ArticlePDF Available

Gastric damage and granulocyte infiltration induced by indomethacin in tumour necrosis factor receptor 1 (TNF-R1) or inducible nitric oxide synthase (iNOS) deficient mice

Authors:

Abstract and Figures

Tumour necrosis factor alpha (TNF-alpha) is involved in non-steroidal anti-inflammatory drug induced gastropathy. Nitric oxide (NO) is a mediator of gastrointestinal mucosal defence but, paradoxically, it also contributes to mucosal damage. We optimised the C57BL/6 mouse model of indomethacin induced gastropathy to evaluate the role of TNF-alpha and inducible nitric oxide synthase (iNOS) generated NO in gastric damage and granulocyte infiltration using tumour necrosis factor receptor 1 (TNF-R1-/-) or iNOS (iNOS-/-) deficient mice. Different doses of indomethacin (2.5, 5, 10, 20 mg/kg) were administered and animals were assessed 6, 12, or 24 hours later. Gastric damage was measured by the sum of all erosions in the gastric mucosa, and gastric granulocyte infiltration was determined by myeloperoxidase (MPO) activity. Other groups of wild-type mice received thalidomide, dexamethasone, fucoidin, L-NAME, or 1400W, and then indomethacin was administered. Additionally, indomethacin was administered to TNF-R1-/- or iNOS-/-. Gastric damage and MPO activity were evaluated 12 hours later. Indomethacin induced dose and time dependent gastric damage and increase in MPO activity in wild-type mice, with the greatest effect at a dose of 10 mg/kg and after 12 hours. Treatment with thalidomide, dexamethasone, or fucoidin reduced gastric damage and MPO activity induced by indomethacin. After indomethacin administration, TNF-R1-/- had less gastric damage and MPO activity than controls. Genetic (knockout mice) or pharmacological (1400W and L-NAME) inhibition of iNOS activity reduced indomethacin induced gastric damage, despite no reduction in MPO activity. TNF-alpha, acting via TNF-R1, is involved in indomethacin induced gastric damage and granulocyte infiltration. Furthermore, iNOS generated NO is involved in gastric damage induced by indomethacin.
Content may be subject to copyright.
STOMACH
Gastric damage and granulocyte infiltration induced by
indomethacin in tumour necrosis factor receptor 1 (TNF-R1)
or inducible nitric oxide synthase (iNOS) deficient mice
M H L P Souza, H Paula Lemos, R B Oliveira, F Q Cunha
...............................................................................................................................
See end of article for
authors’ affiliations
.......................
Correspondence to:
Dr M H L P Souza,
Departamento de
Fisiologia e Farmacologia,
Universidade Federal do
Ceara´, Rua Cel. Nunes de
Melo, 1127, Rodolfo
Teo´filo, Fortaleza-CE,
Brasil; souzamar@ufc.br
Accepted for publication
12 November 2003
.......................
Gut 2004;53:791–796. doi: 10.1136/gut.2002.012930
Background: Tumour necrosis factor a(TNF-a) is involved in non-steroidal anti-inflammatory drug induced
gastropathy. Nitric oxide (NO) is a mediator of gastrointestinal mucosal defence but, paradoxically, it also
contributes to mucosal damage.
Aims: We optimised the C57BL/6 mouse model of indomethacin induced gastropathy to evaluate the role
of TNF-aand inducible nitric oxide synthase (iNOS) generated NO in gastric damage and granulocyte
infiltration using tumour necrosis factor receptor 1 (TNF-R12/2) or iNOS (iNOS2/2) deficient mice.
Methods: Different doses of indomethacin (2.5, 5, 10, 20 mg/kg) were administered and animals were
assessed 6, 12, or 24 hours later. Gastric damage was measured by the sum of all erosions in the gastric
mucosa, and gastric granulocyte infiltration was determined by myeloperoxidase (MPO) activity. Other
groups of wild-type mice received thalidomide, dexamethasone, fucoidin, L-NAME, or 1400W, and then
indomethacin was administered. Additionally, indomethacin was administered to TNF-R12/2or
iNOS2/2. Gastric damage and MPO activity were evaluated 12 hours later.
Results: Indomethacin induced dose and time dependent gastric damage and increase in MPO activity in
wild-type mice, with the greatest effect at a dose of 10 mg/kg and after 12 hours. Treatment with
thalidomide, dexamethasone, or fucoidin reduced gastric damage and MPO activity induced by
indomethacin. After indomethacin administration, TNF-R12/2had less gastric damage and MPO activity
than controls. Genetic (knockout mice) or pharmacological (1400W and L-NAME) inhibition of iNOS
activity reduced indomethacin induced gastric damage, despite no reduction in MPO activity.
Conclusion: TNF-a, acting via TNF-R1, is involved in indomethacin induced gastric damage and
granulocyte infiltration. Furthermore, iNOS generated NO is involved in gastric damage induced by
indomethacin.
Non-steroidal anti-inflammatory drugs (NSAIDs) are
some of the most widely used drugs in the world.
1
NSAID induced gastric damage is the major side effect
associated with usage of this type of drug. Although the
mechanism of NSAID induced gastropathy is generally
believed to be related to the ability of these agents to inhibit
gastric prostaglandin generation,
1
there is a great deal of
evidence to suggest that neutrophils are important in the
pathogenesis of the gastric damage induced by NSAIDs.
226
It is becoming increasingly appreciated that tumour
necrosis factor a(TNF-a) plays a critical role in NSAID
induced gastric injury by modulating neutrophil infiltra-
tion.
45
The activities of TNF-aare mediated by two distinct
cell surface receptors: TNF receptor 1 (TNF-R1) and TNF
receptor 2 (TNF-R2).
7
Several studies have shown that TNF-R1
is the dominant signalling receptor for the inflammatory
effect of TNF-a.
7
However, the role of TNF-R1 in NSAID
induced gastric damage has not been studied.
Nitric oxide (NO) is a crucial mediator of gastrointestinal
mucosal defence but, paradoxically, it also contributes to
mucosal damage.
8
This can be illustrated by the ability of
different NO concentrations to produce completely opposite
effects in the same tissue.
9
In general, the neuronal and
endothelial nitric oxide synthase (NOS) isoforms produce
low amounts of NO.
9
In contrast, the inducible form of NOS
(iNOS) produces NO in higher quantities.
9
Piotrowski et al
showed that when animals received an ulcerogenic dose of
indomethacin, there was a 12-fold increase in gastric
epithelial expression of iNOS activity compared with controls,
and this increase was positively correlated with damage to
the epithelium.
10
On the other hand, Wallace and Miller
stated that NO plays a critical role in modulating several
components of mucosal defence, including increased gastric
blood flow, reduced neutrophil adhesion, and increased
mucus secretion.
9
However, the relationship between iNOS
and NSAID induced gastric damage has not been studied
extensively and requires further clarification.
The recent development of genetically engineered mice
with gene deletions (knockout) makes it theoretically
possible to identify the specific inflammatory mediators that
may be involved in NSAID induced gastropathy.
6
In the
present study, we optimised a C57BL/6 mouse model of
indomethacin induced gastropathy to evaluate the role of
TNF-aand iNOS generated NO in gastric damage and
granulocyte infiltration using TNF-R1 and iNOS deficient
mice.
MATERIAL AND METHODS
Animals
C57BL/6 mice (weight 18–20 g) were fasted for 18–24 hours
before the experiments. Breeding pairs of mice with a
targeted disruption of the TNF-areceptor R1 (p55) and
iNOS genes were obtained from Jackson Laboratories (Bar
Abbreviations: NSAIDs, non-steroidal anti-inflammatory drugs; NO,
nitric oxide; TNF-a, tumour necrosis factor a; TNF-R1, tumour necrosis
factor receptor 1; TNF-R2, tumour necrosis factor receptor 2; NOS, nitric
oxide synthase; iNOS, inducible nitric oxide synthase; MPO,
myeloperoxidase; TNF-R1 2/2, TNF-R1 deficient mice; iNOS 2/2,
iNOS deficient mice
791
www.gutjnl.com
Harbor, Maine, USA). Breeding stock backcrossed to C57BL/6
was obtained and the genotype of p55 and iNOS mice was
determined by polymerase chain reaction of DNA, as
previously described.
