Gastric Secretion. Acid secretion was mea-
sured in wild-type mice provided with an acute gastric fistula under
urethane anesthesia (1.25 g/kg i.p.). Briefly, the abdomen was in-
cised, and both the stomach and duodenum were exposed. An acute
fistula (inside diameter, 2 mm) made with a polyethylene tube was
548 Kotani et al.
at ASPET Journals on December 31, 2015jpet.aspetjournals.orgDownloaded from
inserted into the stomach from a small incision made in the fore-
stomach and was held in place by a ligature. The stomach was filled
with 0.4 ml of saline (154 mM NaCl) through the fistula, and the
solution was changed every 20 min. The collected samples were
centrifuged at 3000 rpm for 15 min and titrated to pH 7.0 against 10
mM NaOH using an autoburette (Hiranuma Comtite-8, Tokyo, Ja-
pan). Indomethacin (5 mg/kg), SC-560 (5 mg/kg), or rofecoxib (5
mg/kg) was given i.d. 1 h before instillation of saline in the stomach
whereas iloprost (0.13
g/kg) was given i.v. 5 min before.
Preparation of Drugs. The drugs used were urethane (Tokyo
Kase, Tokyo, Japan), indomethacin (Sigma-Aldrich), SC-560 (Cay-
man Chemical, Ann Arbor, MI), rofecoxib (synthesized in our labo-
ratory), iloprost (Nacalai Tesque, Kyoto, Japan), and PGE
2
(Funa
-
koshi, Tokyo, Japan). All COX inhibitors were suspended in a
hydroxy propyl cellulose solution (Wako, Osaka, Japan). Iloprost was
dissolved in saline, whereas PGE
2
was first dissolved in absolute
ethanol and diluted with saline to the desired concentrations. Each
agent was prepared immediately before use and administered p.o.,
i.d., i.p., or i.v. in a volume of 0.5 ml/100 g of body weight. Control
animals received saline as the vehicle.
Statistics. Data are presented as the mean S.E.M. for four to
eight mice per group. Statistical analyses were performed using a
one-way analysis of variance and Student’s t test or Dunnett’s mul-
tiple comparison test where appropriate, and values of P 0.05 were
considered significant.
Results
I/R-Induced Gastric Lesions in Wild-Type Mice. Lap-
arotomy without clamping of the gastric artery (sham oper-
ation) did not produce any damage in the gastric mucosa of
wild-type mice. In the animals subjected to I/R treatment
(30-min ischemia followed by reperfusion for 60 min), how-
ever, multiple hemorrhagic lesions were observed in the gas-
tric mucosa, the lesion score being 7.2 4.9 mm
2
(n 5).
Ischemia for 30 min did not induce any macroscopically vis-
ible damage in the mucosa. Histologically, most of the dam-
age induced by I/R was restricted to the surface epithelium,
but some damage occurred deep in the mucosa, extending to
the region of pits and glands (Fig. 1, A–D). In sham-operated
animals, no damage was detected even by histological obser-
vation.
Effect of COX Inhibitors on I/R-Induced Gastric Le-
sions in Wild-Type Mice. Following I/R treatment in wild-
type mice, the gastric mucosa developed multiple hemor-
rhagic lesions, the lesion score being 9.9 3.6 mm
2
.
Pretreatment of the animals with indomethacin (5 mg/kg
p.o.) significantly aggravated these lesions, the lesion score
being 22.3 4.5 mm
2
(Fig. 2
A). The severity of I/R-induced
gastric lesions was also significantly increased by prior ad-
ministration of the selective COX-2 inhibitor rofecoxib (5
Fig. 2. A, effects of COX inhibitors on the I/R-induced gastric lesions in
wild-type mice. Indomethacin (5 mg/kg), SC-560 (5 mg/kg), or rofecoxib (5
mg/kg) was given p.o. 60 min before ischemia. Data are presented as
mean S.E.M. from five to seven mice. , significant difference from
control at p 0.05. B, COX mRNA expression in the gastric mucosa of
wild-type (WT) mice subjected to a sham operation or I/R treatment. I,
ischemia for 30 min.
TABLE 1
PCR primer sequences and product size
Molecule Primer Sequence Product Size
bp
GAPDH 5-AACGACCCCTTCATTGAC-3 191
5-TCCACGACATACTCAGCAC-3
COX-1 5-CTGCATGTGGCTGTGGATGTCATC-3 389
5-GGTCTTGGTGAGGCAGACCAG-3
COX-2 5-GTCTGATGATGTATGCCACAATCTG-3 276
5-GATGCCAGTGATAGAGGGTGTTGAA-3
EP1 5-AATACATCTGTGGTGCTGCCAACA-3 829
5-CCACCATTTCCACATCGTGTGCGT-3
EP3 5-GGTATGCCAGCCACATGAAGAC-3 529
5-CAAGATCTGGTTCAGCGAAGCC-3
IP 5-GGCACGAGAGGATGAAGTTTAC-3 408
5-GTCAGAGGCACAGCAGTCAATGG-3
GAPDH, glyceraldehyde-3-phosphate dehydrogenase; bp, base pair(s).
Fig. 1. Gross appearance and histological observations of the gastric
mucosa in wild-type mice subjected to a sham operation or I/R treatment.
A and C, sham; B and D, I/R. Bar, 100
m
PGI
2
and Ischemia/Reperfusion-Induced Gastric Injury 549
at ASPET Journals on December 31, 2015jpet.aspetjournals.orgDownloaded from
mg/kg p.o.), and the lesion score was almost equivalent to
that observed in indomethacin-pretreated animals. However,
the COX-1-selective inhibitor SC-560 (5 mg/kg p.o.) had no
effect on the development of gastric lesions induced by I/R.
The gene expression of glyceraldehyde-3-phosphate dehy-
drogenase, the housekeeping gene, as well as COX-1 was
clearly detectable in the stomach of control wild-type mice
and was not affected by either ischemia or I/R (Fig. 2B).
Although the expression of COX-2 mRNA was negligible in
the gastric mucosa of control wild-type mice, it was markedly
up-regulated following I/R but not ischemia alone.
I/R-Induced Gastric Lesions in EP1, EP3, and IP Re-
ceptor Knockout Mice. To further investigate which pro-
stanoid receptor is involved in the mucosal defense against
I/R-induced gastric lesions, we compared the gastric ulcero-
genic response to I/R in wild-type mice and the animals
lacking EP1, EP3, or IP receptors. As shown in Fig. 3A, the
expression of EP1, EP3, and IP receptor mRNAs was clearly
detectable in the stomach of wild-type mice.
Following I/R treatment, wild-type mice in each group
developed hemorrhagic lesions in the gastric mucosa, the
lesion score being 7.6 2.8 to 8.0 3.2 mm
2
. Development of
gastric lesions was observed in the animals lacking EP1,
EP3, or IP receptors, although the severity of the lesions
differed in these groups of mice. As shown in Fig. 3B, the
gastric ulcerogenic response to I/R was significantly in-
creased in IP receptor knockout mice, the lesion score reach-
ing roughly 2 times that in wild-type mice. However, the
severity of these lesions in EP1 or EP3 receptor knockout
animals was not significantly different compared with wild-
type mice.
Effect of Iloprost on I/R-Induced Gastric Lesions in
Wild-Type Mice. Since the severity of the I/R-induced gas-
tric lesions was found to increase in IP receptor knockout
animals, we examined the effect of a stable PGI
2
analog,
iloprost, on the ulcerogenic response to I/R in wild-type mice,
in the absence or presence of COX inhibitors.
Pretreatment of the animals with iloprost (0.33
g/kg
i.v.) dose-dependently prevented the development of I/R-in-
duced gastric lesions in wild-type mice, the degree of protec-
tion being 72.4% (Fig. 4A). However, this agent, even at 3
g/kg, had no effect on these lesions in the animals lacking IP
receptors (Fig. 4B). On the other hand, the I/R-induced gas-
Fig. 3. A, expression of mRNA for prostanoid receptors (EP1, EP3, and
IP) in the mouse stomach. M, marker. B, gastric lesions induced by I/R in
wild-type mice and those lacking EP1, EP3, or IP receptors. Under
urethane anesthesia, the celiac artery was clamped, and then reperfusion
was achieved 30 min later by removal of the clamp. After a 60-min
reperfusion period, hemorrhagic lesions were induced in the gastric mu-
cosa. Data are presented as mean S.E.M. from five to seven mice. ,
significant difference from each group of wild-type mice at p 0.05.
