Sei sulla pagina 1di 20

GASTROENTEROLOGY 2008;135:41– 60

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
REVIEWS IN BASIC AND CLINICAL
GASTROENTEROLOGY
David Metz, Section Editor

Gastric Mucosal Defense and Cytoprotection: Bench to Bedside

LOREN LAINE,* KOJI TAKEUCHI,‡ and ANDRZEJ TARNAWSKI§


*Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California; ‡Division
of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto, Japan; and
§
Gastroenterology Section, VA Long Beach Healthcare System and Division of Gastroenterology, University of California Irvine School of Medicine, Irvine, California

The gastric mucosa maintains structural integrity and


function despite continuous exposure to noxious fac-
tors, including 0.1 mol/L HCl and pepsin, that are
T he gastric mucosa is continuously exposed to many
noxious factors and substances. How the gastric
mucosa maintains structural integrity and resists auto-
capable of digesting tissue. Under normal conditions, digestion by substances such as 0.1 mol/L HCl and pep-
mucosal integrity is maintained by defense mecha- sin puzzled clinicians and investigators for more than
nisms, which include preepithelial factors (mucus- 200 years. An early hypothesis proposed by Hunter in
bicarbonate-phospholipid “barrier”), an epithelial 1772 and supported by Virchow in 1853 was that con-
“barrier” (surface epithelial cells connected by tight tinuous circulation of alkaline blood through the mu-
junctions and generating bicarbonate, mucus, phos- cosa neutralizes acid.1,2 Subsequent work demonstrated
pholipids, trefoil peptides, prostaglandins (PGs), and that a large number of mucosal defense mechanisms
heat shock proteins), continuous cell renewal accom- prevent mucosal damage and maintain mucosal integrity.
plished by proliferation of progenitor cells (regulated The discovery by Vane that the major mechanism by
by growth factors, PGE2 and survivin), continuous which aspirin and other NSAIDs produce gastric damage
blood flow through mucosal microvessels, an endo- is inhibition of prostaglandin (PG) synthesis,3 and the
thelial “barrier,” sensory innervation, and generation concept of cytoprotection developed in the late 1970s
and early 1980s by Robert et al4,5 sparked tremendous
of PGs and nitric oxide. Mucosal injury may occur
interest in gastric mucosal defense.
when noxious factors “overwhelm” an intact mucosal
Gastric mucosal injury may occur when noxious fac-
defense or when the mucosal defense is impaired. We
tors “overwhelm” an intact mucosal defense (eg,
review basic components of gastric mucosal defense
Zollinger Ellison syndrome) or when the mucosal defen-
and discuss conditions in which mucosal injury is
sive mechanisms are impaired. This review focuses on
directly related to impairment in mucosal defense, conditions in which mucosal injury is directly related to
focusing on disorders with important clinical sequelae: impairment in mucosal defense. Conditions in which the
nonsteroidal anti-inflammatory drug (NSAID)-asso- inciting factor is an injurious agent (eg, acid, Helicobacter
ciated injury, which is primarily related to inhibition pylori) are covered in other reviews in this series. We first
of cyclooxygenase (COX)-mediated PG synthesis, and review the basic components and mechanisms that pro-
stress-related mucosal disease (SRMD), which occurs vide gastric mucosal defense and then discuss clinical
with local ischemia. The annual incidence of NSAID- conditions that occur primarily because of impairment in
associated upper gastrointestinal (GI) complications gastric mucosal defense. Finally, we will review the pre-
such as bleeding is approximately 1%–1.5%; and re- vention and treatment of 2 important clinical conditions
ductions in these complications have been demon- that result from impaired mucosal defense: NSAID-asso-
strated with misoprostol, proton pump inhibitors ciated injury and SRMD.
(PPIs) (only documented in high-risk patients), and
COX-2 selective inhibitors. Clinically significant Abbreviations used in this paper: COX, cyclooxygenase; CRF, corti-
bleeding from SRMD is relatively uncommon with cotrophin-releasing factor; EGF, epidermal growth factor; NSAID, non-
modern intensive care. Pharmacologic therapy with steroidal anti-inflammatory drug; PGs, prostaglandins; PGI2, prostacy-
antisecretory drugs may be used in high-risk patients clin; PPIs, proton pump inhibitors; SRMD, stress-related mucosal
disease; TFFs, trefoil factor family peptides.
(eg, mechanical ventilation >48 hours), although the © 2008 by the AGA Institute
absolute risk reduction is small, and a decrease in 0016-5085/08/$34.00
mortality is not documented. doi:10.1053/j.gastro.2008.05.030
42 LAINE, TAKEUCHI, AND TARNAWSKI GASTROENTEROLOGY Vol. 135, No. 1
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

Figure 1. Diagrammatic representation of gastric mucosal defense, modified and updated from Tarnawski.9

Gastric Mucosal Defense Mechanisms pholipids, which cover the mucosal surface. This un-
Defense mechanisms permit the gastric mucosa to stirred layer retains bicarbonate secreted by surface epi-
withstand frequent exposure to damaging factors across thelial cells to maintain a neutral microenvironment (pH
a wide range of pH, osmolality, and temperature.6 –12 ⬃7.0) at the surface epithelial cells and prevents penetra-
These include local defense mechanisms and neurohor- tion of pepsin and thus proteolytic digestion of the
monal mechanisms described below (Figure 1). surface epithelium.14,15 The mucus gel contains phospho-
lipids, and its luminal surface is coated with a film of
Local Gastric Mucosal Defense Mechanisms surfactant phospholipids with strong hydrophobic
Mucus-bicarbonate-phospholipid “barrier.” The properties.8,16
mucus-bicarbonate-phospholipid “barrier” constitutes Mucus gel is secreted by apical expulsion from surface
the first line of mucosal defense.8,13–15 This barrier is epithelial cells and contains ⬃95% water and ⬃5% mucin
formed by mucus gel, bicarbonate, and surfactant phos- glycoproteins, products of mucin (MUC) genes. The gel-
July 2008 GASTRIC MUCOSAL DEFENSE AND CYTOPROTECTION 43