11
Animals were housed in a sterile
laminar flow cabinet until the start of the experiments, in
temperature controlled rooms, and received water and food
ad libitum. All animal treatments and surgical procedures
were performed in accordance with the Guide for Care and
Use of Laboratory Animals, National Institutes of Health
(Bethesda, Maryland, USA).
Drugs
Drugs used in this study were indomethacin (Prodome
Quı´mica e Farmaceˆutica, Sa˜o Paulo, Sa˜o Paulo, Brazil), Tris
(Merck, Sa˜o Paulo, Sa˜o Paulo, Brazil), 1400W (Cayman
Chemical Co, Ann Arbor, Michigan, USA), L-NAME, thali-
domide, dexamethasone, fucoidin, 3,39, 5,59’-tetramethyl-
benzidine and hexadecyltrimethyl-ammonium bromide (all
from Sigma Chemicals, St Louis, Missouri, USA).
Gastric damage induce by indomethacin in mice
Gastric damage was induced in wild-type C57BL/6 mice by
intragastric installation of indomethacin (2.5, 5, 10, 20 mg/
kg) dissolved in Tris buffer. The control group received only
vehicle (Tris buffer). Animals were killed 6, 12 or 24 hours
later by decapitation after light anaesthesia. Other groups of
wild-type mice were treated with fucoidin, a sulphated
fucosylated polysaccharide that binds to and blocks the
function of L- and P-selectins
12 13
(two doses of 25 mg/kg, five
minutes before and six hours after, intravenously) or saline,
and then indomethacin was administered (10 mg/kg).
Twelve hours later, animals were sacrificed and their
stomachs rapidly removed, opened by an incision along the
greater curvature, and pinned onto a wax platform.
Haemorrhagic or ulcerative lesions were counted and their
length measured with analogue callipers. A gastric damage
score (lesion index) was then calculated as the sum of the
lengths of all linear erosions,
14
which was measured by an
observer (HPL) who was unaware of the treatment given to
the animals. After scoring the damage, a sample of the corpus
region of each stomach was excised for measurement of
myeloperoxidase (MPO) activity.
15
MPO is an enzyme found
primarily in the azurophilic granules of neutrophils and
therefore has been used extensively as a biochemical marker
of granulocyte infiltration into various tissues, including the
gastrointestinal tract.
Role of TNF-ain indomethacin induced gastric
damage in mice
Gastric damage was induced in TNF-R1 deficient mice (TNF-
R12/2) or wild-type C57BL/6 mice by intragastric installa-
tion of indomethacin (10 mg/kg). Other groups of wild-type
mice were treated with the TNF-asynthesis inhibitor
thalidomide
16
(50 mg/kg, one hour before, and 25 mg/kg,
six h after, intraperitoneally), or with the glucocorticoid
dexamethasone (two doses of 1 mg/kg, one hour before and
six hours after, intraperitoneally). One hour later, indo-
methacin was administered (10 mg/kg). The control group
received only vehicle. After a further 12 hours, gastric
damage was determined as described above. Thereafter, full
0
5
10
15
20
Lesion index (mm)
0
4
6
2
MPO activity (U/mg)
2.5C5102.5C 5 10 20
Indomethacin (mg/kg) Indomethacin (mg/kg)
AB
*
*
*
0
5
10
15
Lesion index (mm)
0
4
6
2
MPO activity (U/mg)
6C12246C1224
Time after indomethacin (h) Time after indomethacin (h)
CD
*
*
*
*
*
Figure 1 Intragastric instillation of indomethacin induced both gastric damage and an increase in gastric myeloperoxidase (MPO) activity in C57BL/6
mice. Animals were treated with indomethacin (2.5–20 mg/kg), and gastric damage (A) and gastric MPO activity (B) were determined 12 hours later.
The control (C) group received only Tris buffer. Gastric lesions and gastric MPO activity were greatest at a dose of 10 mg/kg. The time course of
indomethacin induced gastropathy showed that both gastric damage (C) and gastric MPO activity (D) peaked at 12 hours. Results are expressed as
means (SEM) of at least five animals per group. *p,0.05 compared with the control group (ANOVA and Newman-Keuls test).
792 Souza, Paula Lemos, Oliveira, et al
www.gutjnl.com
thickness pieces of the gastric corpus were weighed, frozen,
and stored at 270˚
C until assay of MPO activity.
Role of iNOS derived NO in indomethacin induced
gastric damage in mice
Gastric damage was induced in iNOS deficient (iNOS2/2)or
wild-type C57BL/6 mice by intragastric instillation of
indomethacin (10 mg/kg). Other groups of wild-type or
iNOS2/2mice were treated with L-NAME (100 mg/kg, one
hour before, and 50 mg/kg, six hours after indomethacin
administration, intraperitoneally), and then indomethacin
was administered (10 mg/kg). Another group of wild type
mice was treated with a selective inhibitor of inducible NOS,
1400W
17
(1.5 mg/kg, one hour before, intravenously), and
then mice received indomethacin (10 mg/kg). The control
group received only vehicle. Twelve hours later, gastric
damage was determined as described above. Finally, full
thickness pieces of the gastric corpus were weighed, frozen,
and stored at 270˚
C until assay of MPO activity.
Gastric MPO activity
The extent of granulocyte accumulation in the gastric mucosa
was measured by assaying MPO activity, as previously
described.
15
Briefly, 50–100 mg of gastric tissue were homo-
genised in two volumes of ice cold buffer (0.1 M NaCl,
20 mM NaPO
4
, 15 mM Na EDTA), pH 4.7, and centrifuged at
3000 rpm for 15 minutes. The pellet was then subjected to
hypotonic lysis (900 ml of 0.2% NaCl solution followed
30 seconds later by addition of an equal volume of a solution
containing 1.6% NaCl and 5% glucose). After further
centrifugation, the pellet was resuspended in 50 mM
NaPO
4
buffer, pH 5.4, containing 0.5% H-TAB, and re-
homogenised. The homogenate was then frozen and thawed
three times and centrifuged again at 10 000 rpm for
15 minutes at 4˚
C. MPO activity in the resuspended pellet
was assayed by measuring the change in absorbance at
450 nm using tetramethylbenzidine (1.6 mM) and H
2
O
2
(0.5 mM). Firstly, the results were reported as total number
of neutrophils by comparing absorbance of the tissue
supernatant with that of rat peritoneal neutrophils processed
in the same way. To this end, neutrophil migration was
induced in the peritoneum of rats by injecting carrageenin
(300 mg/animal). A standard curve relating neutrophil
numbers (.90% purity, 12 500 to 195.3 neutrophils/50 ml)
and absorbance was obtained by processing purified neu-
trophils, as described above, and assaying for MPO activity.
The correlation between the number of neutrophils and units
of MPO was determined using the technique described by
Bradley and colleagues.
18
The neutrophil standard curve was
processed using 0.0005% hydrogen peroxide as substrate for
MPO. A unit of MPO activity was defined as that converting
1mmol of hydrogen peroxide to water in one minute at 22˚
C.
Statistical analysis
Statistical analysis was performed using one way analysis of
variance (ANOVA) followed by the Newman-Keuls test,
when appropriate. Statistical significance was set at p,0.05.
RESULTS
Intragastric administration of indomethacin induced linear
haemorrhagic erosions in the corpus of the animals’
stomachs. As shown in fig 1, indomethacin induced gastric
lesions and increased gastric MPO activity dose dependently,
with the greatest effect at a dose of 10 mg/kg (fig 1A, B). The
time course of both gastric damage and the increase in gastric
MPO activity induced by indomethacin, at a dose of 10 mg/
kg, peaked at 12 hours (fig 1C, D).
Gastric damage (fig 2A) and gastric MPO activity (fig 2B)
induced by indomethacin (10 mg/kg) were significantly
reduced by treatment with fucoidin. Figure 3 shows that
thalidomide or dexamethasone treatment significantly inhib-
ited both the gastric damage (fig 3A) and gastric MPO
activity (fig 3B) induced by indomethacin. Furthermore,
TNF-R1 deficient mice presented less gastric damage (fig 3C)
and gastric MPO activity (fig 3D) than wild-type mice.