Fig. 4. A, effect of iloprost, the IP agonist, on I/R-induced gastric lesions
in wild-type mice. Iloprost (0.33
g/kg) was given i.v. 5 min before
reperfusion. Data are presented as mean S.E.M. from six to eight mice.
, significant difference from control at p 0.05. B, effect of iloprost on
I/R-induced gastric lesions in IP receptor knockout mice. Iloprost (1
g/kg) was given i.v. 5 min before reperfusion. Data are presented as
mean S.E.M. from six to eight mice.
550 Kotani et al.
at ASPET Journals on December 31, 2015jpet.aspetjournals.orgDownloaded from
tric lesions were markedly worsened by prior administration
of either indomethacin (5 mg/kg p.o.) or rofecoxib (5 mg/kg
p.o.), the degree of aggravation being 52.2 or 67.3%, respec-
tively (Fig. 5A). The aggravating effect of these COX inhibi-
tors was significantly abrogated by pretreatment with ilo-
prost (1
g/kg i.v.; Fig. 5B).
Mucosal MPO Activity during I/R in Wild-Type and
IP Receptor Knockout Mice. The severity of I/R-induced
gastric lesions was increased in IP receptor knockout mice
and reduced by supplementation of PGI
2
, respectively. To
confirm that the mucosal inflammatory response during I/R
is also affected by these treatments, we measured MPO ac-
tivity in the gastric mucosa of both wild-type and IP receptor
knockout mice after I/R in the presence or absence of iloprost.
Tissue-associated MPO activity in the gastric mucosa of
sham-operated mice was less than 0.3
mol H
2
O
2
/min/mg of
tissue. Gastric MPO activity in wild-type mice was markedly
increased after I/R, reaching about 4 times over the control
levels, the values being 1.11 0.2
mol H
2
O
2
/min/mg of
tissue (Fig. 6A). The MPO activity was further increased in
IP receptor knockout animals in response to I/R, the values
being 2.56 0.41
mol H
2
O
2
/min/mg of tissue, which is
significantly greater than that observed in wild-type mice.
On the other hand, the increase in gastric MPO activity
following I/R in wild-type mice was dose-dependently sup-
pressed by prior administration of iloprost (0.33
g/kg), and
a significant effect was observed at 1 and 3
g/kg, the inhi-
bition being 44.1 and 44.0%, respectively (Fig. 6B).
Gastric Mucosal 6-Keto-PGF
1
and PGE
2
Contents in
Wild-Type Mice. Levels of 6-keto-PGF
1
, the stable metab
-
olite of PGI
2
, in the gastric mucosa were significantly in
-
creased in wild-type mice following I/R treatment compared
with a sham operation (Fig. 7A). This increase was signifi-
cantly prevented by prior administration of indomethacin (5
mg/kg p.o.) and rofecoxib (5 mg/kg p.o.) but not SC-560 (5
mg/kg p.o.). Likewise, the mucosal PGE
2
content was also
significantly increased by I/R treatment, and the response
was significantly attenuated by both indomethacin and rofe-
coxib but not SC-560 (Fig. 7B).
Effect of Various COX Inhibitors and Iloprost on
Gastric Acid Secretion. The stomach of wild-type mice
secreted acid at a rate of about 3.01 to 3.42
Eq for 2 h.
Fig. 5. Effect of iloprost (1
g/kg) on the I/R-induced gastric lesions in
wild-type mice pretreated with COX inhibitors. Indomethacin (5 mg/kg)
or rofecoxib (5 mg/kg) was given p.o. 60 min before ischemia. Iloprost (1
g/kg) was given i.v. 5 min before reperfusion. Data are presented as the
mean S.E.M. from five to six mice. Significant differences at p 0.05:
, from the corresponding control; #, from treatment with (A) indometh-
acin or (B) rofecoxib.
Fig. 6. A, I/R-induced changes in gastric MPO activity in wild-type and IP
receptor knockout mice. Under urethane anesthesia, the celiac artery was
clamped (ischemia), and then reperfusion was achieved 30 min later by
removal of the clamp. After a 60-min reperfusion period, gastric MPO
activity was measured. Data are presented as the mean S.E.M. from six
mice. Significant difference at p 0.05: , from sham; #, from control. B,
the effect of iloprost on I/R-induced gastric MPO activity in wild-type
mice. Iloprost (0.33
g/kg) was given i.v. 5 min before reperfusion. Data
are presented as mean S.E.M. from six mice. , significant difference
from control at p 0.05.
PGI
2
and Ischemia/Reperfusion-Induced Gastric Injury 551
at ASPET Journals on December 31, 2015jpet.aspetjournals.orgDownloaded from
Neither indomethacin, SC-560, nor rofecoxib had any effect
on basal acid secretion in wild-type mice, the values being
equivalent in all groups (Table 2). Likewise, iloprost (0.33
g/kg) given i.v. did not significantly affect acid output at any
dose, although a slight decrease (12.6%) was observed at the
highest dose, 3
g/kg. In addition, no difference in basal acid
secretion was observed between wild-type and IP receptor
knockout mice, the values being 3.01 0.40
Eq/2 h and
3.51 0.43
Eq/2 h, respectively (Table 3).
Effect of PGE
2
on I/R-Induced Gastric Lesions in
Wild-Type Mice. The severity of I/R-induced gastric lesions
remained unaltered in EP1 or EP3 receptor knockout mice.
However, because the levels of PGE
2
content in the gastric
mucosa were found to increase following I/R with the up-
regulation of COX-2 expression, there is a possibility that
PGE
2
plays some role in mucosal defense of the stomach
during I/R through a different EP receptor subtype. We
therefore examined the effect of PGE
2
on I/R-induced gastric
lesions in both wild-type and IP receptor knockout mice.
PGE
2
, given i.v. at 0.1 to 1 mg/kg 5 min before reperfusion,
dose-dependently reduced the severity of I/R-induced gastric
lesions in wild-type mice, and a significant effect was ob-
tained only at 1 mg/kg, the degree of protection being 39.1%,
which is less pronounced compared with that of iloprost at 3
g/kg (67.3%; Fig. 8A). The protective effect of PGE
2
was
observed even in IP receptor knockout mice, although the
degree of protection was 25.8%, slightly less than that in
wild-type mice (Fig. 8B).
Discussion
Ischemia followed by reperfusion leads to tissue injury
(Farber et al., 1981; Cheung et al., 1986; Piper et al., 2003).
Whereas there is a substantial body of experimental data
characterizing the factors that promote gastric lesions under
I/R-induced conditions, tissue defense reactions that counter-
balance the noxious effects of I/R remain less understood.
The present study clearly demonstrated that endogenous
PGs derived from COX-2 play a crucial role in the mucosal
defense of the stomach under I/R-induced conditions, and
this action is mainly mediated by PGI
2
through activation of
IP receptors.
It has been thought that COX-1 functions as a housekeep-
ing enzyme, catalyzing the formation of PGs that contribute
to the maintenance of the mucosal integrity of the stomach
through the modulation of various functions (Soll et al., 1991;
Vane and Botting, 1995), whereas COX-2, the inducible en-
zyme up-regulated by proinflammatory cytokines and growth
factors, mediates pathological reactions such as inflamma-
tion and tumor growth (Davies et al., 1997; Koga et al., 2004).