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
forming mucin units polymerize into large mucin mul- sin.7,6,9,14 The surface epithelial cells are metabolically
timers essential for gel formation.14,15,17 The structure of and electrically coupled by gap junctions. Heat shock
each of the gel-forming mucins, MUC2, MUC5AC, proteins are generated by gastric epithelial cells in re-
MUC5B, and MUC6, has been elucidated.14,18 In the sponse to stress, such as increased temperature, oxidative
stomach, MUC5AC is expressed in the surface epithelial stress, and cytotoxic agents.27–29 They prevent protein
cells of cardia, fundus, and antrum; and MUC6 is ex- denaturation and protect cells against injury. Activation
pressed in the neck cells of the fundus and in antral of heat shock protein response is one of the mucosal
glands. Alternating layers of MUC5AC and MUC6 have protective mechanisms of the antacid hydrotalcite.30
been demonstrated in the mucus layer in human gastric Cathelicidin and ␤ defensins are cationic peptides that
mucosa.14,19 Mucus gel is cosecreted with low-molecular- play roles in the innate defensive system at mucosal
weight trefoil factor family peptides (TFFs).20,21 TFFs are surfaces preventing bacterial colonization. They have
an integral part of the intracellular mucus secretory ves- been demonstrated in gastric epithelial cells, and they
icles and may play a role in the intracellular assembly accelerate ulcer healing.31 Surface epithelial cells secrete
and/or packaging of mucins.20 TFF2 increases the viscos- TFFs that regulate reepithelialization and exert mucosal
ity of gastric mucin and stabilizes the gel network.22 protective action.32
Mucus secretion is stimulated by gastrointestinal hor- Continuous cell renewal from mucosal progenitor
mones, including gastrin and secretin, as well as PGE2 cells. Continuous cell renewal from mucosal progenitor
and cholinergic agents.7,14 Ulcerogenic substances such cells maintains structural integrity of the mucosa. The
as aspirin and bile salts cause dissipation of the mucus epithelium is continually renewed by a well-coordinated
gel and phospholipid layer, leading to acid back-diffusion and controlled proliferation of progenitor cells that en-
and mucosal injury.14,23 ables replacement of damaged or aged surface epithelial
The secretion of HCO3⫺ into a stable, adherent mucus cells. Complete replacement of the gastric surface epithe-
gel layer creates a pH gradient at the epithelial surface in lium usually takes 3–7 days, whereas months are required
the stomach and duodenum and provides the first line of to replace the glandular cells. In gastric glands, a single
mucosal defense against luminal acid.14,15,24 pH gradient stem cell undergoes division to produce committed pro-
studies provided experimental evidence for the existence genitor cells, which further differentiate into an adult
of the mucus-bicarbonate barrier in vivo and presence of epithelial cell type.33 The stem/progenitor cell niche is
a nearly neutral pH at the epithelial surface.15 The surface made up of proliferating and differentiating epithelial
epithelium in acid-secreting gastric mucosa exports cells and surrounding mesenchymal cells.34 The latter
HCO3⫺ at rates only 10% of the acid secretion rate. generate growth factors and thus promote mesenchymal
Mucus gel minimizes luminal loss of HCO3⫺ sufficiently to epithelial cross talk and signaling to maintain the
to maintain a neutral pH at the apical cell surfaces. niche progenitor cell survival. Restitution of the surface
Experimental studies show that Na⫹HCO3⫺ cotransport epithelium after superficial injury occurs within minutes
at the basolateral membrane is the major mechanism for by migration of preserved epithelial cells located in the
import of HCO3⫺. Studies of rat and rabbit gastric mu- neck area of gastric glands.35,36 This migration precedes
cosa demonstrated expression of a Cl⫺/HCO3⫺ anion and is independent of proliferation of progenitor cells,
exchanger in the apical membranes of gastric surface which occurs later— hours after injury.36,37
epithelial cells.25 In the stomach, PGs stimulate HCO3⫺ Cell proliferation of progenitor cells is controlled by
secretion via EP1 receptors.26 Luminal acid, corticotro- growth factors. The major growth factor receptor ex-
phin-releasing factor (CRF), melatonin, uroguanylin, and pressed in gastric progenitor cells is epidermal growth
orexin A also stimulate HCO3⫺ secretion.7,14 factor receptor (EGF-R),38 and the major mitogenic
The mucus bicarbonate barrier is the only preepithelial growth factors that activate this receptor are transform-
barrier between lumen and epithelium. When it is over- ing growth factor ␣ (TGF-␣) and insulin-like growth
whelmed or breaks down in disease, the next series of factor-1.39 PGE2 and gastrin transactivate EGF-R and
protective mechanisms come into play, including intra- trigger the mitogen-activated protein kinase pathway
cellular neutralization of acid, rapid epithelial repair, and thereby stimulating cell proliferation and exerting a tro-
maintenance and distribution of mucosal blood flow. phic action on gastric mucosa.40 EGF peptide itself is
Surface epithelial cells. The next line of mucosal absent in normal gastric mucosa. However, it is present
defense is formed by a continuous layer of surface epi- in the gastric lumen, derived from salivary and esopha-
thelial cells (Figure 2), which secrete mucus and bicar- geal glands, and can stimulate progenitor cell prolifera-
bonate and generate PGs, heat shock proteins, TFFs, and tion in case of injury. An important new finding is the
cathelicidins. Because of the presence of phospholipids expression of survivin, an antiapoptosis protein, in gas-
on their surfaces, these cells are hydrophobic, repelling tric progenitor cells, which allows these cells to avoid
acid- and water-soluble damaging agents.8 Intercon- apoptosis and promotes mitosis (Figure 2C).41
nected by tight junctions, the surface epithelial cells form Alkaline tide. “Alkaline tide” occurs because
a “barrier” preventing back diffusion of acid and pep- stimulated parietal cells secreting hydrochloric acid
44 LAINE, TAKEUCHI, AND TARNAWSKI GASTROENTEROLOGY Vol. 135, No. 1
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

Figure 2. Structural elements of gastric mucosal defense. (A) Histology of upper part of human gastric mucosa visualizing surface epithelial cells
(SEC), foveoli (F), and upper gland area. (H&E staining; original magnification, ⫻50). Blood microvessels with erythrocytes in the lumen are present
in the lamina propria (arrows). (B) Scanning electromicrograph of human gastric mucosal luminal surface. The unstirred mucus gel layer is not seen
because of dissolution during fixation. Individual surface epithelial cells (SEC) are clearly visible as are lumina of the gastric pits (arrows). Reproduced
with permission from Tarnawski A, Krause WJ, Ivey KJ. The effect of glucagon on aspirin-induced gastric mucosal damage in man. Gastroenterology
1978;74:240 –245. (C) Immunostaining of human gastric mucosa with survivin (antiapoptosis protein) antibody. Survivin is strongly expressed
(brown-red staining) in the progenitor cells located in the foverolar/neck area (arrowheads). Reproduced with permission from Tarnawski et al.159 (D)
Vascular cast study of capillary blood vessels in the gastric mucosa. The remaining components of the mucosa were dissolved in concentrated NaOH
Reproduced with permission from Ichikawa et al.165 (E) Transmission electronmicrograph of normal human gastric mucosa. Surface epithelial cells
(SEC) contain prominent, dark mucus granules. Below the surface epithelial cells, a capillary blood vessel (CAP) with erythrocytes (E) in the lumen is
present in the lamina propia. N, nucleus of endothelial cell lining capillary vessel (original magnification, ⫻2000). Reproduced with permission from
Tarnawski et al.172 (F) Transmission electronmicrograph of a portion of human gastric capillary blood vessel. The structure of the capillary wall and
endothelial cell cytoplasm is normal with a characteristic fenestration (arrows) and presence of endothelial vesicles. BM, basement membrane; E,
erythrocytes in the capillary lumen; J, junction between 2 neighboring endothelial cells; CF, collagen fibers. Original magnification, ⫻17,400.
Reproduced with permission from Tarnawski et al.172

into the gastric gland lumen concurrently secrete bi- upward in proximity to gastric glandular epithelial
carbonate into the interstitium and lumen of adjacent cells (Figures 1 and 2). At the base of surface epithelial
capillary blood vessels (Figure 1). This bicarbonate is cells, capillaries converge into collecting venules.42
transported upward to the base of the surface epithe- The endothelial cells lining the microvessels generate
lial cells and to the gastric lumen, where it contributes potent vasodilators such as nitric oxide (NO) and pros-
to the unstirred layer of mucus and bicarbonate. tacyclin (PGI2), which protect the gastric mucosa
Mucosal microcirculation. Mucosal microcircu- against injury and oppose the mucosal damaging ac-
lation is essential for delivery of oxygen and nutrients tion of vasoconstrictors such as leukotriene C4, throm-
and removal of toxic substances. At the level of the boxane A2, and endothelin. PGI2 and NO main-
muscularis mucosae, most gastric arteries branch into tain viability of endothelial cells and prevent platelet
capillaries, which enter the lamina propria and travel and leukocyte adherence to the microvascular endo-
July 2008 GASTRIC MUCOSAL DEFENSE AND CYTOPROTECTION 45

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
Figure 3. Sensory innervation of gastric mucosa. Photomicrographs of rat gastric specimens immunostained with calcitonin gene-related peptide
(CGRP) antibody demonstrate the distribution of CGRP immunoreactivity reflecting sensory nerves. CGRP-immunoreactive fibers are present in the
submucosa, often close to the blood vessels (A; arrowheads). The nerve fibers form a dense plexus at the mucosal base (A; arrows). From this plexus,
sensory nerves enter the lamina propria and together with microvessels penetrate toward the mucosal surface (B; arrowheads), ending just beneath
the basement membrane of the surface epithelial cells. Through the acid sensing ion channels, these nerve endings sense luminal contents such as
acid and other irritants. Reproduced with permission from Tarnawski et al.48