Treatment with L-NAME or 1400W reduced the gastric
damage (fig 4A) induced by indomethacin. Gastric MPO
activity induced by indomethacin was increased with
L-NAME treatment and unchanged with 1400W treatment
(fig 4B). In addition, after indomethacin administration,
iNOS deficient mice had less severe gastric damage and
similar indomethacin induced gastric MPO activity compared
with wild-type mice (fig 4C, D).
Table 1 shows that L-NAME treatment did not change
indomethacin induced gastric damage in iNOS deficient mice
but increased gastric MPO activity compared with iNOS
deficient mice.
DISCUSSION
The recent development of genetically engineered mice
makes this species particularly attractive as an animal model
to define the pathogenic mechanism responsible for NSAID
induced gastric damage.
6
In the present study, we optimised
a mouse model of indomethacin induced gastropathy and
used TNF-R1 or iNOS deficient mice to evaluate the role of
TNF-aand iNOS derived NO in the gastric damage and
granulocyte infiltration induced by indomethacin. This last
event was analysed because indomethacin induces gastric
0
5
10
15
Lesion index (mm)
*
SalineC Fucoidin
Indomethacin (10 mg/kg)
0
2
4
6
8
MPO activity (U/mg)
*
SalineC Fucoidin
Indomethacin (10 mg/kg)
A
B
Figure 2 Gastric damage and gastric myeloperoxidase (MPO) activity
induced by indomethacin were significantly reduced by treatment with
fucoidin. Wild-type mice were treated with saline or fucoidin. Five
minutes later, indomethacin (10 mg/kg) was administered. Gastric
lesions (A) and gastric MPO activity (B) were determined 12 hours later.
The control (C) group received only Tris buffer. Results are expressed as
means (SEM) of at least five animals per group. *p,0.05 compared with
the saline group (ANOVA and Newman-Keuls test).
Indomethacin induced gastric damage in TNF-R1 or iNOS deficient mice 793
www.gutjnl.com
damage is a neutrophil dependent process.
2–6
Our data
showed that TNF-a, acting via TNF-R1, mediates gastric
damage and granulocyte infiltration, measured as MPO
activity, in indomethacin induced gastropathy.
Furthermore, iNOS generated NO is involved in gastric
damage induced by indomethacin.
Indomethacin induced dose and time dependent gastric
erosions and an increase in gastric MPO activity in mice, with
the greatest effect 12 hours after administration. Gastric
damage observed was associated with increased MPO activity
because the severity and time course of the gastric damage
were coincident with such infiltration. Moreover, treatment
with fucoidin, a sulphated fucosylated polysaccharide that
binds to and blocks the function of L- and P-selectins,
12 13
reduced both gastric MPO activity and gastric damage
induced by indomethacin. Similarly, other studies have
shown that NSAID induced gastric damage in rats depends
on neutrophil mucosal infiltration.
25
Furthermore, treatment
with anti- ICAM-1, P-selectin, and E-selectin attenuated the
severity of indomethacin induced gastric in rats,
19
and mice
deficient in L- or P-selectins developed less indomethacin
induced gastric damage.
6
We observed that TNF-a, acting via TNF-R1, is an
important mediator in NSAID induced gastric damage and
granulocyte infiltration as both events were reduced in
TNF-R12/2mice. The involvement of TNF-ain this event
was further supported by the observation that thalidomide, a
drug that inhibits TNF-aproduction,
16
and also dexametha-
sone, reduced both gastric MPO activity and gastric damage
induced by indomethacin. These results are in agreement
with previous studies that showed a reduction in gastric
susceptibility to indomethacin in rats treated with pentoxifyl-
line, thalidomide, or dexamethasone.
45
With regard to the
TNF-R1 receptor, there is much evidence that it is the main
receptor that mediates the inflammatory effect of TNF-a,
being responsible for neutrophil migration and activation,
including NO production.
7
Furthermore, nuclear factor kb,an
important transcription factor for inflammatory events,
including gastrointestinal inflammatory diseases,
20
is acti-
vated by TNF-avia the TNF-R1 receptor.
7
The glucocorticoid
dexamethasone, apart from inhibiting TNF-aproduction,
also inhibits release of the majority of other inflammatory
cytokines, such as interleukin1band chemokines,
21
and
lipoxygenase products,
21
which are mediators thought to be
involved in the gastrointestinal inflammatory process.
22
Our
results do not eliminate the possibility that these molecules
may also mediate indomethacin induced gastropathy in mice.
In spite of the many studies showing involvement of NO in
the gastric damage induced by chemical substances, such as
serotonin or compound 48/80,
23 24
by stress,
25
or by Helicobacter
pylori infection,
26
the role of iNOS derived NO in NSAID
induced gastric damage has not been completely elucidated.
There is evidence that low doses of NO releasing substances
protect against NSAID induced gastropathy
27
and increase the
healing rate of gastric ulcers.
28
However, high doses of these
substances could induce extensive haemorrhagic mucosal
damage.
29 30
Furthermore, there are studies showing that
L-NAME, a non-specific NOS inhibitor, at doses of 15–50 mg/kg,
0
5
10
15
Lesion index (mm)
0
4
6
2
MPO activity (U/mg)
SalineC Thalid DexaSalineC Thalid Dexa
Indomethacin (10 mg/kg) Indomethacin (10 mg/kg)
AB
*
*
*
*
0
10
20
30
40
Lesion index (mm)
0.0
1.0
1.5
0.5
MPO activity (U/mg)
WildC TNF-RI _/_
WildC TNF-RI _/_
Indomethacin (10 mg/kg) Indomethacin (10 mg/kg)
CD
*
*
Figure 3 Tumour necrosis factor a(TNF-a) mediates indomethacin induced gastric damage and an increase in gastric myeloperoxidase (MPO)
activity. Gastric damage (A) and gastric MPO activity (B) induced by indomethacin (10 mg/kg) were significantly reduced by treatment with
thalidomide (Thalid) or dexamethasone (Dexa), but not with saline. In addition, intragastric administration of indomethacin induced less gastric
damage (C) and gastric MPO activity (D) in tumour necrosis factor receptor 1 deficient mice (TNF-R12/2) than in wild-type mice (Wild). Results are
expressed as means (SEM) of at least five animals per group. *p,0.05 compared with the saline group (A, B) or wild-type mice (C, D) (ANOVA and
Newman-Keuls test).
794 Souza, Paula Lemos, Oliveira, et al
www.gutjnl.com
increases indomethacin induced gastric damage,
31
reduces
gastric blood flow,
32
and delays healing of chronic gastric
ulcers.
33
Our results showed that iNOS generated NO is
implicated in indomethacin induced gastric damage as iNOS
deficient mice exhibited less gastric damage when indo-
methacin was administered, and wild-type mice treated with
L-NAME at a dose of 100 mg/kg, which inhibits both cNOS
and iNOS activity,
34
or with 1400W, a selective inhibitor of
iNOS,
17
decreased indomethacin induced gastric damage. The
fact that both genetic (knockout mice) and pharmacological
(1400W) specific inhibition of iNOS reduced gastric damage
indicates that the effect of L-NAME on indomethacin induced
gastric damage was also determined by its effect on iNOS
activity, and not by causing a decrease in gastric blood flow,
which—due its inhibitory effect on cNOS—could, in turn,
render less conspicuous the haemorrhagic expression of the
gastric damage. Confirmation that the L-NAME effect was
due to inhibition of iNOS was demonstration that L-NAME
treatment did not enhance reduction of gastric damage
caused by indomethacin in iNOS deficient mice (table 1).
These results are in apparent contradiction with others
described in the literature which showed an increase in
indomethacin induced gastric damage by a non-specific NOS
inhibitor, L-NAME, at a dose of 50 mg/kg.
31
One possible
explanation for this discrepancy is that at this dose, L-NAME
is unable to efficiently inhibit iNOS activity.