Recent studies, however, showed that COX-2 is also involved
in mucosal defense under certain conditions (Muscara et al.,
2000; Tanaka et al., 2002) and plays an important role in the
healing of gastric ulcers (Mizuno et al., 1997). In the present
study, we found that the selective COX-2 inhibitor rofecoxib
significantly aggravated the development of gastric lesions in
response to I/R, similar to indomethacin, confirming the in-
Fig. 7. Effect of COX inhibitors on mucosal levels of 6-keto-PGF
1
, the
stable metabolite of PGI
2
(A), and PGE
2
(B) in the stomach of wild-type
mice subjected to I/R treatment. Indomethacin (5 mg/kg), SC-560 (5
mg/kg), or rofecoxib (5 mg/kg) was given p.o. 60 min before ischemia. Data
are presented as the mean S.E.M. from five mice. Significant difference
at p 0.05: , from sham; #, from control.
TABLE 2
Effect of various COX inhibitors and iloprost on gastric secretion in
wild-type mice
Acid secretion was measured in wild-type mice provided with an acute gastric fistula
under urethane anesthesia. The stomach was filled with 0.4 ml of saline through the
fistula, the solution was changed after 20 min, and the collected samples were used
to measure acid output. Indomethacin (5 mg/kg), SC-560 (5 mg/kg), or rofecoxib (5
mg/kg) was given i.d. 1 h before the instillation of saline in the stomach, whereas
iloprost (0.13
g/kg) was given i.v. 5 min before. Data are presented as the mean
S.E.M. from four to five mice.
Group Dose No. of Mice Total Acid Output
Eq/2 h
Saline 4 3.02 0.36
Indomethacin 5 mg/kg 5 2.67 0.39
SC-560 5 mg/kg 5 2.86 0.29
Rofecoxib 5 mg/kg 5 3.21 0.17
Iloprost 0.3
g/kg 5 3.01 0.40
1
g/kg 5 3.18 0.10
3
g/kg 5 2.64 0.55
TABLE 3
Basal acid secretion in wild-type and IP receptor knockout mice
Acid secretion was measured in wild-type and IP receptor knockout mice provided
with an acute gastric fistula under urethane anesthesia. The stomach was filled with
0.4 ml of saline through the fistula, the solution was changed after 20 min, and the
collected samples were used to measure acid output. Data are presented as the
mean S.E.M. from four mice.
Group No. of Mice Total Acid Output
Eq/2 h
Wild-type mice 4 3.01 0.40
IP knockout mice 4 3.51 0.43
552 Kotani et al.
at ASPET Journals on December 31, 2015jpet.aspetjournals.orgDownloaded from
volvement of COX-2/PGs in mucosal defense during I/R.
These results are consistent with the findings of Maricic et al.
(1999), who showed that I/R-induced gastric damage was
aggravated by administration of selective COX-2 inhibitors
such as NS-398 and DFU as assessed both macroscopically
and histologically. They also showed that I/R increased
COX-2 mRNA levels in the gastric mucosa and that dexa-
methasone aggravated the I/R-induced gastric lesions at a
dose that inhibited COX-2 expression following I/R (Maricic
et al., 1999). We also found in the present study the up-
regulation of COX-2 expression in the stomach following I/R.
These findings suggest that the gastric mucosa has the ca-
pacity to rapidly up-regulate COX-2 expression in response
to various stimuli. It should be noted that ischemia alone
neither caused any macroscopic damage nor up-regulated
COX-2 expression in the gastric mucosa. Schmedtje et al.
(1997) reported that hypoxia increased the expression of the
COX-2 gene in human vascular endothelial cells in vitro. It is
possible that the expression is up-regulated by ischemia
alone when examined at a later period. Notwithstanding, it is
assumed that COX-2 expression is up-regulated as one of the
protective mechanisms when the stomach is exposed to ul-
cerogenic stimuli.
On the other hand, we observed in this study that the
selective COX-1 inhibitor SC-560 did not significantly affect
the severity of I/R-induced gastric lesions, suggesting no role
for COX-1 in mucosal defense during I/R. However, Hirat-
suka et al. (2004) reported that SC-560 reduced the severity
of I/R-induced gastric lesions in mice, probably due to down-
regulation of free radical production during reperfusion by
decreasing blood flow. The discrepancy in these results re-
mains unexplained at present, yet it may be due to different
experimental conditions such as the dose of SC-560; they
used 40 mg/kg of this agent, 8 times higher than that (5
mg/kg) in the present study.
The most important finding of the present study is that the
severity of I/R-induced gastric lesions was markedly in-
creased in IP receptor knockout mice but not in the animals
lacking EP1 or EP3 receptors. These results suggest that
although I/R stimulated the generation of PGs to increase the
mucosal levels of both PGE
2
and 6-keto-PGF
1
, the stable
metabolite of PGI
2
, in the stomach, the type of prostanoid
responsible for mucosal defense during I/R is PGI
2
not PGE
2
.
These results are understandable, because the expression of
COX-2 in the gastric mucosa following I/R was observed
mainly in the endothelial cells (Hiratsuka et al., 2004) and
because PGI
2
is a major prostanoid produced in the endothe
-
lial cells (Konturek and Robert, 1982). We observed in this
study that iloprost, a stable analog of PGI
2
, significantly
prevented the I/R-induced gastric lesions in the absence or
presence of COX inhibitors, supporting the involvement of
endogenous PGI
2
in mucosal defense during I/R. This PGI
2
analog has an affinity for not only IP receptors but also EP
receptors as well (Narumiya and FitzGerald, 2001). In the
present study, however, iloprost had no effect on the devel-
opment of I/R-induced gastric lesions in IP receptor knockout
mice, excluding the involvement of EP receptors in the pro-
tective action of this agent. Harada et al. (1999, 2000) re-
ported that iloprost prevented stress-induced gastric lesions,
primarily by inhibiting leukocytes from accumulating. Since
I/R injury is a neutrophil-dependent response (Zimmerman
et al., 1990), it is assumed that selective COX-2 inhibitors
promote the adherence of leukocyte to the vascular endothe-
lium during I/R, thereby resulting in aggravation of the le-
sions in the stomach (Muscara et al., 2000). In the present
study, we observed a marked increase in MPO activity in the
gastric mucosa following I/R in wild-type mice, and this re-
sponse was significantly reduced by iloprost and further en-
hanced in IP receptor knockout animals, confirming the in-
hibitory role for PGI
2
/IP receptors in the neutrophil-related
process of I/R-induced gastric injury. These results all
strongly suggest that endogenous PGI
2
produced by COX-2
plays a role in mucosal defense during I/R through the acti-
vation of IP receptors.
We previously examined, using various subtype-specific
EP receptor agonists and antagonists, the relationship be-
tween EP receptor subtypes and PGE
2
-induced gastric cyto
-
protection and found that PGE
2
exhibits a protective action
against a variety of gastric lesions mediated by the activation
of EP1 receptors (Araki et al., 2000; Suzuki et al., 2001;
Takeuchi et al., 2002a). The present results, however, sug-
gest that neither EP1 nor EP3 receptors participate in mu-
cosal defense during I/R in the stomach, although the muco-
Fig. 8. A, effect of PGE
2
on I/R-induced gastric lesions in wild-type mice.
PGE
2
(0.11 mg/kg) was given i.v. 5 min before reperfusion. Data are
presented as the mean S.E.M. from five to six mice. , significant
difference from control at p 0.05. B, effect of PGE
2
on I/R-induced
gastric lesions in IP receptor knockout mice. PGE
2
(1 mg/kg) was given
i.v. 5 min before reperfusion. Data are presented as mean S.E.M. from
four to six mice.