thelial cells, thus preventing compromise of the Sensory innervation of gastric mucosa. Gastric
microcirculation.43 mucosa and submucosal vessels are innervated by pri-
When the gastric mucosa is exposed to an irritant or mary afferent sensory neurons and nerves forming a
when acid back-diffusion occurs, a marked and rapid dense plexus at the mucosal base (Figure 3A).45,48,49,50 The
increase in mucosal blood flow occurs. This increase nerves fibers from this plexus enter the lamina propria
allows removal and/or dilution of the back-diffusing acid (accompanying capillary vessels) and end just beneath the
and/or noxious agents. This response seems to be essen- surface epithelial cells (Figure 3B). These nerve endings can
tial for mucosal defense because its abolition through sense the luminal content and/or entry of acid into the
mechanical restriction of blood flow leads to hemor- mucosa via acid-sensing channels. Activation of these nerves
rhagic necrosis. This hyperemic response is mediated by directly affects the tone of submucosal arterioles, which
sensory afferent nerves (see below). The increased muco- regulate mucosal blood flow.43 Stimulation of gastric sen-
sal blood flow in response to acid secretion is mediated, sory nerves leads to the release of neurotransmitters such as
at least in part, by NO, generated by NO synthase. NO calcitonin gene-related peptide and substance P in the nerve
plays a major role in mucosal defense by modulating the terminals located within or in close proximity to the large
mucosal circulation.43– 45 Endogenous and exogenous submucosal vessels.45,48,49,50 Calcitonin gene-related peptide
NO protects gastric mucosa against injury by ethanol exerts a mucosal protective action, most likely through
and endothelin 1, whereas inhibition of NO synthase vasodilatation of submucosal vessels mediated by NO gen-
(resulting in reduced NO generation) increases gastric eration. Interference with any aspect of the sensory inner-
mucosal injury.9,43,45 vation, such as ablation of the sensory afferent nerves with
Hydrogen sulfide is another endogenously generated chronic, large doses of capsaicin, impairs the hyperemic
compound that exerts a strong mucosal protective action response and thus diminishes resistance of the gastric mu-
similar to NO; it reduces tumor necrosis factor ␣ cosa to injury.44,45,49,50
(TNF-␣) expression, decreases leukocyte adherence to Continuous generation of PGE2 and PGI2.
vascular endothelium, and inhibits NSAID-induced gas- Continuous generation of PGE2 and PGI2 by the mu-
tric mucosal injury.46,47 cosa is crucial for the maintenance of mucosal integ-
46 LAINE, TAKEUCHI, AND TARNAWSKI GASTROENTEROLOGY Vol. 135, No. 1
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

rity and protection against ulcerogenic and necrotizing hancement of gastric mucosal blood flow via stimulation
agents.9 –12,44,51–53 Almost all of the mucosal defense of NO production and calcitonin gene-related peptide
mechanisms are stimulated and/or facilitated by PGs. release from sensory afferent nerves.70
PGs inhibit acid secretion; stimulate mucus, bicarbon- Adrenal glucocorticoids also play an important, facili-
ate, and phospholipid secretion; increase mucosal tating role in gastric mucosal defense.68,69,71–73 An acute
blood flow; and accelerate epithelial restitution and rise of corticosterone during stress is a potent gastropro-
mucosal healing.9 –12,44,51–54 tective component of the hormonal response to stress.71
PGs also inhibit mast cell activation and leukocyte and Pretreatment with glucocorticoid at pharmacologic doses
platelet adherence to the vascular endothelium.9 –12,51–53 causes a long-lasting decrease of stress-induced cortico-
The importance of PGE2 and PGI2 in gastric mucosal sterone rise and increased gastric ulcerogenic responses
defense is demonstrated by the fact that immunoneutral- to different models of stress, whereas corticosterone re-
izing antibodies to these PGs cause development of gas- placement reduces gastric lesions in rats.72 Administra-
tric and duodenal ulcers in rabbits and dogs55,56 identical tion of a specific glucocorticoid receptor antagonist (RU-
to those produced by NSAIDs that inhibit PG generation. 38486) increases the severity of stress-induced erosions,
Endogenous and exogenous E-type PGs and their an- further supporting a gastroprotective role of glucocorti-
alogs exert biologic actions via EP receptors 1– 4.57 The coids during stress.71 The protective action of glucocor-
mucosal protective action of PGs is mainly mediated via ticoids is assumed to occur via their maintenance of
EP-1 receptors, which also increase bicarbonate secretion glucose homeostasis, gastric blood flow, and mucus se-
and mucosal blood flow in damaged mucosa and de- cretion and their attenuation of enhanced gastric motil-
crease gastric motility.58 – 60 EP-3 and EP-4 receptors af- ity and microvascular permeability.73
fect acid and mucus secretion, respectively.61,62
Gastric Mucosal Injury Because of
Neurohormonal Regulation of Gastric Impaired Mucosal Defense
Mucosal Defense The most important conditions in clinical prac-
In addition to local mucosal factors, gastric mu- tice that are initiated by impairment of gastric mucosal
cosal defense is regulated, at least in part, by the central defense are NSAID-related injury and SRMD. Other fac-
nervous system and hormonal factors.10,44,50,63–70 Central tors that reduce gastric mucosal defense include aging
vagal activation increases mucus gel and surface cell and portal hypertension.
intracellular pH in rat stomach,63 and central CRF sig-
naling pathways are involved in the endocrine and vis- NSAID-Related Gastric Injury
ceral responses to stress.64,65 Peripheral CRF-related NSAIDs are the most widely used medications in
mechanisms also contribute to stress-induced changes in the United States.74,75 Eleven percent of the US adult
gut motility and mucosal function. The CRF2 receptor is population use an NSAID regularly (nearly every day for
present in the human stomach in which it plays a pro- a month or more),75 and a much larger number use
tective role by inhibiting apoptosis.64 Peripheral injection NSAIDs intermittently.74
of CRF or urocortin inhibits gastric emptying and mo- Traditional, nonselective NSAIDs induce predictable
tility through interaction with CRF2 receptors. Con- gastric mucosal injury. Initial early injury with agents
versely, CRF receptor antagonists given peripherally pre- such as aspirin may occur because of a topical effect.
vent acute restraint and stress-induced delayed gastric Most nonselective NSAIDs are weakly acidic. In gastric
emptying. These findings provide evidence that activa- juice, they are relatively nonionized and lipophilic, allow-
tion of peripheral CRF receptors and mast cells are im- ing them to move across cell membranes into the interior
portant mechanisms involved in stress-related alterations of cells.76 The neutral pH within the cell causes conver-
of gut physiology.10 –12,44,51,63– 65 Also, activation of central sion to the ionized form of the NSAID, leading to accu-
opioid receptors enhances gastric mucosal defense.66 mulation within the cell and local injury. Within 10
Various peptides including gastrin 17, cholecystokinin, minutes of a single dose of 650 mg of aspirin, the gastric
thyrotropin-releasing hormone, bombesin, CRF, EGF, potential difference is decreased, and 25% of surface ep-
peptide YY, neurokinin A analogs, and intragastric pep- ithelial cells show damage on light and electron micro-
tone exert gastroprotection that is abolished by afferent scopic examination.77 On endoscopic examination, sub-
nerve denervation, blockade of calcitonin gene-related epithelial hemorrhages develop within 15–30 minutes.
peptide receptors, and inhibition of NO synthase.44,67 Virtually 100% of subjects given aspirin 650 mg once
Ghrelin, a peptide hormone produced by gastric A-like develop gastric subepithelial hemorrhages, and virtually
cells in rodents and P/D1 cells in humans, regulates all subjects given aspirin 650 mg 4 times in 24 hours
growth hormone secretion at the hypothalamic and pi- develop gastric erosions—primarily in the antrum.76 Sub-
tuitary level and elicits orexigenic (appetite stimulating) epithelial hemorrhages and erosions are confined to the
action.70 Ghrelin also may exert peripheral actions such mucosa and therefore do not cause clinically severe GI
as gastric mucosal protective and healing action by en- complications such as major bleeding or perforation.
July 2008 GASTRIC MUCOSAL DEFENSE AND CYTOPROTECTION 47

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
Table 1. Upper GI Complications and Clinical Events From Traditional NSAID Arms of GI Outcome Studies ⱖ6 Months
Duration Without GI Protective Cotherapy
MUCOSA CLASS VIGOR TARGET
(n ⫽ 4439)87 (n ⫽ 3981)88,89 (n ⫽ 4029)90 (n ⫽ 9127)91
Type of arthritis Rheumatoid Osteoarthritis (73%); Rheumatoid Osteoarthritis
rheumatoid (27%)
NSAID 10 specified Ibuprofen 800 mg 3 times Naproxen 500 mg Ibuprofen 800 mg three times
NSAIDs daily; diclofenac 75 mg twice daily daily; naproxen 500 mg
twice daily twice daily
Low-dose aspirin Not stated 22% Exclusion criterion 24%
Median follow-up 6 mo 9 mo 9 mo 12 mo
Upper GI complications 1.5 1.0 1.4 1.3
(annualized incidence) (%)
Upper GI clinical events 2.7 2.8 4.5 2.8
(annualized incidence) (%)

NOTE. Upper GI complications include bleeding, perforation, or obstruction. Clinical events include complications plus uncomplicated symptom-
atic ulcers.