With regard to the role of NO in granulocyte infiltration
induced by indomethacin, our results suggest that
small amounts of NO released by cNOS are sufficient to
downregulate indomethacin induced gastric granulocyte
0
10
20
30
Lesion index (mm)
0
4
6
8
2
0
4
6
8
2
MPO activity (U/mg)
SalineC L-NAME 1400WSalineC L-NAME 1400W
Indomethacin (10 mg/kg) Indomethacin (10 mg/kg)
AB
*
*
*
0
10
20
30
40
Lesion index (mm)
MPO activity (U/mg)
WildC iNOS _/_
WildC iNOS _/_
Indomethacin (10 mg/kg) Indomethacin (10 mg/kg)
CD
*
Figure 4 Involvement of inducible nitric oxide synthase (iNOS) generated nitric oxide in the gastric damage and increase in gastric myeloperoxidase
(MPO) activity by indomethacin. Treatment with L-NAME or 1400W reduced gastric damage (A) but promoted different effects on the indomethacin
induced increase in gastric MPO activity. L-NAME increased and 1400W did not change gastric MPO activity induced by indomethacin (B).
Furthermore, after indomethacin administration, iNOS deficient mice had less gastric damage than wild-type mice (Wild) (C), despite no change in
indomethacin induced gastric MPO activity (D). Results are expressed as means (SEM) of at least five animals per group. *p,0.05 compared with the
saline group (A, B) or wild-type mice (C, D) (ANOVA and Newman-Keuls test).
Table 1 Effect of L-NAME treatment in indomethacin induced gastropathy in iNOS
deficient mice
Group Lesion index*(mm) MPO activity*(U/mg)
Vehicle (Tris buffer) 0.0 (0.0) 0.64 (0.06)
Indomethacin (wild-type mice) 23.1 (3.7) 5.36 (0.26)
Indomethacin+saline (iNOS2/2) 12.5 (2.6) 6.46 (1.02)
Indomethacin+L-NAME (iNOS2/2) 9.3 (0.7)``` 8.52 (0.74)```
*Data are mean (SEM) of 4–6 mice per group.
iNOS, inducible nitric oxide synthase; MPO, myeloperoxidase.
p,0.01, indomethacin+saline (iNOS2/2) versus indomethacin (wild-type mice).
```p,0.001, indomethacin+L-NAME (iNOS2/2) versus indomethacin (wild-type mice).
Indomethacin induced gastric damage in TNF-R1 or iNOS deficient mice 795
www.gutjnl.com
infiltration. This interpretation is supported by the fact that
we observed enhancement of gastric MPO activity only when
both NOS isofoms (cNOS and iNOS) were inhibited by
L-NAME in both wild-type and iNOS2/2mice. In line with
our results, there is evidence in the literature that NO inhibits
expression of cell adhesion molecules on endothelial cells,
which is an important step in neutrophil migration.
35–37
The fact that the reduction in indomethacin induced
gastric lesions observed in iNOS2/2,inL-NAME or 1400W
treated mice, was not accompanied by a similar reduction in
granulocyte infiltration appears to contradict the hypothesis
that NSAID induced gastric damage is a neutrophil depen-
dent process. However, one possible explanation is that
although neutrophils, which could be the source of the NO
involved in gastric damage, are present in the gastric mucosa,
iNOS in these cells is not able to produce NO and/or other
reactive nitrogen species.
In summary, our results indicate that TNF-a, acting via
TNF-R1, is involved in indomethacin induced gastric damage
and neutrophil infiltration in mice. Furthermore, iNOS
generated NO is involved in gastric damage induced by
indomethacin.
ACKNOWLEDGEMENTS
The authors gratefully acknowledge the technical assistance of Ieda R
dos Santos Schivo. Grants from FAPESP, PRONEX, and CNPq
(Brazil) supported this work.
Authors’ affiliations
.....................
M H L P Souza, Department of Physiology and Pharmacology, School of
Medicine, Federal University of Ceara´, Brazil, and Department of
Medicine (Division of Gastroenterology), School of Medicine Ribeira˜o
Preto, University of Sa˜o Paulo, Brazil
H Paula Lemos, Department of Medicine (Division of Gastroenterology),
School of Medicine Ribeira˜o Preto, University of Sa˜o Paulo, Brazil, and
Department of Pharmacology, School of Medicine Ribeira˜o Preto,
University of Sa˜o Paulo, Brazil
R B Oliveira, Department of Medicine (Division of Gastroenterology),
School of Medicine Ribeira˜o Preto, University of Sa˜o Paulo, Brazil
F Q Cunha, Department of Pharmacology, School of Medicine Ribeira˜o
Preto, University of Sa˜o Paulo, Brazil
REFERENCES
1Laine L. Approaches to nonsteroidal anti-inflammatory drug use in the high-
risk patient. Gastroenterology 2001;120:594–606.
2Wallace JL, Keenan CM, Granger DN. Gastric ulceration induced by
nonsteroidal anti-inflammatory drugs is a neutrophil-dependent process.
Am J Physiol 1990;259:G462–7.
3Trevethick MA, Clayton NM, Strong P, et al. Do infiltrating neutrophils
contribute to the pathogenesis of indomethacin induced ulceration of the rat
gastric antrum?Gut 1993;34:156–60.
4Santucci L, Fiorucci S, Di Matteo FM, et al. Role of tumor necrosis factor alpha
release and leukocyte margination in indomethacin-induced gastric injury in
rats. Gastroenterology 1995;1108:393–401.
5Appleyard CB, McCafferty DM, Tigley AW, et al. Tumor necrosis factor
mediation of NSAID-induced gastric damage: role of leukocyte adherence.
Am J Physiol 1996;270:G42–8.
6Morise Z, Granger DN, Fuseler JW, et al. Indomethacin induced gastropathy
in CD18, intercellular adhesion molecule 1, or P-selectin deficient mice. Gut
1999;45:523–8.
7Tartaglia LA, Goeddel DV. Two TNF receptors. Immunol Today
1992;13:151–3.
8Muscara MN, Wallace JL. Nitric oxide. V. therapeutic potential of nitric oxide
donors and inhibitors. Am J Physiol 1999;276:G1313–16.
9Wallace JL, Miller MJ. Nitric oxide in mucosal defense: a little goes a long
way. Gastroenterology 2000;119:512–20.
10 Piotrowski J, Slomiany A, Slomiany BL. Activation of apoptotic caspase-3 and
nitric oxide synthase-2 in gastric mucosal injury induced by indomethacin.
Scand J Gastroenterol 1999;34:129–34.
11 Pfeffer K, Matsuyama T, Kundig TM, et al. Mice deficient for the 55 kd tumor
necrosis factor receptor are resistant to endotoxic shock, yet succumb to L
monocytogenes infection. Cell 1993;73:457–67.
12 Varki A. Selectin ligands. Proc Natl Acad Sci U S A 1994;91:7390–7.
13 Bevilacqua MP. Endothelial-leukocyte adhesion molecules. Annu Rev Immunol
1993;11:767–804.
14 Santucci L, Fiorucci S, Giansanti M, et al. Pentoxifylline prevents
indomethacin-induced acute gastric-mucosal damage in rats—role of tumor-
necrosis-factor-alpha. Gut 1994;35:909–15.
15 Souza DG, Cassali GD, Poole S, et al. Effects of inhibition of PDE4 and TNF-a
on local and remote injuries following ischaemia and reperfusion injury.
Br J Pharmacol 2001;134:985–94.
16 Sampaio EP, Sarno EN, Galilly R, et al. Thalidomide selectively inhibits tumor
necrosis factor alpha production by stimulated human monocytes. J Exp Med
1991;173:699–703.
17 Garvey EP, Oplinger JA, Furfine ES, et al. 1400W is a slow, tight binding,
and highly selective inhibitor of inducible nitric-oxide synthase in vitro and in
vivo. J Biol Chem 1997;272:4959–63.
18 Bradley PP, Christensen RD, Rothstein G. Cellular and extracellular
myeloperoxidase in pyogenic inflammation. Blood 1982;3:618–22.
19 Wallace JL, McKnight W, Miyasaka M, et al. Role of endothelial adhesion
molecules in NSAID-induced gastric mucosal injury. Am J Physiol
1993;265:G993–8.
20 Schmid RM, Adler G. NF-kappaB/rel/IkappaB: implications in
gastrointestinal diseases. Gastroenterology 2000;118:1208–28.
21 Haynes RC. Adrenocorticotropic hormone; adrenocortical steroids and their
synthetic analogs; inhibitors of the synthesis and actions of adrenocortical
hormones. In: Gilman AG, Rall TW, Nies AS, et al, eds. The pharmacological
basis of therapeutics, 8th edn. New York: Pergamon Press, 1990:1431–62.