PGI
2
and Ischemia/Reperfusion-Induced Gastric Injury 553
at ASPET Journals on December 31, 2015jpet.aspetjournals.orgDownloaded from
sal PGE
2
content was significantly elevated following I/R. Of
course, the present data do not totally exclude the involve-
ment of PGE
2
in mucosal defense during I/R. It has been
reported that PGE
2
protects against ischemia- or I/R-induced
injury in brain, liver, and heart through EP2 or EP4 recep-
tors (McCullough et al., 2004; Xiao et al., 2004; Kuzumoto et
al., 2005). We also found in the present study that PGE
2
significantly reduced the severity of these lesions in both
wild-type and IP receptor knockout mice, yet the effective
dose was much higher and the effect was much less pro-
nounced compared with iloprost. In a preliminary study, we
also observed that the effect of PGE
2
was significantly af
-
fected by neither EP1, EP3, nor EP4 antagonists (data not
shown). At present, it remains unknown whether or not this
effect of PGE
2
is physiological action, yet there is a possibility
that PGE
2
exhibits a protective effect against the I/R-induced
gastric lesions, probably through EP2 receptors. Hoshino et
al. (2003) reported that PGE
2
inhibited the irritant-induced
apoptosis via EP2/EP4 receptors and speculated that this
action is involved in the gastroprotective action of PGE
2
in
vivo conditions. However, we previously reported that nei-
ther specific EP2 nor EP4 agonists protected the stomach
against acidified ethanol or indomethacin in rats (Araki et
al., 2000; Suzuki et al., 2001). Furthermore, no evidence has
been reported the involvement of apoptotic changes in the
I/R-induced gastric lesions. On the other hand, it is known
that PGE
2
inhibits the neutrophil migration in the gastric
mucosa via EP2/EP4 receptors (Suzuki et al., 2001). Thus, it
is assumed that PGE
2
prevents I/R-induced gastric lesions
via inhibition of the neutrophil-related process but not apo-
ptosis, similar to PGI
2
.
It has been reported that gastric acid secretion is substan-
tially decreased after ischemia and remained reduced for
several hours even after reperfusion (Takeuchi et al., 1986;
Nakamoto et al., 1998). However, Kitano et al. (1997) showed
that cimetidine, the histamine H
2
receptor antagonist, had a
protective effect on I/R-induced gastric lesions through the
suppression of acid secretion. In a preliminary study, we
observed that lansoprazole, a proton pump inhibitor, also
significantly reduced the severity of I/R-induced gastric le-
sions in wild-type mice (data not shown), suggesting the
participation of gastric acid in the pathogenesis of these
lesions. Since iloprost at the highest dose (3
g/kg) caused a
slight decrease in acid secretion, consistent with the finding
by Seidler et al. (1989), it is possible that the protective effect
of this prostanoid, especially at high doses, on I/R-induced
gastric lesions is partly accounted for by its antisecretory
action.
Based on all the results of the present study, we confirmed
that I/R induced gastric lesions with the up-regulation of
COX-2 expression and that the damage was significantly
aggravated by indomethacin as well as the selective COX-2
inhibitor rofecoxib but not the selective COX-1 inhibitor SC-
560. We further showed for the first time that the I/R-induced
gastric lesions were significantly worsened in IP receptor
knockout mice but not in the animals lacking EP1 or EP3
receptors. Thus, it is assumed that endogenous PGs derived
from COX-2 play a crucial role in gastric mucosal defense
during I/R, and this action is mainly mediated by PGI
2
through the activation of IP receptors.
Acknowledgments
We thank Professor Shu Narumiya, Kyoto University Faculty
(Kyoto, Japan) of medicine for kindly supplying EP1, EP3, and IP
receptor knockout mice.
References
Araki H, Ukawa H, Sugawa Y, Yagi K, Suzuki K, and Takeuchi K (2000) The roles
of prostaglandin E receptor subtypes in the cytoprotective action of prostaglandin
E2 in rat stomach. Aliment Pharmacol Ther 14 (Suppl 1):116–124.
Chamoun F, Burne M, O’Donnell M, and Rabb H (2000) Pathophysiologic role of
selectins and their ligands in ischemia reperfusion injury. Front Biosci 5:E103–
E109.
Cheung JY, Bonventre JV, Malis CD, and Leaf A (1986) Calcium and ischemic
injury. N Engl J Med 314:1670 –1676.
Davies NM, Sharkey KA, Asfaha S, Macnaughton WK, and Wallace JL (1997)
Aspirin causes rapid up-regulation of cyclo-oxygenase-2 expression in the stomach
of rats. Aliment Pharmacol Ther 11:1101–1108.
Dimmitt RA, Glew R, Colby C, Brindle M, Skarsgard E, and Moss RL (2003) Serum
cytosolic beta-glucosidase activity in a rat model of necrotizing enterocolitis. Pe-
diatr Res 54:462–465.
Farber JL, Chien KR, and Mittnacht S Jr (1981) Myocardial ischemia: the patho-
genesis of irreversible cell injury in ischemia. Am J Pathol 102:271–281.
Futaki N, Takahashi S, Yokoyama M, Arai I, Higuchi S, and Otomo S (1994) NS-398,
a new anti-inflammatory agent, selectively inhibits prostaglandin G/H synthase/
cyclooxygenase (COX-2) activity in vitro. Prostaglandins 47:55–59.
Granger DN and Kubes P (1994) The microcirculation and inflammation: modulation
of leukocyte-endothelial cell adhesion. J Leukoc Biol 55:662–675.
Harada N, Okajima K, Liu W, and Uchiba M (2000) Activated neutrophils impair
gastric cytoprotection role of neutrophil elastase. Dig Dis Sci 45:1210–1216.
Harada N, Okajima K, Murakami K, Isobe H, and Liu W (1999) Gastric prostacyclin
(PGI
2
) prevents stress-induced gastric mucosal injury in rats primarily by inhib
-
iting leukocyte activation. Prostaglandins Other Lipid Mediat 57:291–303.
Hiratsuka T, Futagami S, Tatsuguchi A, Ueki N, Suzuki K, Shinji Y, Kusunoki M,
Shinoki K, Nishigaki H, Wada K, et al. (2004) Distinct roles of COX-1 and COX-2
in gastric ischemia-reperfusion injuries in mice (Abstract). Gastroenterology 126:
A-543.
Hoshino T, Tsutsumi S, Tomisato W, Hwang HJ, Tsuchiya T, and Mizushima T
(2003) Prostaglandin E
2
protects gastric mucosal cells from apoptosis via EP2 and
EP4 receptor activation. J Biol Chem 278:12752–12758.
Kitano M, Wada K, Kamisaki Y, Nakamoto K, Kishimoto Y, Kawasaki H, and Itoh
T (1997) Effects of cimetidine on acute gastric mucosal injury induced by ischemia-
reperfusion in rats. Pharmacology 55:154 –164.
Koga T, Shibahara K, Kabashima A, Sumiyoshi Y, Kimura Y, Takahashi I, Kakeji Y,
and Maehara Y (2004) Overexpression of cyclooxygenase-2 and tumor angiogene-
sis in human gastric cancer. Hepatogastroenterology 51:1626 –1630.
Konturek SJ and Robert A (1982) Cytoprotection of canine gastric mucosa by pros-
tacyclin: possible mediation by increased mucosal blood flow. Digestion 25:155–
163.
Krawisz JE, Sharon P, and Stenson WF (1984) Quantitative assay for acute intes-
tinal inflammation based on myeloperoxidase activity: assessment of inflamma-
tion in rat and hamster models. Gastroenterology 87:1344–1350.
Kuzumoto Y, Sho M, Ikeda N, Mizuno T, Hamada K, Akashi S, Tsurui Y, Kashizuka
H, Nomi T, Kanehiro H, and Nakajima Y (2005) Role of EP4 prostaglandin E2
receptor in the ischemic liver. Transplant Proc 37:422–424.
Laudanno OM, Cesolari JA, Esnarriaga J, Rista L, Piombo G, Maglione C, Aram-
berry L, Sambrano J, Godoy A, and Rocaspana A (2001) Gastrointestinal damage
induced by celecoxib and rofecoxib in rats. Dig Dis Sci 46:779 –784.
Maricic N, Ehrlich K, Gretzer B, Schuligoi R, Respondek M, and Peskar BM (1999)
Selective cyclo-oxygenase-2 inhibitors aggravate ischaemia-reperfusion injury in
the rat stomach. Br J Pharmacol 128:1659 –1666.
McCullough L, Wu L, Haughey N, Liang X, Hand T, Wang Q, Breyer RM, and
Andreasson K (2004) Neuroprotective function of the PGE2 EP2 receptor in cere-
bral ischemia. J Neurosci 24:257–268.