Although direct topical injury may occur, the major complicated events (bleeding, perforation, or obstruc-
mechanism via which NSAIDs cause ulcers and GI com- tion) and uncomplicated events (eg, symptomatic ulcer
plications is thought to be systemic: inhibition of COX- identified on endoscopy done for abdominal pain). The
mediated PG synthesis, as discussed below. Clinical in- risk of upper GI complications in observational popula-
formation supports the primacy of a systemic NSAID tion studies in NSAID users is approximately 4-fold
effect in induction of gastric injury. NSAIDs given by the higher than in people not taking NSAIDs.86 Four large
rectal, intravenous, and intramuscular routes increase the prospective GI outcome studies of ⱖ6 months assessed
risk of ulcers and ulcer complications.78 upper GI complications and overall upper GI clinical
Gastroduodenal ulcers identified at endoscopy are events in arthritis patients taking traditional NSAIDs
common in patients taking nonselective NSAIDs regu- without GI-protective cotherapy (Table 1).87–91 Annual-
larly, with a prevalence of ⬃15%–30%76 and a 6-month ized incidences of upper GI complications were 1%–1.5%,
incidence in double-blind trials as high as ⬃45%.79 Gas- and annualized incidences of all upper GI clinical events
tric ulcers are approximately 4 times more common than were 2.7%– 4.5%.
duodenal ulcers in patients taking NSAIDs79,80: the inci- Pathogenesis of NSAID-Induced Gastric Injury:
dence of gastric ulcers in a pooled analysis of 3– 6 month Prostaglandin Synthesis and the Role of COX-1 and
randomized trials of traditional NSAIDs was 19% vs 5% COX-2 in Gastric Mucosal Defense. COX-1 and COX-2
for duodenal ulcers.80 are key enzymes in the biosynthesis of PGs. COX-1 is
Continued use of nonselective NSAIDs after ulcer for- constitutively expressed in many tissues,92–94 whereas
mation decreases healing rates even with concomitant COX-2 has little or no expression in most tissues but is
antisecretory therapy: 8-week gastric ulcer healing rates
rapidly induced in response to growth factors and cyto-
with histamine H2-receptor antagonist (H2RA) therapy
kines.95,96 The COX-1-mediated PG synthesis is mainly
were 95% with discontinuation vs 63% with continuation
responsible for maintaining gastric mucosal integrity at
of NSAIDs.81 Whether COX-2 selective NSAIDs also de-
baseline. The traditional NSAIDs such as indomethacin
crease healing has been a question of interest. Induction
or ibuprofen are dual, nonselective inhibitors of both
of gastric ulcers in animal models is associated with a
marked rise in local COX-2 expression, and selective COX-1 and COX-2. They cause damage in the stomach
COX-2 inhibitors delay healing of these experimental with a marked decrease in the gastric mucosal PGE2
ulcers similarly to nonselective NSAIDs.82– 84 To study content.97,98 Gastric injury was assumed to be related to
this issue, patients with NSAID-associated gastric ulcers inhibition of gastric mucosal PG production by COX-1.
were randomly assigned to celecoxib 200 mg twice daily This contention was further supported by the fact that
or placebo in a double-blind trial.85 All received famoti- COX-2 selective NSAIDs, which do not inhibit COX-1 at
dine 40 mg daily and had endoscopy at 4 and 8 weeks. A therapeutic doses, do not affect the mucosal PG produc-
significantly lower healing rate was seen with celecoxib tion and do not produce gross gastric injury in experi-
(66% vs 80%, respectively; P ⫽ .015), indicating that mental models. However, a selective COX-1 inhibitor
COX-2 selective inhibitors delay ulcer healing in ulcers (SC-560) also does not cause gross damage in the stom-
and supporting the concept that the PGs produced by ach, despite inhibiting PG production in the gastric mu-
the up-regulated COX-2 are important for ulcer healing. cosa.99 –101 In contrast, the combined administration of
The major clinical concern regarding NSAID use is COX-1 selective and COX-2 selective inhibitors induces
their induction of upper GI clinical events, composed of mucosal damage. These data challenge the contention
48 LAINE, TAKEUCHI, AND TARNAWSKI GASTROENTEROLOGY Vol. 135, No. 1
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

that only COX-1 plays a “housekeeping” role in the resulting hypoxia and decreased mucosal defense.104
stomach. Inhibition of COX-1 but not COX-2 also increases acid
Selective inhibition of COX-1 induces a PG deficiency secretion in the damaged stomach.110
but up-regulates COX-2 expression.100,101 This conten- In summary, the likely sequence of events in NSAID-
tion is supported by the fact that gastric mucosal PGE2 induced gastric injury is inhibition of PG synthesis and
content in a rat model is markedly decreased by indo- induction of gastric hypermotility, followed by microvas-
methacin and SC-560, yet the values are partially restored cular disturbances and neutrophil activation (Figure 4).
8 hours after SC-560 but not indomethacin, a time suf- Gastric hypermotility and microvascular disturbances are
ficient for COX-2 induction.100,101 In addition, combined associated with a PG deficiency caused by COX-1 inhibi-
treatment with SC-560 and rofecoxib induces gross gas- tion. However, the inhibition of COX-1 up-regulates the
tric injury, but the damage is prevented by administra- expression of COX-2, and the PGs produced by COX-2
tion of PGE2 4 hours after the selective COX inhibitors. suppress the neutrophil-endothelial interaction caused
Furthermore, a selective COX-2 inhibitor by itself in- by the vascular disturbances because of COX-1 inhibi-
duces damage in the rat stomach when the expression of tion. These sequential events related to COX-1 and
COX-2 is up-regulated in the gastric mucosa by adrenal- COX-2 inhibition may explain why gastric damage occurs
ectomy, adjuvant-induced arthritis, or the administration only when both COX isozymes are inhibited and indicate
of COX-1 inhibitors.101–103 Thus, both COX-1 and a “housekeeping” role of COX-1 as well as COX-2 in the
COX-2 may play a role in PG synthesis and maintenance stomach.
of gastric mucosal integrity. COX-2 plays a “back-up” Potential Prostaglandin-Independent Mecha-
role by alleviating PG deficiency in situations in which nisms of NSAID-Induced Gastric Injury. NSAIDs may
COX-1-induced PG synthesis has been decreased. induce tissue and cell injury by mechanisms independent
Wallace et al99 found that neutrophil adherence to of prostaglandin inhibition, related to the inhibition of
gastric mesenteric venules was increased by indometha- oxidative phosphorylation in mitochondria, inhibition of
cin and celecoxib but not by SC-560, whereas gastric kinases (phosphorylating enzymes), and/or activation of
mucosal blood flow was decreased by indomethacin and apoptosis.111 A potential role for leukotrienes in NSAID-
SC-560 but not celecoxib. These data indicate that induced gastric injury also has been postulated. With the
NSAID-induced reduction in gastric mucosal blood flow decrease in arachidonic acid metabolism via the COX
is due to COX-1 inhibition, whereas NSAID-induced neu- pathway in NSAID users, arachidonic acid metabolism
trophil adherence to vascular endothelium is due to may be shifted to the alternative 5-lipoxygenase pathway,
COX-2 inhibition. Thus, reduction in mucosal blood with a resultant increase in leukotriene production. This
flow alone with COX-1 inhibition is insufficient to cause suggestion was supported by experimental studies docu-
gross gastric damage. In line with the finding that COX-1 menting that licofelone, an inhibitor of COX-1, COX-2,
is responsible for maintaining mucosal blood flow, SC- and 5-lipoxygenase, did not increase gastric mucosal in-
560 by itself, but not rofecoxib, increased microvascular jury. A randomized 4-week endoscopic trial in 121
permeability in the stomach, similarly to indometha- healthy volunteers revealed no gastric ulcers with placebo
cin.100,104 However, both COX-1 and COX-2 may be re- or licofelone (vs 20% with naproxen); the gastric mucosa
quired for enhancement of neutrophil migration in the was normal in 90% of subjects receiving placebo and 91%
gastric mucosa. Neither SC-560 nor rofecoxib alone af- of those receiving licofelone, compared with 37% of those
fect myeloperoxidase activity in the gastric mucosa, yet given naproxen.112
together they increase myeloperoxidase activity to levels Potential Lessening of Gastric Mucosal Injury
comparable with those induced by indomethacin.100,104 –107 With Aspirin. Interestingly, aspirin may provide some
This may reflect the combination of COX-1-mediated mu- protection against COX-mediated gastric injury in exper-
cosal blood flow and COX-2-mediated neutrophil adher- imental studies. Unlike other NSAIDs, acetylation of
ence to vascular endothelium. COX-2 by aspirin leads to generation of 15(R)15-epili-
Several investigators, using a variety of experimental poxin A4, termed aspirin-triggered lipoxin. 15(R)15-epili-
models, have also demonstrated a role of motility in poxin A4 and analogs exert protective actions on the
gastric mucosal defense. Normal gastric motility, me- gastric mucosa by inhibiting neutrophil chemotaxis, re-
diated by COX-1-induced PG synthesis, may be in- ducing neutrophil-mediated tissue injury and epithelial
volved in maintaining normal gastric mucosal blood apoptosis, and mediating adaptation of the gastric mu-
flow.100,104,108,109 A marked increase in gastric motility cosa to long-term aspirin administration. High-dose as-
is reported after treatment with the selective COX-1 pirin in rats induces COX-2 expression and lipoxin A4
inhibitor SC-560 but not the selective COX-2 inhibitor production in the gastric mucosa,113 and coadministra-
rofecoxib, although the duration of hypermotility is tion of aspirin and a selective COX-2 inhibitor resulted in
shorter than that induced by nonselective COX inhib- substantially more severe gastric injury than that pro-
itors.101 High amplitude contractions may result in a duced with either agent alone. In human volunteers,
temporal restriction of blood flow to the mucosa with low-dose aspirin increases urinary excretion of lipoxin A4,
July 2008 GASTRIC MUCOSAL DEFENSE AND CYTOPROTECTION 49