22 Wallace JL, Ma L. Inflammatory mediators in gastrointestinal defense and
injury. Exp Biol Med (Maywood) 2001;226:1003–15.
23 Yasuhiro T, Korolkiewicz RP, Kato S, et al. Role of nitric oxide in pathogenesis
of serotonin-induced gastric lesions in rats. Pharmacol Res 1997;36:333–8.
24 Yasuhiro T, Konaka A, Kato S, et al. Gastric mucosal damage induced by
compound 48/80: roles of serotonin and nitric oxide. J Gastroenterol Hepatol
1998;13:1099–106.
25 Nishida K, Ohta Y, Ishiguro I. Relation of inducible nitric oxide synthase
activity to lipid peroxidation and nonprotein sulfhydryl oxidation in the
development of stress-induced gastric mucosal lesions in rats. Nitric Oxide
1998;2:215–23.
26 Mannick EE, Bravo LE, Zarama G, et al. Inducible nitric oxide synthase,
nitrotyrosine, and apoptosis in Helicobacter pylori gastritis: effect of antibiot ics
and antioxidants. Cancer Res 1996;56:3238–43.
27 Calatayud S, Sanz MJ, Canet A, et al. Mechanisms of gastroprotection by
transdermal nitroglycerin in the rat. Br J Pharmacol 1999;127:1111–18.
28 Elliott SN, McKnight W, Cirino G, et al. A nitric oxide-releasing nonsteroidal
anti-inflammatory drug accelerates gastric ulcer healing in rats.
Gastroenterology 1995;109:524–30.
29 Lamarque D, Whittle BJ. Role of oxygen-derived metabolites in the rat gastric
mucosal injury induced by nitric oxide donors. Eur J Pharmacol
1995;277:187–94.
30 Gurbuz V, Alican I, Berrak, et al. Role of nitric oxide in indomethacin-induced
gastric mucosal dysfunction in the rat. Exp Physiol 1999;84:319–32.
31 Khattab MM, Gad MZ, Abdallah D. Protective role of nitric oxide in
indomethacin-induced gastric ulceration by a mechanism independent of
gastric acid secretion. Pharmacol Res 2001;43:463–7.
32 Ferraz JG, Tigley A, Wallace JL. Paradoxical effects of L-arginine on gastric
mucosal integrity. Eur J Pharmacol 1994;260:107–11.
33 Ma L, Wallace JL. Endothelial nitric oxide synthase modulates gastric ulcer
healing in rats. Am J Physiol Gastrointest Liver Physiol 2000;279:G341–6.
34 Ferraz JG, Sharkey KA, Reuter BK, et al. Induction of cyclooxygenase 1 and 2
in the rat stomach during endotoxemia: role in resistance to damage.
Gastroenterology 1997;113:195–204.
35 Fox-Robichaud A, Payne D, Kubes P. Inhaled NO reaches distal vasculatures
to inhibit endothelium- but not leukocyte-dependent cell adhesion. Am J Physiol
1999;277:L1224–31.
36 Kubes P, Suzuki M, Granger DN. Nitric oxide: an endogenous modulator of
leukocyte adhesion. Proc Natl Acad Sci U S A 1991;88:4651–5.
37 Benjamim CF, Silva JS, Fortes ZB, et al. Inhibition of leukocyte rolling by nitric
oxide during sepsis leads to reduced migration of active microbicidal
neutrophils. Infect Immun 2002;70:3602–10.
796 Souza, Paula Lemos, Oliveira, et al
www.gutjnl.com
... Results revealed that there was a time dependent increase in the extent and severity of ulceration and neutrophil infiltration into the gastric antrum after treatment with indomethacin. 26 Flavocoxid containing catechin from Acacia catechu attenuates inflammation and neutrophil invasion in a carrageenan-induced paw edema model. 27 Number of inflammatory cells in subgroup B 2 was significantly greater than subgroup C1.Similarly subgroup D 1 showed significantly decrease in inflammatory cells as compared to subgroup C 1 . ...
... Inflammation and neutrophil infiltration are vital in causing gastric damage induced by NSAIDs. 26 The inflammation results in increased TNF-α production, which enhances neutrophil-derived superoxide generation. 26 This stimulates IL-1 production resulting in neutrophil accumulation. ...
... 26 The inflammation results in increased TNF-α production, which enhances neutrophil-derived superoxide generation. 26 This stimulates IL-1 production resulting in neutrophil accumulation. 29 Inhibition of inflammation by Acacia catechu can be attributed to quercetin. ...
Article
Full-text available
Background: Aspirin is amongst the most widely used drugs and has many adverse effects on gastric mucosa. Anti-inflammatory properties of Acacia catechu have been established already. Objective was to evaluate the histopathological changes induced by aspirin in the stomach of albino rats and to assess the protective effect of different doses of Acacia catechu.Methods: Experimental study Postgraduate Medical Institute, Lahore for 21 days. Forty-eight adult albino rats, both males and female, were divided into four groups A, B, C and D randomly; each comprising of 12 rats. Group A was control, group B was given aspirin 100 mg/kg and group C and D were given aspirin 100 mg/kg along with Acacia catechu 250 mg/kg and 500 mg/kg respectively by oral route. The rats from individual group were sacrificed on 3rd day, 7th day and 14th day and stomachs were examined under light microscope to observe the inflammatory cells infiltration.Results: Gross and microscopic findings on days 3, 7 and 14 were similar. Control groups A1, A2 and A3 showed normal healthy gastric mucosa and the least number of inflammatory cells. In group B, aspirin produced ulcerations and linear breaks; with highest inflammatory infiltrates. On microscopic examination, numerous inflammatory cells were noted. Group C and D rats had minimum ulcer index and fewer inflammatory cells.Conclusions: Acacia catechu has protective role against gastric injury by inhibiting inflammation.
... Additionally, histopathological studies have shown that oral intake of Indian Gooseberry reduces gastric lesions and mucosal injury in vivo. Previous reports have also shown the fruit extract to decrease Myeloperoxidase (MPO) activity which is generally considered as a potent ulcerogenic marker [17] . Pepticare, a herbal formulation containing extracts of Phyllanthus emblica has proven its anti-oxidant mediated antiulcer effects in mammalian models [18] . ...
... Low levels of Nitric Oxide (NO) are maintained by eNOS. 17 The extract of Phyllanthus emblica has been documented to attenuate ulcer lesions by upregulating eNOS and downregulating iNOS mediated expression of NO. Gallic acid enriched Amla extract has been demonstrated to exhibit ulcer healing effects via cyclooxygenease dependent increase in PG E2, proangiogenesis factors and endothelial NOS. ...
Article
Full-text available
The Indian Gooseberry is one of the commonly used plants in the Indian system of medicine. Amla is a wonder superfood, belonging to the genus Phyllanthus L. which is mainly distributed in tropical areas. It represents a phytochemical reservoir of biologically important molecules. The plant contains tannins, alkaloids, amino acids, carbohydrates, vitamins and organic acids. Various parts of the plant have been used to treat a wide array of diseases. The present article highlights the importance of Phyllanthus emblica in the prevention and treatment of ulcer. Gastrointestinal ulcer results due to an increase in the offensive factors as compared to defensive ulcer protective elements. The fruit extracts possesses potent anti-oxidant potential which is the key to its therapeutic effect. Additionally it is also capable of inducing neo-angiogenesis thereby helping in repair of gastric lesions. The anti-inflammatory potential of the above further accelerates ulcer healing. Owing to its anti-secretory and cyto-protective capacities, Phyllanthus emblica either alone or in combination represents a valuable natural strategy to treat several chronic diseases especially ulcer.
... It was observed that a rise in the neutrophilic infiltration succeeded severe and intense ulceration into the gastric antrum leading to ulceration. 22 Flavocoxid present in Acacia catechu prevents pro inflammatory cytokines formationsuch as tumor necrosis factor-α, hence forth hindering reactive oxidative species synthesis resulting in reduced leukocyte infiltration. 23 Flavocoxid contains catechin which reduces neutrophil invasion, thus inflammation. ...