Miura S, Tatsuguchi A, Wada K, Takeyama H, Shinji Y, Hiratsuka T, Futagami S,
Miyake K, Gudis K, Mizokami Y, et al. (2004) Cyclooxygenase-2-regulated vascu-
lar endothelial growth factor release in gastric fibroblasts. Am J Physiol 287:
G444–G551.
Mizuno H, Akamatsu T, and Kasuga M (1997) Induction of cyclooxygenase-2 in
gastric mucosal lesions and its inhibition by the specific antagonist delays healing
in mice. Gastroenterology 112:387–397.
Muscara MN, Vergnolle N, Lovren F, Triggle CR, Elliott SN, Asfaha S, and Wallace
JL (2000) Selective cyclo-oxygenase-2 inhibition with celecoxib elevates blood
pressure and promotes leukocyte adherence. Br J Pharmacol 129:1423–1430.
Nakamoto K, Wada K, Kitano M, Kishimoto Y, Ashida K, Kamisaki Y, Kawasaki H,
and Itoh T (1998) The role of endogenous acid in the development of acute gastric
ulcer induced by ischemia-reperfusion in the rat. Life Sci 62:PL63–PL69.
Narumiya S and FitzGerald GA (2001) Genetic and pharmacological analysis of
prostanoid receptor function. J Clin Investig 108:25–30.
Piper HM, Meuter K, and Schafer C (2003) Cellular mechanisms of ischemia-
reperfusion injury. Ann Thorac Surg 75:S644 –S668.
Riaz AA, Wan MX, Schafer T, Dawson P, Menger MD, Jeppsson B, and Thorlacius H
(2002) Allopurinol and superoxide dismutase protect against leucocyte-
endothelium interactions in a novel model of colonic ischaemia-reperfusion. Br J
Surg 89:1572–1580.
Saika M, Ueyama T, and Senba E (1999) Prostacyclin analog prevents stress-induced
expression of immediate early genes and gastric mucosal lesions in the rat stom-
ach. Life Sci 64:PL235–PL240.
Schmedtje JF, Ji YS, Liu WL, DuBois RN, and Runge MS (1997) Hypoxia induces
554 Kotani et al.
at ASPET Journals on December 31, 2015jpet.aspetjournals.orgDownloaded from
cyclooxygenase-2 via the NF-kappaB p65 transcription factor in human vascular
endothelial cells. J Biol Chem 272:601– 608.
Seidler U, Beinborn M, and Sewing KF (1989) Inhibition of acid formation in rabbit
parietal cells by prostaglandins is mediated by the prostaglandin E2 receptor.
Gastroenterology 96:314 –320.
Soll AH, Weinstein WM, Kurata J, and McCarthy D (1991) Nonsteroidal anti-
inflammatory drugs and peptic ulcer disease. Ann Intern Med 114:307–319.
Sugimoto Y, Namba T, Honda A, Hayashi Y, Negishi M, Ichikawa A, and Narumiya
S (1992) Cloning and expression of a cDNA for mouse prostaglandin E receptor
EP3 subtype. J Biol Chem 267:6463– 6466.
Suzuki K, Araki H, Mizoguchi H, and Takeuchi K (2001) E type prostaglandin
inhibits indomethacin-induced gastric lesions in rats through EP1 receptors: Im-
portance of antigastric motility action. Digestion 63:92–101.
Takeeda M, Yamato M, Kato S, and Takeuchi K (2003) Cyclooxygenase isozymes
involved in adaptive functional responses in rat stomachs following barrier dis-
ruption. J Pharmacol Exp Ther 307:713–719.
Takeuchi K, Araki H, Komoike Y, Umeda M, and Suzuki K (2002a) Adaptive gastric
cytoprotection is mediated by prostaglandin EP1 receptors: a study using rats and
knockout mice. J Pharmacol Exp Ther 297:1160 –1165.
Takeuchi K, Kato K, and Tanaka A (2002b) Gastrointestinal protective action of
prostaglandin E
2
and EP receptor subtypes, in Frontiers of Gastrointestinal Re
-
search: Gastrointestinal Mucosal Repair and Experimental Therapeutics (Cho CH
and Wang JY eds) vol 25, pp 227–242, Karger AG, Basel.
Takeuchi K, Ohno T, and Okabe S (1986) Variations of gastric transmucosal poten-
tial difference and lesion formation in the rat during hemorrhagic shock. Gastro-
enterology 91:1113–1122.
Tanaka A, Hase S, Miyazawa T, and Takeuchi K (2002) Up-regulation of cyclooxy-
genase-2 by inhibition of cyclooxygenase-1: a key to nonsteroidal anti-
inflammatory drug-induced intestinal damage. J Pharmacol Exp Ther 300:754
761.
Ushikubi F, Segi E, Sugimoto Y, Murata T, Matsuoka T, Kobayashi T, Hizaki H,
Tuboi K, Katsuyama M, Ichikawa A, et al. (1998) Impaired febrile response in mice
lacking the prostaglandin E receptor subtype EP3. Nature (Lond) 395:281–284.
Vane JR and Botting RM (1995) New insights into the mode of action of anti-
inflammatory drugs. Inflamm Res 44:1–10.
Wada K, Kamisaki Y, Kitano M, Kishimoto Y, Nakamoto K, and Itoh T (1996) A new
gastric ulcer model induced by ischemia-reperfusion in the rat: role of leukocytes
on ulceration in rat stomach. Life Sci 59:PL295–PL301.
Whittle BJ, Kauffman GL Jr, and Boughton-Smith NK (1984) Stimulation of gastric
alkaline secretion by stable prostacyclin analogues in rat and dog. Eur J Pharma-
col 100:277–283.
Xiao CY, Yuhki K, Hara A, Fujino T, Kuriyama S, Yamada T, Takayama K, Taka-
hata O, Karibe H, Taniguchi T, et al. (2004) Prostaglandin E2 protects the heart
from ischemia-reperfusion injury via its receptor subtype EP4. Circulation 109:
2462–2468.
Yasue N, Chan ET, Kaplowitz N, and Guth PH (1992) Effect of phorone and allo-
purinol on ischemia-reperfusion injury in gastrointestinal mucosa of the rat.
Pharmacology 44:334 –343.
Zimmerman BJ, Guillory DJ, Grisham MB, Gaginella TS, and Granger DN (1990)
Role of leukotriene B4 in granulocyte infiltration into the postischemic feline
intestine. Gastroenterology 99:1358–1363.
Address correspondence to: Dr. Koji Takeuchi, Department of Pharma-
cology and Experimental Therapeutics, Kyoto Pharmaceutical University,
Misasagi, Yamashina, Kyoto 607-8414, Japan. E-mail: takeuchi@mb.
kyoto-phu.ac.jp
PGI
2
and Ischemia/Reperfusion-Induced Gastric Injury 555
at ASPET Journals on December 31, 2015jpet.aspetjournals.orgDownloaded from
... Gastrointestinal injury after DCF administration can occur through its pharmacologic effects, namely inhibition of cyclooxygenase (COX) enzymes. COX metabolism of arachidonic acid gives rise to a number of physiologically important derivatives such as prostaglandin E 2 (PGE 2 ) which functions to protect the GI mucosa (Kotani et al., 2006). ...
... Our in vitro assays repeatedly demonstrated that DCF-AG possesses the ability to inhibit both COX-1 and COX-2, and that the inhibition leads to decreased synthesis of prostaglandin E 2 from arachidonic acid. Considering the role PGE 2 has in protecting the GI tract and that inhibition of both COX enzymes is necessary for intestinal injury to develop (Tanaka et al., 2001;Sigthorsson et al., 2002;Hoshino et al., 2003;Kotani et al., 2006), DCF-AG modulation of COX activity may have exacerbated the GI injury normally ascribed to DCF. Ostensibly, we believe our studies demonstrate that DCF-AG directly contributed to enteropathy in our in vivo models via active uptake by mouse Oatp2b1 resulting in enhanced cytotoxicity in Mrp3 KO animals, due to decreased basolateral excretion (and greater enterocyte retention) from the combined effects of ROS generation, SOD inhibition, and attenuation of the PGE 2 -derived protection of the intestinal tract. ...