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
Figure 4. Current hypotheses
for roles of COX-1 and COX-2 in
the pathogenic mechanism of
NSAID-induced gastric damage.
The motility hypothesis suggests
that gastric motility plays an im-
portant role in NSAID damage.
NSAIDs induce vagal-depen-
dent gastric hypermotility via in-
hibition in COX-1-mediated
prostaglandin (PG) production
and CNS actions. Subsequent
microvascular disturbances lead
sequentially to neutrophil-endo-
thelial interaction and oxyradical
production. Inhibition of COX-1
leads to up-regulation of COX-2
expression. PG production me-
diated by COX-2, which may
suppress the neutrophil-endo-
thelial interaction, is also de-
creased by COX-2 selective or
nonselective NSAIDs. The neu-
trophil-endothelial interaction
plays a major role in the neutro-
phil hypothesis, which suggests
that NSAIDs activate the neutro-
phil through alteration of arachi-
donic acid metabolites (eg,
PGs), enhancing neutrophil-en-
dothelial cell adhesion.

whereas production of lipoxin A4 is inhibited by a COX-2 results may not be generalizable to patients with central
selective inhibitor.113,114 The latter effect has been sug- nervous system disease, trauma, burns, or transplanta-
gested as a potential explanation for the more than tion because these groups were not well represented in
additive effect of COX-2 selective inhibitors plus low- the population. In a subsequent multivariable analysis of
dose aspirin in causing mucosal injury.115 In addition, 1200 ICU patients on mechanical ventilation receiving
aspirin is reported to not increase basal gastric motil- prophylactic therapy, maximum serum creatinine during
ity.104,108 The clinical relevance of these observations is ICU stay was the sole patient characteristic identified as
uncertain because aspirin in full anti-inflammatory doses a significant predictor of clinically important bleeding
is not “safer” than other NSAIDs. (relative risk [RR], 1.16; 95% confidence interval [95% CI]:
SRMD 1.02–1.32).117
Endoscopic studies generally indicate that approxi-
SRMD occurs in critically ill patients: not all pa-
tients in an intensive care setting but only those who are mately 75%–100% of critically ill patients have gross
extremely ill, eg, severe trauma, burns ⬎one third of the gastric lesions visible when endoscopy is performed
body surface area, major intracranial disease, major sur- within the first 1–3 days of illness; the proportion is
gery, and severe medical illness. Prospective assessment of probably closer to 100% in patients who are extremely
2252 medical-surgical intensive care unit (ICU) patients ill as defined above.118 –120 An exception to this finding
revealed only 2 independent risk factors for clinically was reported in a study of 40 patients with intracranial
important stress bleeding: mechanical ventilation for disease requiring neurosurgery and mechanical venti-
ⱖ48 hours (odds ratio [OR], 15.6) and coagulopathy lation ⬎48 hours121: less than half of patients under-
(OR, 4.3).116 These data were generated primarily in pa- going endoscopy within 12 hours of admission had
tients with cardiovascular surgery and medical illness; the erosions or subepithelial hemorrhage, although the
50 LAINE, TAKEUCHI, AND TARNAWSKI GASTROENTEROLOGY Vol. 135, No. 1
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

proportion increased to ⬎50% at 5 days in those with absence of acid, gross gastric lesions were minimal, in-
no stress prophlyaxis. Lesions are generally diffuse volving 4% of the body and 3% of the antrum surface
subepithelial hemorrhage and erosions and occur more area. However, intragastric instillation of acid markedly
commonly in the proximal stomach.119,122 Bleeding increased the gross lesions in dose-dependent fashion to
from these mucosal lesions tends to be slow; if massive 53% and 45% of the body and antral surface areas, respec-
bleeding occurs, it suggests that an ulcer (ie, extension tively, with 0.1 mol/L HCl. Oxygen-free radicals appear to
below the mucosa) has developed. play a role in the induction of ischemia-reperfusion his-
Overt bleeding (eg, bloody nasogastric aspirate, he- tologic injury (which is prevented by allopurinol) but not
matemesis, melena) is reported in ⬃20% of critically ill injury because of ischemia alone (not prevented by allo-
patients; pooled results from placebo/no therapy control purinol). Ritchie also suggested that bile salts, along with
groups of 10 randomized controlled trials in metaanaly- ischemia and acid, were required to induce acute gastric
ses of prophylactic therapy performed from 1980 to mucosal injury.138
1998120,121,123–132 reveal an incidence of 17%. However, Ischemia/reperfusion increases COX-2 messenger RNA
the most relevant outcome to consider is clinically sig- levels in the rat gastric mucosa,139 and hypoxia increases
nificant bleeding. Clinically significant or clinically im- the expression of the COX-2 gene in human vascular
portant bleeding has been variably defined to include endothelial cells in vitro.140 Ischemia/reperfusion-in-
overt bleeding combined with transfusion, hemodynamic duced gastric damage is also aggravated by administra-
instability, and/or the need for intervention and is now tion of selective COX-2 inhibitors and dexamethasone at
suggested to occur in ⱕ5% of patients.122 Pooled analysis a dose that inhibits COX-2 expression.139 COX-2 expres-
of placebo control groups in the randomized trials from sion is therefore assumed to be up-regulated as one of the
metaanalyses cited above reveals an 8% incidence of clin- protective mechanisms when the stomach is exposed to
ically significant bleeding. The mortality in patients with stress such as ischemia/reperfusion.
stress-related bleeding is high, related primarily to the Reduced local mucosal NO generation and increased
underlying illness. Although stress-related bleeding has endothelin-1 (a potent vasoconstrictor) also appear to
been calculated to increase hospital stay and mortality, play an important role in the mechanisms of
studies of prophylactic therapy have not revealed that SRMD.141,142 Endogenous NO plays a dual role in isch-
decreased bleeding translates into decreased mortality.133 emia/reperfusion-induced gastric injury: constitutive NO
The incidence of SRMD-associated clinically significant synthase/NO is protective, whereas inducible NO syn-
bleeding appears to have markedly decreased in the past thase/NO is proulcerogenic.143,144 The gene expression of
few decades, most likely because of better care of critically inducible NO synthase is markedly up-regulated in the
ill patients.122,134 stomach following ischemia/reperfusion, accompanied
Pathogenesis of SRMD. Stress-related gastric by a significant increase in the mucosal NO content,
mucosal injury appears to be related to local ischemia, supporting a pathogenic role.145
although progression to significant mucosal injury re- Hemorrhagic shock in an animal model has been re-
quires acid as well. Critically ill patients who develop ported to increase gastric mucosal permeability with in-
acute gastric mucosal lesions have a significant decrease creased back diffusion of hydrogen ions,146 presumably
in gastric mucosal blood volume compared with controls, allowing for greater mucosal injury in the presence of
whereas patients without acute gastric lesions do not.135 intraluminal gastric acid. This increase in back diffusion
Studies in the rat model136 reveal a significant correla- has also been reported in critically ill patients.146 Gastric
tion between blood pressure during hypotension and mucosal acidosis can be assessed in critically ill patients
gastric mucosal blood flow. Leung et al reported that a with gastric tonometry. The intramucosal pH, which is
threshold level of hypotension to 40% of baseline was calculated from intraluminal gastric juice pCO2 and ar-
required for lesions to form.136 Similar to reports in terial HCO3⫺, is used as a measure of mucosal ischemia
endoscopic studies, gross gastric lesions develop in a and is reported to be a significant risk factor for major
significantly greater area of the body than the antrum in bleeding from stress ulceration and mortality.147
the rat model. Critically ill patients may have a decrease in gastric
A rat model of hemorrhagic shock and retransfusion- mucosal blood flow through systemic hypotension, but,
induced gastric injury reveals that histologic mucosal in addition, patients may have splanchnic hypoperfusion
damage occurs even without acid, although it increases even when systemic hemodynamics are relatively main-
with intragastric instillation of acid in a dose-dependent tained.122,148 Normotensive critically ill septic patients on
fashion.137 In addition, reperfusion after ischemia in- mechanical ventilation were reported to have a 50%– 60%
creased histologic injury as compared with ischemia reduction in mucosal blood flow measured by reflectance
alone, although increasing the duration of ischemia spectrophotometry compared with controls.122 In addi-
(from 20 to 30 minutes) increased gastric histologic le- tion, induction of an isolated increase in splanchnic
sions to the same extent as a similar period of retransfu- blood flow in critically ill patients, in the absence of any
sion (10 minutes after 20 minutes ischemia). In the significant changes in systemic measurements of hemo-
July 2008 GASTRIC MUCOSAL DEFENSE AND CYTOPROTECTION 51