Article
Full-text available
Background and Objectives: Aspirin, one of the most widely used drugs, causes deleterious effects on gastric mucosa. Anti-inflammatory properties of Acacia catechu have already been established. This study is unique as it evaluated the histopathological changes induced by aspirin in the stomach of albino rats and assessed the protective effect of different doses of Acacia catechu. Methods: Forty eight adult albino rats, both males and female, were divided randomly into four groups A, B, C and D; each comprising of 12 rats. Group A, (control) was given chow and water ad libitum. Group B was treated with aspirin 100 mg/kg. Group C and D were given aspirin 100 mg/kg along with Acacia catechu 250 mg/kg 500 mg/kg respectively by oral route. Half of the rats from individual group were sacrificed on 3 rd day and the rest on 7 th day. Stomach was examined for macroscopic (ulcer index) and microscopic (inflammatory cells) parameters. Results: Gross and microscopic findings on days 3 and 7 were similar. Control groups A1 and A2 showed normal healthy gastric mucosa and the least number of inflammatory cells. In group B, aspirin produced ulcerations and linear breaks; with highest ulcer index. On microscopic examination, numerous inflammatory cells were noted. Group C and D rats had minimum ulcer index and fewer inflammatory cells. Conclusion: Aqueous solution of Acacia catechu has protective role against gastric ulcers by decreasing ulcers, and inflammation.
... Previous studies reported substantial elevation in TNF-α levels when exogenous ADMA was used to treat gastric epithelial cells [38]. TNF-α is a proinflammatory mediator that was previously reported to be highly expressed in indomethacin-induced gastric injury [44]. It promotes inflammation that induces tissue injury via upregulation of adhering molecules, resulting in leukocyte recruitment. ...
Article
Full-text available
Non-steroidal anti-inflammatory drugs (NSAIDs)-induced gastric ulcers represent a significant clinical concern and adversely affect the quality of life. Inducible nitric oxide synthase/endothelial nitric oxide synthase (iNOS/eNOS) and asymmetric dimethylarginine/ dimethylarginine dimethylaminohydrolase-1 (ADMA/DDAH-1) signaling are key players in gastric ulcer pathogenesis. This work was planned to explore the role of iNOS/eNOS and ADMA/DDAH-1 signaling in rats with indomethacin-induced gastric ulcer, as potential pathways for the gastro-protective effect of tadalafil. Split into 5 separate groups, rats were assigned to control, tadalafil (10 mg/kg, p.o), indomethacin (single oral dose of 60 mg/kg), indomethacin + pantoprazole (40 mg/kg, p.o), and indomethacin + tadalafil (10 mg/kg, p.o). The results indicated that pretreatment with tadalafil significantly reduced ulcer index (UI), increased preventive index (PI), and counteracted indomethacin-induced histopathological aberrations. Tadalafil significantly reduced the gastric content of NO while it significantly elevated that of GSH and enhanced SOD activity. It significantly reduced the gastric expression of TNF-α and ADMA while it significantly elevated that of COX-2, PGE-2, and DDAH-1. Western blot analysis revealed that pretreatment with tadalafil significantly reduced iNOS protein expression while it significantly elevated that of eNOS. Collectively, these data suggest that tadalafil exerts potential protective effect against indomethacin-induced ulcer through suppression of inflammation, attenuation of oxidative stress, and boosting of antioxidants. Moreover, tadalafil protective effects are mediated via upregulation of PGE-2 with modulating the signaling pathways of ADMA/DDAH-1, and iNOS/eNOS. As a result, the current evidence corroborates the use of tadalafil in controlling gastric ulcers and preventing NSAID gastric side effects.
... In the present study, we found that MPO activity in the gastric mucosa in the indomethacintreated rats was increase when compared to control treated rats. Our results were in agreement with Souza et al. (41) and this increase in MPO activity is reversed by methotrexate treatment. Our results were in agreement with Alican et al. (26) The results of the present study indicate that were in agreement with Sugimoto et al. (16) Xanthine oxidase has been known as a major source of ROS generation in the pathogenesis of various biological systems including gastrointestinal tract. ...
Article
Full-text available
Introduction: Aspirin, indomethacin, ibuprofen or diclofenac, a representative of non-steroidal anti-inflammatory drugs (NSAIDs) are widely used for treatment of inflammation. NSAIDs cause clinically important gastric damage by several mechanisms. One of them is to inhibition of endogenous prostaglandin synthesis. The anti-inflammatory properties of NSAIDs are mediated through the inhibition of cyclooxy- genase-2, whereas adverse effects, such as gastro-duodenal ulceration, occur as a result of effects on the constitutively expressed cyclooxygenase-1. In recent years, it has become increasingly clear that neutrophile-derived factors play an important role in several forms of experimental gastro-intestinal ulceration and inflammation. Aim of study: The aim of this study was to determine if the gastric ulcer induced by NSAIDs is mediated through oxidative stress. Also to examine the role of neutropenic drugs in the protection against ulcer induced by NSAIDs and examine if the antioxidant play a protective role in such cases. Subjects and methods: This study was carried out on 60 adult male Albino rats. They were divided into the six groups: Control group (n=6), indomethacin-treated group (n=6), neutropenic (methotrexate) group (n=12), zymosan-treated group (n=12), allopurinol-treated group (n=12) and vitamin E-treated group (n=12). The stomachs were examined macroscopically for hemorrhagic lesions developing in the glandular mucosa and the ulcer index was calculated. Tissue reduced glutathione content, myeloperoxidase activity, xanthine oxidase activity, thiobarbituric acid reactive substances and protein oxidation were measured. Results: There are a significantly increase in ulcer index, tissue xanthine oxidase, myeloperoxidase, lipid peroxidation and protein oxidation in indomethacin-treated group with control group of rats. There are no significant difference in tissue xanthine oxidase, myeloperoxidase, lipid peroxidation, protein oxidation and tissue reduced glutathione content in neutropenic group with control group of rats. Conclusion: Free radical-induced lipid peroxidation and suppression of antioxidizing enzymes play an important role in gastric damage induced by indomethacin. Inhibition of neutrophil infiltration by methotrexate may be one of the protective factors decreasing indomethacin-induced gastric mucosal injury. Keywords: Oxidative Stress, Non-steroidal Anti-inflammatory Drugs, Ulcer.
... However, the enhanced generation of NO by iNOS may contribute to the pathogenesis of various gastroduodenal disorders, including peptic ulcers. 73 Thus, the status of eNOS vs iNOS expression in gastric tissue is crucial for maintaining its integrity. Experimental evidence indicated that the upregulation of NF-κB also induces the transcriptional activation of iNOS. ...
Article
Full-text available
Purpose: Previously, we demonstrated that the specific ratio of Korean multi-herbal formula (SR-5) exhibits hepatoprotective properties against ethanol-induced hepatic damage in rats. Chronic and excessive alcohol consumption is a major etiological factor involved in gastric disease and ulcer development induced by the inflammatory response and oxidative stress. Methods: The present study evaluated the gastroprotective effects of SR-5 (100, 150, and 200 mg/kg) against hydrochloride acid/ethanol (HCl/EtOH)-induced and indomethacin/hydrochloride acid (INDO/HCl)-induced gastritis in a mouse model and the mechanisms involved. Results: All the tested doses of SR-5 significantly inhibited gastric lesions in the HCl/EtOH-induced ulcer model mice. Similarly, all the tested doses of SR-5 significantly inhibited gastric lesions in the INDO/HCl-induced ulcer model mice. Furthermore, mice pretreated with SR-5 had significantly increased gastric levels of enzymatic and nonenzymatic antioxidants, namely, catalase (CAT) and glutathione (GSH), with concomitant reductions in malondialdehyde (MDA) and reactive oxygen species (ROS) levels compared with those in the HCl/EtOH or INDO/HCl group. SR-5 suppressed the expression of nuclear factor-kappa B (NF-κB)/p65, inducible nitric oxide synthase (iNOS), tumor necrosis factor-α (TNF-α), and cyclooxygenase-2 (COX-2) to their normal values. Conclusion: These findings are the first to demonstrate the powerful protective effect of SR-5 against gastric injury development and provide hope for clinical application.
... Peptic ulcer refers to the group of ulcerative disorders of the gastro intestinal tract involving principally, the most proximal portion of the stomach and the duodenum, which is commonly due to the effect of acid and pepsin [27] .Stomach lesions are located preferentially along the small curvature in the transition zone between the body and the antrum, whereas in the duodenum those lesions are located in the bulb [28] . The present study investigated the effect of DAG on gastric and duodenal ulcers. ...