Article
Diclofenac (DCF) is commonly used in the treatment of arthritis and pain. Although mechanisms of injury following DCF administration have been characterized, the roles that transporters have as modulators of exposure and toxicity remain unclear. To that end, in vivo transporter knockout (KO) models were used to assess the toxicokinetics of DCF and its major metabolites. DCF and its reactive acyl glucuronide (DCF-AG) were shown to be substrates for the efflux transporter Bcrp. Biliary excretion of DCF and DCF-AG decreased in Bcrp KOs while only DCF-AG plasma levels increased compared to wild-type (WT) mice. DCF-AG was likewise determined to be a substrate for the efflux transporter Mrp3 evidenced by reduced plasma concentrations in Mrp3 KOs without changes in biliary excretion. Toxic DCF challenge resulted in increased intestinal injury in Mrp3 KOs compared to WTs. In vitro assays revealed that human MRP3 can transport DCF-AG with apparent high affinity compared to human MRP2. The active uptake of DCF-AG was shown to be mediated by human OATPs in an in vitro system, specifically OATP2B1 exhibited concentration-dependent kinetics. Mechanistic studies indicated that DCF-AG was able to induce oxidative stress through creation of reactive oxygen species and inhibition of superoxide dismutase. Furthermore, DCF-AG was shown to inhibit both COX-1 and COX-2, which may have contributed to intestinal injury. Lastly it was demonstrated that DCF-AG can inhibit human MRP4 that is upregulated during oxidative stress. In summary, the data presented herein demonstrate the multifactorial pathways by which DCF-AG is transported and contributes to tissue injury.
... It functions in the stomach through EP2/EP4 and IP receptors. 24 Prostaglandins also inhibit mast cell stimulation as well as adhesion of leukocytes and platelets to the vascular endothelium (Halter et al, Brzozowski et al). 25 Coxibs are selective COX-2 inhibitors. ...
Article
Full-text available
Background: Around the globe acid peptic disease and its potential complications are among the major cause of morbidity and is a significant burden on health system. Till now the main stay of treatment are the pharmaceutical agents which decrease the secretion of acid. But these have potential complications and tolerability issues. Among such plants one is Acacia catechu commonly available in India and Pakistan.Methods: This experimental study involved 48 albino rats that were divided into four groups. Group A, B, C, and D were given water, 100mg aspirin, 100 mg aspirin+ 250 mg Acacia catechu and 100 mg aspirin+500 mg Aacacia catechu respectively, and were sacrificed on day 3, 7 and 14th day of study and observed for changes.Results: Difference between blood vessels in subgroup B1 and subgroup C1 with subgroup D1 was statistically significant (p=0.05) and (p=0.001). Group C1 and D1 had normal vessels in mucosa and submucosa. However, dilatation of blood vessels was noted in both groups receiving Acacia catechu along with ASA.Conclusions: Aqueous extracts of Acacia catechu stem and bark showed significant anti-ulcer and anti-inflammatory activities by increasing blood flow to the stomach.
... NO vasodilatation, a basic gastric mucosal defense, provides blood to the mucosa to resist damage from gastric acid and pepsin (32,33). However, the role of NO in the gastrointestinal tract is not just a defense mechanism, and previous studies (18,19) have revealed that NO may play a dual role in ischemia-reperfusion-induced mucosal damage (34). ...
Article
Full-text available
Stress-related mucosal disease (SRMD) is a common complication in patients in the intensive care unit (ICU). The aim of the present study was to investigate the possible mechanisms for the pathogenesis of SRMD. In total, 38 patients with SRMD were enrolled from an ICU, as well as 15 healthy volunteers. The disease severity of patients in ICU was evaluated using the Acute Physiology and Chronic Health Evaluation (APACHE) II score. Gastric mucosa with the most severe lesions were biopsied for hematoxylin and eosin staining and then assessed by pathological damage scoring. The serum levels of malondialdehyde (MDA), superoxide dismutase (SOD) and ischemic modified albumin (IMA) were also detected. In addition, claudin-3 and inducible nitric oxide (NO) synthase (iNOS) in the gastric mucosa were assessed by western blotting and immunohistochemistry. The average APACHE II score of the patients with SRMD was significantly higher compared with the controls. Moreover, the levels of MDA (4.74±2.89 nmol/ml) and IMA (93.61±10.78 U/ml) in patients with SRMD were significantly higher compared with the controls (P<0.001), while those of SOD (89.66±12.85 U/ml) in the patients with SRMD were significantly lower compared with the controls (P<0.001). Furthermore, compared with the control, iNOS expression was significantly higher (P=0.034), while the expression of claudin-3 was significantly lower in patients with SRMD (P<0.001). The results indicated that APACHE II score was positively correlated with pathological damage score (r=0.639, P<0.001) and levels of MDA (r=0.743, P<0.001), but negatively correlated with the level of SOD (r=-0.392, P=0.015). In addition, MDA was positively correlated with IMA (r=0.380, P=0.018), but negatively correlated with claudin-3 (r=-0.377, P=0.020). Therefore, it was speculated that oxidative stress may play an important role in the pathogenesis of SRMD, and NO levels and cell membrane permeability are altered during this process.
... ;Wallace (2006);Takeuchi et al. (2014);Kotani et al. (2006) Calcitonin gene-related peptide (CGRP) and nitric oxide -Increasing the mucin synthesis -Inducing the submucosal vasodilation and enhance mucosal blood flow -Acid secretion inhibition -Inducing the effects of anti-inflammatoryLi et al. (1992); Holzer et al. (1998); Gyires et al. (2007); Holzer et al. (2007) Somatostatin -Decrease the elevated level of substance P, VIP and leukotriens -Has antioxidant, anti-inflammatory and anti-apoptotic roles Karmeli et al. (1994); Nassar et al. Antioxidant or reactive metabolite-eliminating effects -Counteract oxidative stress Szabo et al. (1992); Ali et al. (1995) Hydrogen sulfide (H2S) -Increases mucosal blood flow -Stimulates bicarbonate secretion -Reduces proinflammatory cytokine production and -Leukocyte-endothelial adherence -Increases prostaglandin synthesis -Decreases reactive oxygen metabolite production -Enhances tissue repair Wallace et al. ...
Article
Full-text available
A peptic ulcer is an ulcer of one of those areas of the gastrointestinal tract that are usually acidic. Peptic ulcer formation occurred due to the acid of excess, peptic activity in gastric juice as well as a breakdown in mucosal defenses. Some of anti-ulcer medicines such as proton pump inhibitors and H2 receptor antagonists are used for peptic ulcer treatment. However, these drugs have shown disease relapse, side effects, and even drug interactions. Many medicinal plants exhibit anti-ulcer activity and found useful in the treatment of peptic ulcer. The purpose of this review is to know more about the anti-ulcer properties of the medicinal plants. Serjania marginata (HESM) leaves, thymol obtained from Thymus, Origanum and Cuphea aequipetala infusion (CAI) In this review there is information about some kinds of plants which are very useful in treating peptic ulcer disease such as hydroalcoholic extract obtained from S. marginata (HESM) leaves, thymol obtained from Thymus, Origanum and CAI. These plants were studied by their mechanisms underlying to the gastroprotective effect of thymol and HESM. Furthermore, the study reported the potential effect of involvement of some factors of the protective effect of medical extract, such as prostaglandins (PG), nitric oxide (NO), and a hydrogen sulfide (H2S). Side the effect of these factors in gastro protective, and finally the paper clarified some local mechanisms of actions of some compounds that were involved in mucosal defense or injury.
... Gastroprotection of the gastric mucosa is said to be controlled by the gastric microcirculation. Prostaglandin of the E series (PGE 2 ) is a potent vasodilator, producing its effect in the stomach through EP2/EP4 receptors [22,23]. It increases mucosal blood flow, thereby increasing the resistance of the gastric mucosa to injury [24]. ...