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
dynamics and oxygen delivery, was noted to increase trate.164 Furthermore, a dramatic 10-fold reduction of
gastric intramucosal pH.149 gastric angiogenesis occurs in response to alcohol injury
Of interest, mild stress prevents gastric lesions that in portal hypertensive gastric mucosa.165 The relative
occur in response to severe stress. This effect is mediated contribution of this impairment in gastric mucosal de-
by PGs derived from both COX-1 and COX-2, probably fense to the blood loss seen in some patients with portal
in both the brain and the stomach, and the effect is hypertensive gastropathy has not been defined.
functionally associated with prevention of the decrease in
body temperature that occurs during severe stress.150 Reduction of Upper GI Injury and
Phospholipase A2 plays a role, triggering central PG pro- Clinical Events
duction by COX-1 and COX-2, and thyrotropin-releasing Cytoprotection
hormone also may be involved in the protective action of Cytoprotection is defined as an ability of pharma-
mild stress. cologic agents, originally PGs, to prevent gastric and
intestinal mucosal injury produced by a variety of ulcero-
Aging gastropathy genic agents (eg, aspirin, indomethacin, bile acids) and by
Experimental studies indicate that the aging gas- necrotizing agents (eg, boiling water, absolute alcohol,
tric mucosa has impaired mucosal defense, eg, decreased 0.6 mol/L HCl, 0.2 mol/L NaOH).4,5 Andre Robert and
mucus and bicarbonate secretion and reduced prosta- colleagues demonstrated that pretreatment of rats with a
glandin generation.151,152 Cryer et al demonstrated that minute amount (0.1 to 1 ␮g/kg) of 16,16 dimethyl PGE2
aging reduces gastric and duodenal PG concentrations in given orally or subcutaneously 15–30 minutes prior to
humans.153 Other studies showed that aging gastric mu- intragastric instillation of 100% alcohol or boiling water
cosa has reduced NO synthase activity and impaired completely prevents severe gastric mucosal necrosis from
sensory nerve response to luminal acid.152,154 –156 Further- occurring.4 This remarkable protection is accomplished
more, in experimental models, aging increases the sus- by mechanisms other than reduction of the gastric acid
ceptibility of gastric mucosa to injury by a variety of secretion, as indicated by the fact that certain PGs protect
damaging agents, such as NSAIDs, ethanol, and hyper- the gastric mucosa in nonantisecretory doses and that
tonic solutions,152,156,157 and impairs healing of both even elimination of luminal acid with antisecretory
acute injury and chronic gastric ulcers.151,155,158 agents does not prevent necrosis.4 This novel concept
A recent study159 of aging rats demonstrated that gas- sparked a dramatic expansion in research. The cytopro-
tric mucosal blood flow is decreased by 60% with hypoxia tective action of PGs was found to apply to the human
and atrophy of glandular cells. In addition, expression of gastric and duodenal mucosa170 –172 as well as other organ
multifunctional phosphatase and tensin homolog de- systems (eg, liver, pancreas, kidney, blood vessels, and
leted on chromosome 10 (PTEN) and activated heart).170 Endoscopic studies with assessment of mucosal
caspases-3 and -9 was increased, whereas survivin was appearance, histology, and ultrastructure demonstrated
reduced, all resulting in increased apoptosis. Further- that 16,16 dimethyl PGE2 effectively protects the human
more, this study demonstrated increased susceptibility of gastric and duodenal mucosa against injury produced by
aging gastric mucosa to ethanol injury and that down- concentrated (40%– 60%) alcohol.171,172
regulation of elevated PTEN in gastric mucosa of aging Since the first demonstration of cytoprotection by PGs,
rats completely reverses this increased susceptibility. This other agents also have been shown to be cytoprotective.
study also demonstrated increased PTEN expression and They include sulfhydryl agents; growth factors; and pep-
reduced survivin in aging human gastric mucosa. These tides such as EGF, TGF-␣, TFFs, gastrin, cholecystokinin,
experimental observations may explain at least in part the thyrotropin-releasing hormone, bombesin, CRF, peptide
well-described increase in NSAID-associated ulcers and YY, neurokinin A, somatostatin, leptin, ghrelin, NO, hy-
ulcer complications with increasing age. drogen sulfide, agonists of adenosine and agonists of
proteinase activated receptor-1, and topically active anti-
Portal hypertensive gastropathy ulcer drugs such as sucralfate, aluminum-containing ant-
Experimental studies indicate that the gastric mu- acids, and rebamipide.19,173 Aluminum-containing antac-
cosa of individuals with portal hypertension has im- ids (eg, hydrotalcite) have been shown to exert a potent
paired mucosal defense, reduced oxygenation, increased mucosal protective action against a variety of injurious
susceptibility to injury, and impaired healing,160 –169 sim- factors including ethanol, NSAIDs, pepsin, bile acids,
ilar to that of the aging mucosa. Capillary endothelial stress, and ischemia/reperfusion.174,175 These actions are
abnormalities in portal hypertensive gastropathy result due to activation of PG synthesis; binding to and inacti-
in a 3.5-fold reduction in mucosal capillary lumen diam- vation of pepsin, lysolecithin, bile acids and H pylori
eter168 causing diminished capillary blood flow and re- toxins; activation of heat shock proteins; and activation
duced mucosal oxygenation.167,168 Also, activation of the of genes encoding EGF, basic fibroblast growth factor,
TNF-␣ gene and overexpression of endothelial NO syn- and their receptors.174 –176 Other topically active antiulcer
thase occurs with increased formation of toxic peroxyni- drugs may have similar modes of action.
52 LAINE, TAKEUCHI, AND TARNAWSKI GASTROENTEROLOGY Vol. 135, No. 1
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