Article
The plant Dialium guineense (DAG) has been claimed by local users, to be effective in the treatment of peptic ulcers, especially, when taken as an aqueous decoction. The present study assessed the antiulcer activity of the plant, as well as explored the possible mechanisms of action of the herbal drug, aside identifying some of the various phytoconstituents, which could be responsible for its antiulcer activity. Different ulcerogens (ethanol 99.9 %, indomethacin 50 mg/kg, cysteamine 400 mg/kg, glacial acetic acid) and the pylorus ligation-induced ulcers were used to induce acute and chronic ulcers, with doses of 100, 300 and 750 mg/kg DAG and the standard drugs relative to each model, while assessing drug activity through ulcer scoring and comparing it with both the negative and positive controls. The extract, which has an LD50 of 1584.89 mg/kg when administered intraperitoneally, recorded a significant (p<0.05) antiulcer effect in all the models used in the study. Similarly, in the pylorus-ligated group, DAG compared effectively with atropine (1 mg/kg) and ranitidine (100 mg/kg), the standard antagonists of the secretagogues- carbachol and histamine employed in the study. The herbal drug produced a significant reduction in gastric juice volume, as well as in the free and the total acidity. The results suggest that DAG possesses a significant antiulcer property through cytoprotective and antisecretory actions, and it could be projected that the presence of secondary metabolites such as tannins, saponins and flavonoids could be responsible for its ulcer protective and healing property. The study therefore validates the folkloric use of DAG in the treatment of peptic ulcer.
... Another important consequence of naproxen-induced gastrointestinal injury is the increased concentration and activity of MPO, which is associated with increased neutrophil infiltration in damaged tissues. Therefore, MPO is considered to be a marker of inflammation and neutrophil infiltration in ulcerated conditions, and reduction in its levels occurs during the healing process (Higuchi et al., 2009;Souza et al., 2004). At all doses, LPu and LPe treatment reduced the MPO activity compared to the naproxen-treated group (4.147 U/g tissue ±1.38). ...
Article
Full-text available
This study investigated the anti‐inflammatory and antioxidant effects of hydroalcoholic extracts of mango peel and pulp on oxidative damage in a naproxen‐induced gastric injury rat model. The extracts were assessed for antioxidant activity (ABTS and FRAP methods), and the phenolic profile was investigated with UPLC‐QToF‐MSE. Gastric damage was evaluated in vivo by assessing the membrane lipid peroxidation (malondialdehyde (MDA) content), myeloperoxidase (MPO) enzyme activity, and glutathione (GSH) content. Mango peel and pulp contained high contents of bioactive compounds, particularly phenolics (69.50–5.287.70 mg gallic acid equivalents/100 g), carotenoids (651.30–665.50 μg/100 g), and vitamin C (21.59–108.19 mg/100 g). UPLC‐QToF‐MSE analysis identified 17 phenol compounds, including gallotannins, glycosylated flavonoids, and xanthone. The hydroalcoholic extracts of mango peel and pulp (LPe and LPu, respectively) significantly reduced the MPO activity and MDA content. In addition to preventing naproxen‐induced GSH decline, LPe (30 mg/kg) and LPu (10 mg/kg) restored its content to normal levels. LPe and LPu neutralized the oxidizing agents triggered by naproxen and reduced the severity of gastric lesions owing to their antioxidant properties. Epidemiological studies have shown that products with antioxidant activity can be used as potential agents in the treatment and prevention of chronic inflammatory diseases, such as gastric ulcers. Our study showed the possibility of using mango extracts as antioxidants and gastroprotectants, opening a new perspective in which a cheap and abundant food and its industrial residues can be transformed into pharmacological alternatives or applied as adjuvants in the treatment of chronic inflammatory diseases induced by oxidative stress.
... The current findings demonstrate that GSH decrease was preserved by Melaleuca quinquenervia. Additionally, MPO activity, a marker of inflammation, mostly increases ulcerations [24] also diminished by Melaleuca quinquenervia treatment, while increase in the other inflammation marker, MDA, was limited. ...
Article
Full-text available
Purpose: To evaluate the effects of M. quinquenervia extract on ethanol-induced peptic ulcer in rats. Methods: The following three groups of (n = 6) Sprague Dawley rats were included in this study: vehicle (C), ethanol-administered (E) and ethanol + M. quinquenervia-treated (MQ). MQ group rats received 100 µg/mL M. quinquenervia essential oil just before 96 % ethanol induction (1 ml/kg). One hour after ulcer induction, the animals were euthanized, and gastric and duodenal tissues were removed. Tissue samples were analysed for myeloperoxidase (MPO) activity, malondialdehyde (MDA), glutathione (GSH), myeloperoxidase (MPO) activity, malondialdehyde (MDA) and glutathione (GSH) levels, and histopathological examinations were performed by light microscopy. Results: Gastric and duodenal GSH levels that decreased in the ethanol-administered ulcer groups (p < 0.001), rose following MQ treatment (p < 0.5). Moreover, elevated MPO and MDA levels (p < 0.5) in gastric tissues decreased after MQ-treatment. Similarly, the MQ-treated group showed recovery and control-like morphology compared to the ethanol group in both gastric and duodenal tissues when examined by microscopy. Conclusion: The results indicate that M. quinquenervia extract has a positive impact on gastric injury in rats due to its antioxidant activity. Thus, the plant has a potential for the clinical management of gastric ulcer. This is an Open Access article that uses a funding model which does not charge readers or their institutions for access and distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0) and the Budapest
Article
Indomethacin is a widely used nonsteroidal anti-inflammatory drug; however, its clinical utility is accompanied by serious adverse reactions including peptic ulcers. The current study aims to investigate the protective potential of perindopril against indomethacin-induced gastric injury in rats. Perindopril (4 mg/kg) was administered orally for 7 days and indomethacin (60 mg/kg, single oral dose) was administered on the 7th day, 1 h after perindopril administration. Pantoprazole was used as a standard agent. Ulcer index (UI), preventive index ratio (PI), histopathological examination, oxidative stress, and inflammatory biomarkers were investigated. Perindopril significantly decreased UI while increased PI and counteracted histopathological aberrations induced by indomethacin. It alleviated indomethacin-induced oxidative stress by lowering NO while increasing GSH content and superoxide dismutase activity. Perindopril significantly downregulated TNF-α and asymmetric dimethylarginine (ADMA), while significantly upregulated COX-2, PGE-2, dimethylarginine dimethylaminohydrolase-1 (DDAH-1), ANG-(1-7), and ACE-2 expression. Together, these findings suggest the gastroprotective effects of perindopril through modulation of DDAH-1/ADMA and ACE-2/ANG-(1-7) signaling. • HIGHLIGHTS • Perindopril attenuated gastric histopathological damage. • It increased GSH content and SOD activity while decreased NO content. • It modulated gastric ADMA and DDAH-1 activity. • It reduced TNF-α, while increased COX-2 and PGE-2 expression. • It upregulated ACE-2 activity and ANG-(1-7) protein expression.
Article
Full-text available
The objective of this study was to determine whether endogenous nitric oxide (NO) inhibits leukocyte adhesion to vascular endothelium. This was accomplished by superfusing a cat mesenteric preparation with inhibitors of NO production, NG-monomethyl-L-arginine (L-NMMA) or NG-nitro-L-arginine methyl ester (L-NAME), and observing single (30-microns diameter) venules by intravital video microscopy. Thirty minutes into the superfusion period the number of adherent and emigrated leukocytes, the erythrocyte velocity, and the venular diameter were measured; venular blood flow and shear rate were calculated from the measured parameters. The contribution of the leukocyte adhesion glycoprotein CD11/CD18 was determined using the CD18-specific monoclonal antibody IB4. Both inhibitors of NO production increased leukocyte adherence more than 15-fold. Leukocyte emigration was also enhanced, whereas venular shear rate was reduced by nearly half. Antibody IB4 abolished the leukocyte adhesion induced by L-NMMA and L-NAME. Incubation of isolated cat neutrophils with L-NMMA, but not L-NAME, resulted in direct upregulation of CD11/CD18 as assessed by flow cytometry. Decrements in venular shear rate induced by partial occlusion of the superior mesenteric artery in untreated animals revealed that only a minor component of L-NAME-induced leukocyte adhesion was shear rate-dependent. The L-NAME-induced adhesion was inhibited by L-arginine but not D-arginine. These data suggest that endothelium-derived NO may be an important endogenous modulator of leukocyte adherence and that impairment of NO production results in a pattern of leukocyte adhesion and emigration that is characteristic of acute inflammation.