... Gastrointestinal injury after DCF administration occurs through inhibition of cyclooxygenase (COX) enzymes that metabolize arachidonic acid into physiologically important cytoprotective derivatives such as prostaglandin E 2 (PGE 2 ) (Kotani et al., 2006). DCF, a nonselective inhibitor of COX-1 and COX-2, promotes GI injury though decreased PGE 2 signaling. ...
Article
Full-text available
We have previously reported that mice lacking the efflux transporter Mrp3 had significant intestinal injury after toxic diclofenac (DCF) challenge, and proposed that diclofenac acyl glucuronide (DCF-AG), as a substrate of Mrp3, played a part in mediating injury. Since both humans and mice express the uptake transporter OATP2B1 in the intestines, OATP2B1 was characterized for DCF-AG uptake. In vitro assays using human embryonic kidney (HEK)-OATP2B1 cells demonstrated that DCF-AG was a substrate with a maximal velocity (Vmax) and Km of 17.6 ± 1.5 pmol/min per milligram and 14.3 ± 0.1 μM, respectively. Another key finding from our in vitro assays was that DCF-AG was more cytotoxic compared with DCF, and toxicity occurred within 1-3 hours of exposure. We also report that 1 mM DCF-AG caused a 6-fold increase in reactive oxygen species (ROS) by 3 hours. Investigation of oxidative stress through inhibition of superoxide dismutase (SOD) revealed that DCF-AG had 100% inhibition of SOD at the highest tested dose of 1 mM. The SOD and ROS results strongly suggest DCF-AG induced oxidative stress in vitro. Lastly, DCF-AG was screened for pharmacologic activity against COX-1 and COX-2 and was found to have IC50 values of 0.620 ± 0.105 and 2.91 ± 0.36 μM, respectively, which represents a novel finding. Since cyclooxygenase (COX) inhibition can lead to intestinal ulceration, it is plausible that DCF-AG can also contribute to enteropathy via COX inhibition. Taken in context, the work presented herein demonstrated the multifactorial pathways by which DCF-AG can act as a direct contributor to toxicity following DCF administration.
... Research has shown the increase in pro-inflammatory cytokines including tumor necrosis factor (TNF)-α and interleukin (IL)-1b in rats exposed to stress [51,52]. It was also reported that COX-2 protein is highly localised in fibroblasts, monocytes/macrophages and granulocytes in the base of gastric ulcers in rats [53]. ...
Article
Full-text available
Introduction: Piper sarmentosum (Piperaceae) is traditionally used by Asians to treat numerous common ailments including asthma, fever and gastritis. The aim of the research was to determine and compare the effects of Piper sarmentosum (PS) with omeprazole (OMZ) on gastric parameters in rats exposed to restraint stress. Material and methods: The methanolic extract of PS was prepared in the dose of 500 mg/kg. Twenty-eight male Wistar rats were assigned to 4 equal sized groups: two control groups and two treated groups which were supplemented with either PS or OMZ orally at a dose of 500 mg/kg and 20 mg/kg body weight respectively. After 28 days of treatment, one control group, the PS and OMZ group were subjected to a single exposure of water-immersion restraint stress for 3.5 h. After the last exposure to stress, the stomach was excised for evaluation of the parameters. Results: Oral supplementation of PS was as effective in preventing the formation of gastric lesion when compared with OMZ (p < 0.05). The increased gastric acidity and MDA due to stress was also reduced with supplementation of PS and OMZ. Only PS had the ability to reduce prostaglandin E 2 loss (p = 0.0067) and have the ability to down regulate cyclooxygenase-2 (COX-2) mRNA expression (p = 0.01) with stress exposure. Conclusions: Piper sarmentosum possesses a similar protective effect against stress-induced gastric lesions as omeprazole. The protective effect was associated with decreased lipid peroxidation, increased prostaglandin E 2, reduction in gastric acidity and reduction in COX-2 mRNA expression which was altered by stress.
... Nel modello murino di danno della mucosa gastrica da ischemia-riperfusione, il pretrattamento con iloprost previene significativamente le lesioni gastriche di tipo emorragico, sia in assenza che in presenza di indometacina e di rofecoxib (124) . Sempre nel modello sperimentale murino è stato dimostrato un effetto protettivo sulle causticazioni esofagee provocate da soda caustica (125) . ...
Article
Full-text available
We have previously reported that mice lacking the efflux transporter Mrp3 had significant intestinal injury after toxic diclofenac (DCF) challenge, and proposed that diclofenac acyl glucuronide (DCF-AG), as a substrate of Mrp3, played a part in mediating injury. Since both humans and mice express the uptake transporter OATP2B1 in the intestines, OATP2B1 was characterized for DCF-AG uptake. In vitro assays using HEK-OATP2B1 cells demonstrated that DCF-AG was a substrate with a Vmax and Km of 17.6 ± 1.5 pmol/min/mg and 14.3 ± 0.1 µM, respectively. Another key finding from our in vitro assays was that DCF-AG was more cytotoxic compared to DCF, and toxicity occurred within 1 to 3 hours of exposure. We also report that 1 mM DCF-AG caused a 6-fold increase in reactive oxygen species (ROS) by 3 hours. Investigation of oxidative stress through inhibition of superoxide dismutase (SOD) revealed that DCF-AG had 100% inhibition of SOD at the highest tested dose of 1 mM. The SOD and ROS results strongly suggest DCF-AG induced oxidative stress in vitro. Lastly, DCF-AG was screened for pharmacologic activity against COX-1 and COX-2 and was found to have IC50 values of 0.620 ± 0.105 and 2.91 ± 0.36 µM, respectively, which represents a novel finding. Since COX inhibition can lead to intestinal ulceration, it is plausible that DCF-AG can also contribute to enteropathy via COX inhibition. Taken into context, the work presented herein demonstrated the multifactorial pathways by which DCF-AG can act as a direct contributor to toxicity following DCF administration.
Article
Liver transplantation is the only effective treatment for end-stage liver diseases. Ischemia-reperfusion injury remains a major cause of post-transplantation liver dysfunction and even failure. Ischemic postconditioning is defined as rapid intermittent periods of reperfusion and ischemia in the early phase of repefusion after long ischemia of the tissues and organs. Many investigations have demonstrated that ischemic postconditioning has a protective effect against hepatic ichemia-reperfusion injury. Ischemic postconditioning exerts protective effects through many possible mechanisms such as oxygen free radicals, calcium overload, polymorphonuclear neutrophils, cytokines, cell apoptosis and mitochondria.
Article
Full-text available
A functional cDNA clone for mouse EP3 subtype of prostaglandin (PG) E receptor was isolated from a mouse cDNA library using polymerase chain reaction based on the sequence of the human thromboxane A2 receptor and cross-hybridization screening. The mouse EP3 receptor consists of 365 amino acid residues with putative seven-transmembrane domains. The sequence revealed significant homology to the human thromboxane A2 receptor. Ligand binding studies using membranes of COS cells transfected with the cDNA revealed specific [3H]PGE2 binding. The binding was displaced with unlabeled PGs in the order of PGE2 = PGE1 greater than iloprost greater than PGF2 alpha greater than PGD2. The EP3-selective agonists, M&B 28,767 or GR 63799X, potently competed for the [3H]PGE2 binding, but no competition was found with EP1- or EP2-selective ligands. PGE2 and M&B 28,767 decreased forskolin-induced cAMP formation in a concentration-dependent manner in Chinese hamster ovary cells permanently expressing the cDNA. Northern blot analysis demonstrated that the EP3 mRNA is expressed abundantly in kidney, uterus, and mastocytoma P-815 cells and in a lesser amount in brain, thymus, lung, heart, stomach, and spleen.