Table 2. Metaanalyses of Randomized Controlled duction of NSAID-induced gastric ulcers but do decrease
Endoscopic Trials for Prevention of duodenal ulcers (Table 2).178 Double-dose H2RA therapy
NSAID-Associated Ulcers does significantly decrease NSAID gastric ulcers.178 Ran-
Gastric ulcer Duodenal ulcers domized controlled outcome trials to assess clinical
Misoprostol Cotherapy 0.26 (0.17⫺0.39) 0.47 (0.33⫺0.69) events with H2RAs have not been performed.
H2 receptor antagonist 0.73 (0.50⫺1.09) 0.36 (0.18⫺0.74) PPIs. PPIs significantly decrease NSAID-induced
(standard dose) gastric and duodenal ulcers vs placebo (Table 2).178 A
cotherapy randomized comparison after healing of NSAID ulcers
H2 receptor antagonist 0.44 (0.26⫺0.74) 0.26 (0.11⫺0.65)
(or ⬎10 erosions) revealed significantly lower 6-month
(double dose)
cotherapy ulcer incidences for omeprazole 20 mg daily than raniti-
Proton pump inhibitor 0.40 (0.32⫺0.51) 0.19 (0.09⫺0.37) dine 150 mg twice daily: gastric ulcers, 5% vs 16%; duo-
cotherapy denal ulcers, 0.5% vs 4%, respectively.180 PPIs are superior
COX-2 selective inhibitor 0.21 (0.18⫺0.25) 0.34 (0.25⫺0.45) to misoprostol in prevention of NSAID duodenal ulcers,
NOTE. Relative risk (95% CI) of traditional NSAIDS ⫹ medical co- but, when used at high dose (200 ␮g 4 times daily),
therapy or of COX-2 selective inhibitors vs traditional NSAIDs alone misoprostol was superior to PPI therapy for prevention
(80,178). of gastric ulcers in a randomized trial.178,181
No randomized controlled trial of PPIs for upper GI
Increased production of endogenous PG by mild irri- clinical events in a general population of NSAID users
tants (eg, 20% ethanol, capsaicin) may induce adaptive has been performed. However, trials in high-risk patients
cytoprotection and thereby decrease injury from more hospitalized with NSAID-associated bleeding ulcers are
severe irritants administered subsequently.5 This action is available. After ulcer healing, Chan et al182 resumed
mediated through EP-1 receptors and is blocked by COX NSAID therapy with naproxen 500 mg twice daily in 150
inhibition with indomethacin and by an EP-1 receptor such patients with H pylori infection and randomly as-
antagonist.177 signed them to maintenance PPI therapy or H pylori
eradication therapy. At 6 months, the rate of recurrent
Reduction of NSAID-Associated Injury and ulcer bleeding was significantly lower in the PPI group
Clinical Events (4% vs 19%, respectively). A randomized placebo-con-
Strategies designed to decrease upper GI tract trolled trial demonstrated that PPI therapy decreased
injury because of NSAIDs target either the decrease in recurrent ulcer complications (bleeding) in low-dose as-
mucosal defense related to NSAID-induced inhibition of pirin users who had presented with ulcer complications
PG production (eg, misoprostol cotherapy, COX-2 selec- (2% vs 15%, respectively, at median follow-up of 12
tive inhibitors) or the acid that is required to cause gross months).183
injury in the presence of an impaired mucosal defense COX-2 Selective Inhibitors. Endoscopic random-
(eg, H2RAs, PPIs). ized controlled trials reveal that COX-2 selective inhibitors
Medical Cotherapy. Misoprostol. Misoprostol is a (coxibs) significantly decrease gastroduodenal ulcers as
synthetic PGE1 analog designed to help overcome the compared with nonselective NSAIDs (Table 2).80 Ulcer rates
NSAID-induced deficiency of PGs in the gastric mu- were significantly decreased with all 5 coxibs (celecoxib,
cosa. Misoprostol also has antisecretory properties. A rofecoxib, valdecoxib, etoricoxib, lumiracoxib) and when
metaanalysis of randomized controlled trials found coxibs were tested against any of the most commonly used
that misoprostol significantly decreases NSAID-associ- NSAIDs (ibuprofen, naproxen, or diclofenac).
ated gastric and duodenal ulcers vs placebo (Table 2); Large randomized controlled outcome trials demon-
efficacy increases with increasing daily doses from 400 strate a significant reduction in upper GI complications
to 800 ␮g.178 Misoprostol is superior to H2RAs for and overall upper GI clinical events with coxibs vs tradi-
prevention of gastric but not duodenal ulcers.178 A GI tional NSAIDs (Table 3): a recent metaanalysis revealed a
outcomes study in 8848 rheumatoid arthritis patients RR of 0.39 (95% CI: 0.31– 0.50) for upper GI complica-
followed for up to 6 months revealed a significant tions with a 0.4% absolute risk reduction. Interestingly,
decrease in complicated ulcers (RR, 0.49; 95% CI: 0.27– when diclofenac is the comparator traditional NSAID,
0.89; number needed to treat [NNT], 263) and overall outcome studies have shown a significant benefit in over-
clinical events (RR, 0.46; 95% CI: 0.29 – 0.73; NNT, all upper GI events but not complicated events.80,88,89,184
139).87 In the United States, misoprostol represents This may be due to the lack of effective antiplatelet
only 2% of medical cotherapy prescriptions for NSAID activity in most patients taking diclofenac.
users179; this low rate may be due to misoprostol’s GI
adverse effects, such as diarrhea and abdominal dis- Medical Cotherapy vs COX-2 Selective
comfort,178 and the need for multiple daily doses. Inhibitor to Prevent UGI Complications
Histamine H2-receptor antagonists. Standard doses of A traditional NSAID plus PPI has been compared
H2RAs are not significantly better than placebo for re- with celecoxib in patients presenting with NSAID-asso-
July 2008 GASTRIC MUCOSAL DEFENSE AND CYTOPROTECTION 53

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
Table 3. Upper GI Complications and Clinical Events From Randomized Outcome Trials of COX-2 Selective Inhibitors vs
Traditional NSAIDs
CLASS VIGOR TARGET SUCCESS MEDAL
(n ⫽ 3981)88,89 (n ⫽ 4029)90 (n ⫽ 9127)91 (n ⫽ 13,274)204 (n ⫽ 34,701)184
Type of arthritis Osteoarthritis (73%); Rheumatoid Osteoarthritis Osteoarthritis Osteoarthritis (72%);
rheumatoid (27%) rheumatoid (28%)
COX-2 selective inhibitor Celecoxib 400 mg Rofecoxib 50 mg Lumiracoxib 400 Celecoxib 100, Etoricoxib 60, 90
twice daily daily mg daily 200 mg twice mg daily
daily
Traditional NSAID Ibuprofen 800 mg 3 Naproxen 500 mg Ibuprofen 800 mg Naproxen 500 mg Diclofenac 50 mg 3
times daily; twice daily 3 times daily; twice daily, times daily, 75
diclofenac 75 mg naproxen 500 diclofenac 50 mg twice daily
twice daily mg twice daily mg twice daily
Low-dose aspirin use 22% Exclusion criterion 24% 7% 33%
PPI use Exclusion criterion Exclusion criterion Exclusion criterion Exclusion criterion 40%
Median follow-up 9 mo 9 mo 12 mo 12 wk Mean, 18 mo
Upper GI complications
RR (95% CI) 0.77 (0.41⫺1.46) 0.43 (0.24⫺0.78) 0.34 (0.22⫺0.52) 0.14 (0.03⫺0.69) 0.91 (0.67⫺1.24)
NNT — 128 (1 yr) 120 (1 yr) 714 (12 wk) —
Upper GI clinical events
RR (95% CI) 0.66 (0.45⫺0.98) 0.46 (0.33⫺0.64) 0.46 (0.36⫺0.60) 0.50 (0.26⫺0.96) 0.69 (0.57⫺0.83)
NNT 111 (1 yr) 41 (1 yr) 64 (1 yr) 476 (12 wk) 333 (1 yr)

NOTE. Clinical events include complications plus uncomplicated symptomatic ulcers.


PPI, proton pump inihibitor; NNT, number-needed-to-treat (at specific time period based on individual study).