Article
Full-text available
Thalidomide selectively inhibits the production of human monocyte tumor necrosis factor alpha (TNF-alpha) when these cells are triggered with lipopolysaccharide and other agonists in culture. 40% inhibition occurs at the clinically achievable dose of the drug of 1 micrograms/ml. In contrast, the amount of total protein and individual proteins labeled with [35S]methionine and expressed on SDS-PAGE are not influenced. The amounts of interleukin 1 beta (IL-1 beta), IL-6, and granulocyte/macrophage colony-stimulating factor produced by monocytes remain unaltered. The selectivity of this drug may be useful in determining the role of TNF-alpha in vivo and modulating its toxic effects in a clinical setting.
Article
Full-text available
The hypothesis that neutrophils play an important role in the pathogenesis of gastric ulceration induced by nonsteroidal anti-inflammatory drugs (NSAIDs) was tested in rats. Rats made neutropenic by prior treatment with an antibody to rat neutrophils raised in goat were found to be significantly more resistant to the gastric-damaging actions of indomethacin or naproxen than were control rats or rats pretreated with normal goat serum. The reduction of damage in neutropenic rats was not due to effects of the antineutrophil serum on either gastric acid secretion or the ability of indomethacin or naproxen to inhibit prostaglandin synthesis. Gastric cyclooxygenase activity was inhibited by greater than 95% in both normal and neutropenic rats that received indomethacin or naproxen. Reduction of circulating neutrophil numbers by treating rats with methotrexate also resulted in a significant reduction in the susceptibility to gastric damage induced by indomethacin. Since activation of circulating neutrophils appeared to be important in the development of gastric erosions after administration of indomethacin, and in the significant changes in vascular endothelial integrity (Monastral Blue staining) observed within 15 min of indomethacin administration, we investigated the possibility that leukotrienes (LTs) and platelet-activating factor (PAF) might be involved in the pathogenesis of indomethacin-induced ulceration. Changes in gastric LTB4 synthesis were not observed after indomethacin administration. Pretreatment with either an LTD4 antagonist or a PAF antagonist was without significant effect on the extent of gastric damage induced by indomethacin. These results suggest an important role for neutrophils in the pathogenesis of NSAID-induced gastric ulceration. Neutrophils may be important in the vascular injury that occurs early after administration of these compounds.
Article
Nitric oxide is a crucial mediator of gastrointestinal mucosal defense, but, paradoxically, it also contributes to mucosal injury in several situations. Inhibitors of nitric oxide synthesis and compounds that release nitric oxide have been useful pharmacological tools for evaluating the role of nitric oxide in gastrointestinal physiology and pathophysiology. Newer inhibitors with selectivity for one of the isoforms of nitric oxide synthase are even more powerful tools and may have utility as therapeutic agents. Also, agents that can scavenge nitric oxide or peroxynitrite are promising as drugs to prevent nitric oxide-associated tissue injury. Compounds that release nitric oxide in small amounts over a prolonged period of time may also be very useful for prevention of gastrointestinal injury associated with shock and with the use of drugs that have ulcerogenic effects. Indeed, the coupling of a nitric oxide-releasing moiety to nonsteroidal anti-inflammatory drugs has proven to be a valid means of substantially reducing the gastrointestinal toxicity of these drugs without decreasing their efficacy.
Article
Gastric ulceration was induced in rats by i.p. injection of the non-steroidal anti-inflammatory drug (NSAID), indomethacin (IND) (30 mg kg(-1)). Pyloric ligation was carried out in each animal before injection to enable collection of the gastric juice. Three hours later, the animals were killed and their stomachs were removed. In the gastric juice, the amounts of mucin, pepsin and HCl were assessed. Gastric mucosa were scrapped for the determination of nitric oxide (NO) (as nitrite) after evaluation of the gastric ulcer index. The influence of arginine (ARG) (300 mg kg(-1)), a NO precursor, N(G)-nitro- l -arginine methyl ester (l -NAME) (50 mg kg(-1)), a non-selective constitutive nitric oxide synthase/inducible nitric oxide synthase (cNOS/iNOS) inhibitor, and the selective iNOS inhibitor aminoguanidine (AMG) (50 mg kg(-1)) were studied. Each NO modulator was injected i.p. 30 min before IND administration. Results indicated that IND elevated gastric acidity by 80% of the normal group, decreased non-significantly mucosal nitrite by 22% and exhibited a remarkably high ulcer index (chi = 17). Neither mucin nor pepsin levels were significantly altered. In comparison with the IND group, pretreatment with l -NAME caused a significant decrease in gastric HCl, further decrease in mucosal nitrite (50% of normal) and a two-fold increase in the ulcer index score (chi = 34), despite the decrease in HCl. AMG did not alter gastric acidity, decreased mucosal nitrite by 38% of the normal value and failed to alter significantly the ulcer index of IND. On the other hand, pretreatment with ARG did not alter the gastric acidity and raised mucosal nitrite by 10% above normal. Surprisingly, ARG improved the gastric ulcer score (chi = 1) almost similar to the normal score (chi = zero). Therefore, this study creates a new pathway for the potential treatment of NSAID gastric ulceration through modulation of NO synthesis, regardless of the effect on gastric acidity.
Article
Several studies have shown that polymorphonuclear neutrophil leukocyte (PMN) margination is an early and critical event in the pathogenesis of gastric mucosal injury caused by nonsteroidal anti-inflammatory drugs. Tumor necrosis factor (TNF) alpha is a proinflammatory cytokine that causes PMN margination by up-regulating expression of adhesion molecules on both PMN and endothelial cells. This study investigated whether substances that modulate TNF synthesis and release influence PMN margination and indomethacin-induced gastric damage. Rats were treated with several doses of indomethacin alone or in association with substances known to increase (interleukin 2 and lipopolysaccharide) or inhibit (pentoxifylline, dexamethasone, granulocyte colony-stimulating factor [G-CSF]) TNF synthesis and release. Indomethacin administration caused dose-dependent damage and increased PMN margination and plasma TNF concentrations. Pretreatment with interleukin 2 and lipopolysaccharide significantly increased TNF release, PMN margination, and gastric mucosal damage, but administration of dexamethasone, pentoxifylline, and G-CSF provided almost total protection. The administration of G-CSF alone caused a significant increase in gastric PMN margination but protected against the indomethacin-induced gastropathy. Agents that regulate TNF synthesis and release influence gastric susceptibility to indomethacin by modulating PMN margination. G-CSF increased PMN infiltration but protected against the mucosal injury, suggesting that PMN margination alone is not sufficient to induce mucosal damage.
Article
In the past few years, considerable progress has been made on the identification and characterization of tumor necrosis factor (TNF) receptors. The relative roles played by the two receptor types in signaling the diverse functions of TNF are less clear. Here, Louis Tartaglia and David Goeddel summarize progress to date and propose a model of TNF receptor triggering that reconciles the seemingly conflicting data.
Article
We explored the effect of in vitro phagocytosis and in vivo inflammation on the MPO content of functioning neutrophils and on the ability of these cells to export active MPO into the extracellular environment. After ingestion of staphylococci, neutrophils retained 52% of their MPO and released 8% into the medium in active form; the remaining 40% of their MPO could no longer be detected. During bacterial infection induced by intradermally injecting staphylococci, neutrophils harvested from minced infected lesions contained 52% of the MPO of circulating neutrophils that had not reached the lesions. Extracellular fluid from the lesions contained active MPO secreted by the neutrophils, and concentrations of 10-45 U/ml were detected. These data demonstrate that functioning neutrophils can lose approximately half of their MPO. In vitro, 4%-8% of neutrophilic MPO appears in the extracellular space and 40% is inactivated. In vivo, the MPO content of inflammatory neutrophils also decreases, and MPO appears in the extracellular fluid in active form where it is available to participate in a variety of physiologic processes.