Article
Nonsteroidal anti-inflammatory drugs (NSAIDs) induce gastrointestinal ulceration as the adverse reaction. This effect of NSAIDs is attributable to endogenous prostaglandin (PG) deficiency caused by inhibition of cyclooxygenase (COX), yet the relation between COX inhibition and the gastrointestinal ulcerogenic property of NSAIDs remains controversial. Using selective COX inhibitors, we examined whether inhibition of COX-1 or COX-2 alone is sufficient for induction of intestinal damage in rats. Various COX inhibitors were administered p.o. in rats, and the animals were killed 24 h later. Mucosal PGE2 levels were determined by enzyme immunoassay, whereas the gene expression of COX isozymes was examined by reverse transcription-polymerase chain reaction. Nonselective COX inhibitors such as indomethacin inhibited PGE2 production and caused damage in the small intestine. Selective COX-2 inhibitors (rofecoxib or celecoxib) had no effect on the generation of PG, resulting in no damage. A selective COX-1 inhibitor (SC-560) did not cause damage, despite reducing PGE2 content. However, the combined administration of COX-1 and COX-2 inhibitors provoked intestinal damage with an incidence of 100%. COX-2 was up-regulated in the small intestine after administration of SC-560, and the PGE2content was restored 6 h later, in a rofecoxib-dependent manner. The intestinal lesions induced by SC-560 plus rofecoxib were significantly prevented by later administration of 16,16-dimethyl PGE2. These results suggest that the intestinal ulcerogenic property of NSAID is not accounted for solely by inhibition of COX-1 and requires inhibition of COX-2 as well. The inhibition of COX-1 up-regulates COX-2 expression, and this may be a key to NSAID-induced intestinal damage.
Article
We studied the effect of inhibition of oxyradical formation and of endogenous glutathione (GSH) depletion on lesion formation in the gastrointestinal tract in a modified rat hemorrhagic shock model (1 h hypotension and 1 h reperfusion). Allopurinol, an inhibitor of xanthine oxidase, did not protect against lesion formation. This suggests that oxygen radicals generated from xanthine oxidase may not be the major cause of injury under these conditions of prolonged 'ischemia'-reperfusion. Phorone (diisopropylideneacetone), a GSH depletor, decreased mucosal GSH levels in the corpus, duodenum and small intestine, and also significantly reduced lesion formation histologically in the corpus, antrum, duodenum and small intestine. However, there was no significant differences in mucosal blood flow (as estimated by changes in mucosal hemoglobin concentrations and oxygen saturation of mucosal hemoglobin) in the corpus, antrum, duodenum and small intestine between phorone-pretreated and control rats. We conclude that phorone decreased mucosal GSH concentrations and exerted a protective effect against hemorrhagic shock-induced gastrointestinal mucosal lesions. The protective effect appears to be independent of mucosal blood flow.
Article
Evidence has accumulated that nonsteroidal anti-inflammatory drugs (NSAIDs) cause clinically important gastroduodenal ulcers. The pathogenesis, which involves the impairment of mucosal resistance to injury in an acid-peptic environment, is multifactorial and controversial. Ulcers caused by NSAIDs can occur either in mucosa inflamed because of infection with Helicobacter pylori or in histologically normal mucosa. The use of these drugs has been linked to an unexpectedly high incidence of ulcer complications, and a history of peptic ulcer disease is common in such cases. Nonsteroidal anti-inflammatory drugs thus appear both to exacerbate an underlying peptic diathesis and to cause de novo ulcers. The association between the use of these drugs and ulcer complications is supported by ulcer prevalence data from cross-sectional studies, and by data from case-controlled and cohort studies, and from randomized, experimental trials. Drug-induced gastric ulcers have been prevented by misoprostol, but not by H2 blocker therapy. Several therapies have been reported to promote ulcer healing despite continued use of NSAIDs, but adequate controlled trials have not been done. Small gastric and duodenal ulcers readily heal, whereas larger gastric ulcers require vigorous and prolonged therapy. The relative efficacies of various therapies in preventing ulcers, healing ulcers, or preventing complications remain to be established.
Article
Several studies have demonstrated that granulocytes accumulate in the intestinal mucosa following ischemia/reperfusion. It has been suggested that leukotriene B4 may be released during ischemia/reperfusion and consequently may promote granulocyte infiltration into the mucosa. The objectives of this study were to determine whether (a) leukotriene B4 is produced in the gut mucosa during ischemia and reperfusion, and (b) inhibition of leukotriene B4 attenuates granulocyte infiltration into the postischemic intestinal mucosa. Isolated segments of cat intestine were subjected to 3 hours of ischemia and 1 hour of reperfusion. Mucosal samples were obtained during baseline, ischemia at 3 hours and reperfusion at 1 hour. Leukotriene B4 production was determined by radioimmunoassay. Tissue-associated myeloperoxidase activity was used to quantitate granulocyte accumulation in the mucosal samples. In untreated animals, mucosal leukotriene B4 concentration was higher at reperfusion compared with baseline levels. The reperfusion-induced increase in mucosal leukotriene B4 was entirely prevented by pretreatment with either nordihydroguaiaretic acid (Sigma Chemical Co., St. Louis, MO) or L663,536 (Merck-Frosst, Montreal, Quebec, Canada), two potent lipoxygenase inhibitors. Both lipoxygenase inhibitors, as well as leukotriene B4 antagonist (SC-41930) significantly attenuated the reperfusion-induced infiltration of granulocytes. These results indicate that leukotriene B4 plays an important role in mediating the granulocyte accumulation elicited by reperfusion of the ischemic bowel.
Article
The affinities of seven natural and synthetic prostaglandins [PGE2; 16,16-dimethyl PGE2; iloprost (stable prostacyclin analogue); PGF2 alpha; PGD2; BW245c (stable PGD2 analogue); and U46619 (stable thromboxane analogue)] to the PGE2 binding site of rabbit gastric mucosa were determined by measuring [3H]PGE2 displacement from its high-affinity plasma membrane binding sites. In parallel, the potency of each prostaglandin in inhibiting acid generation in vitro was determined by measuring the inhibition of histamine-stimulated [14C]aminopyrine accumulation in rabbit parietal cells prepared by enzymatic dispersion and enriched by counterflow elutriation. All seven prostaglandins displaced [3H]PGE2 and inhibited histamine-stimulated [14C]aminopyrine accumulation in a concentration-dependent manner. For all tested prostaglandins, the IC50 values were in excellent agreement for both variables measured. It is concluded that (a) a PGE2 receptor is localized on the parietal cell and mediates inhibition of acid formation by all prostaglandins and (b) the different in vitro antisecretory potencies of prostaglandins can be attributed to their different affinities to this PGE2 receptor.
Article
We measured transmucosal potential difference (PD) of the stomach in anesthetized rats before, during, and after hemorrhagic shock, and investigated the effects of various drugs on the PD and gastric lesion during this period. After hemorrhagic shock, there was a decrease of PD and an increase of luminal pH in the saline-perfused stomach, the degree of these changes being dependent on a fall in the arterial blood pressure. The graded reduction of PD in response to hemorrhagic shock was similarly observed in the acid-perfused stomach as in the saline-perfused one. However, gastric lesions developed only in the former, and a significant correlation was found between the lesion index and the fall in blood pressure, the reduction in PD, or the concentration of HCl as the perfusate. Subcutaneously administered propantheline bromide (30 mg/kg) or cimetidine (100 mg/kg) had no effect on gastric lesion and PD reduction caused by hemorrhagic shock. These lesions were significantly inhibited by 16,16-dimethyl prostaglandin E2 (10 micrograms/kg) or sulindac (100 mg/kg), a scavenger of OH., and aggravated by indomethacin (1 mg/kg), with less effect on the PD reduction. Intravenous infusion of NaHCO3 (0.5 M) also significantly prevented the lesion with a concomitant suppression of the PD reduction in response to hemorrhagic shock, but these effects were significantly reversed by pretreatment of the animals with acetazolamide (50 mg/kg). These results indicate that during hemorrhagic shock the PD may largely reflect the impairment of mucosal blood flow and may be used as an indicator of mucosal vulnerability to acid, gastric lesions develop only in the presence of exogenous acid, and production of prostaglandins and superoxide radicals may be involved in the pathogenesis of gastric lesions.