ciated gastroduodenal ulcer bleeding in 2, 6-month ran- protective cotherapy, these agents enhance mucosal de-
domized controlled trials.185,186 Differences in recurrent fense (eg, sucralfate, misoprostol) or decrease the effects
ulcer complications between the 2 groups were not sig- of gastric acid (eg, antacids, H2RAs, PPIs). Antacids were
nificant: celecoxib rates were 1.5% and 2.6%, respectively, among the earliest medications used. Metaanalysis of
lower than the traditional NSAID plus PPI. Most impor- randomized controlled trials failed to show a significant
tantly, the rates of recurrent ulcer complications were decrease in overt or clinically important bleeding, and the
unacceptably high in both groups. Chan et al found frequent large volumes required were inconvenient and
recurrent ulcer bleeding in 4.9% and 6.4%, respectively, of potentially could increase risk of aspiration.123 Intrave-
patients in the 2 treatment arms at 6 months, with an nous administration of H2RAs significantly decreased
additional 19% and 26%, respectively, having endoscopic overt bleeding (OR, 0.58; 95% CI: 0.42– 0.79) and clini-
ulcers identified.185,187 These results suggest that very cally important bleeding (OR, 0.44; 95% CI: 0.22– 0.88) as
high-risk patients should receive a coxib plus a PPI. A compared with placebo or no therapy.123 Metaanalysis of
recent randomized controlled trial in patients presenting sucralfate vs placebo/no therapy showed a decrease in
with NSAD-associated ulcer bleeding assessed this strat- overt bleeding (OR, 0.58; 95% CI: 0.34 – 0.99), although
egy by comparing celecoxib alone vs celecoxib plus PPI
limited data show no decrease in clinically important
after ulcer healing.188 The incidence of recurrent ulcer
bleeding.123,189
bleeding at 1 year was 9% with celecoxib alone vs 0% with
The randomized trials done in the 1970s to the 1990s
combined therapy.
were small and of variable methodologic quality with
Summary. Misoprostol, PPIs, and COX-2 selec-
variable definitions of bleeding, and metaanalysis of these
tive inhibitors all significantly decrease the risk of ulcers
diverse and heterogeneous trials is no substitute for a
and ulcer complications associated with NSAID use.
Strategies to decrease NSAID-induced GI complications randomized controlled trial of proper sample size. A
should be employed in patients at increased risk for GI landmark multicenter Canadian double-blind random-
clinical events: older age (eg, ⬎65 years); prior upper GI ized trial comparing ranitidine and sucralfate in 1200
clinical event; concomitant use of corticosteroid, antico- patients was published in 1998. Medical-surgical ICU
agulant, or antithrombotic (eg, aspirin). Patients at very patients requiring mechanical ventilation ⱖ48 hours
high-risk for ulcer complications, such as those with were randomly assigned to ranitidine 50 mg intrave-
prior ulcer complications or multiple GI risk factors, nously every 8 hours or sucralfate 1 g every 6 hours via
should receive a coxib plus a PPI. nasogastric tube.190 Clinically important bleeding was
significantly lower with ranitidine (1.7% vs 3.8%, respec-
Reduction in SRMD tively [RR, 0.44; 95% CI: 0.21– 0.92]) with no difference in
A variety of agents have been used in an attempt mortality (23.5% vs 22.8%, respectively [RR, 1.03; 95% CI:
to decrease SRMD-related bleeding. Similar to NSAID 0.84 –1.26]) (Figure 5).
54 LAINE, TAKEUCHI, AND TARNAWSKI GASTROENTEROLOGY Vol. 135, No. 1
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

patients were receiving enteral nutrition in that study, rani-


tidine was still associated with a significant decrease in
bleeding.
PPIs have been used increasingly for stress prophylaxis,
despite very limited trial data supporting their use in past
years. A recent large double-blind randomized trial com-
pared PPI (immediate-release suspension via nasogastric
tube) and constant infusion H2RA in 359 patients who
required mechanical ventilation for ⱖ48 hours and had 1
additional risk factor.199 Significantly fewer patients had
overt bleeding with the PPI (19% vs 32%, respectively), but
there was no significant difference in persistent bleeding
(3.9% vs 5.5%, respectively), transfusion-requiring bleed-
ing (2.8% vs 2.8%, respectively), nosocomial pneumonia
Figure 5. Results of a double-blind randomized trial of ranitidine intra- (11.2% vs 9.4%, respectively), or death (15.2% vs 11.6%,
venously vs. sucralfate via nasogastric tube in 1200 intensive care unit
respectively); bleeding was not the cause of death in any
patients requiring mechanical ventilation ⱖ48 hours.162
patient.
Limited information is available for use of misopros-
An additional concern with stress prophylaxis relates tol. No placebo-controlled trials of misoprostol for pre-
to the development of ventilator-associated pneumonia, vention of stress-induced ulcers are available, but trials
a common and serious problem in critically ill patients. with antacid and H2RA controls show no significant
Gastric colonization with gram-negative bacilli is sug- differences in outcomes assessed.200 –202
gested to play a causal role, and an increase in gastric pH Summary. Preventive cotherapy aimed at decreas-
increases gastric colonization.191 However, an association ing SRMD bleeding should not be used in all patients in
of gastric organisms with ventilator-assisted pneumonia the ICU but only in those at high risk for stress bleeding,
is not identified in all studies.192–194 Furthermore, meta- eg, severe trauma, burns ⬎ one third of the body surface
analyses of randomized controlled trials failed to show a area, major intracranial disease, severe illness requiring
significant increase in pneumonia with H2RAs (RR, 1.25; mechanical ventilation ⬎48 hours. Intravenous H2RAs
95% CI: 0.78 –2.00) or sucralfate (RR, 2.11; 95% CI: 0.82– or PPIs via nasogastric tube are appropriate therapies,
5.44) vs placebo/no therapy, although there was a trend although the absolute risk reduction with these agents in
to more pneumonias with H2RAs than sucralfate (RR, modern ICUs is likely small. If the rate of clinically
1.19; 95% CI: 0.98 –1.44).123 However, methodologic de- important bleeding is 5% or less, then a 50% RR reduc-
ficiencies and small sample sizes made a large prospective tion with prophylactic therapy means that 40 patients or
randomized trial necessary to confirm or refute these more will need to be treated to prevent 1 additional
findings. The large multicenter trial mentioned above episode of clinically important bleeding—and little or no
carefully assessed ventilator-associated pneumonia and benefit in mortality can be expected.203
found no significant difference (19.1% vs 16.2%, respec-
tively [RR, 1.18; 95% CI: 0.92–1.51]).190 These results are Future Directions
very similar to those of the earlier metaanalysis and do In the clinical arena, knowledge regarding gas-
not rule out a small RR increase with antisecretory ther- tric mucosal defense mechanisms has led to the devel-
apy compared with sucralfate. opment of current and potential future therapies to
Based on the results of the large trial, intravenous reduce NSAID gastrointestinal injury, including miso-
H2RAs have generally been recommended for preventive prostol, coxibs, NO-NSAIDs, H2S-NSAIDs, and phos-
therapy in high-risk patients. However, some authors phatidylcholine-NSAIDs. Future pharmacogenomic
have suggested, because the rate of clinically important studies may help better stratify patients for their risk
bleeding is very low and the benefit of therapy uncertain of NSAID complications (eg, polymorphisms influenc-
in recent studies, that prophylaxis is unnecessary.189,195–197 ing drug metabolism and COX activity). Gastric mu-
In addition, if nosocomial pneumonia is increased with cosal defense mechanisms also may be applicable to
antisecretory therapy, this may offset some or all of the the esophagus, intestine, and extraintestinal tissues.
benefit because of the decrease in bleeding. Furthermore, For example, a PGE2 analog effectively protects liver,
early enteral nutrition is suggested to decrease the risk of pancreas, myocardium, and other tissues from injury.
stress-related bleeding, although it also increases gastric pH Other important areas for future research include bac-
and colonization as well as gastric volume.197,198 In the terial-epithelial interactions through toll-like and
multicenter Canadian trial, enteral nutrition was a signifi- other receptors; epithelial/endothelial cell interactions
cant independent predictor of decreased bleeding risk (RR, with matrix through integrins, focal adhesion proteins,
0.30; 95% CI: 0.13– 0.67).117 However, regardless of whether and cytoskeletal elements; and mucosal immune re-
July 2008 GASTRIC MUCOSAL DEFENSE AND CYTOPROTECTION 55

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
sponse. Future studies will allow a better recognition 18. Gendler S, Spicer AP. Epithelial mucin genes. Annu Rev Physiol
of cellular and subcellular defensive mechanisms, such 1995;57:607– 634.
19. Ho SB, Takamura K, Anway R, et al. The adherent gastric
as ATP-ase pumps; K⫹ and Ca⫹⫹ channels in cell mem-
mucous layer is composed of alternating layers of MUC5AC and
branes; mitochondrial membrane potential and perme- MUC6 mucin proteins. Dig Dis Sci 2004;49:1598 –1606.
ability; activation of pro- and antiapoptotic pathways, 20. Newton J, Allen A, Westley BM, et al. The human trefoil peptide,
including PI-3K and survivin prosurvival signaling and TFFl, is present in different molecular forms that are intimately
PTEN-induced proapoptotic signaling; intracellular associated with the adherent mucus gel in normal stomach. Gut
pH, hypoxia, and oxygen cellular sensing mechanisms; 2000;46:312–320.
21. Poulsom R, Wright NA. Trefoil peptides: a newly recognized
and nuclear transport of transcription factors by im- family of epithelial mucin associated molecules. Am J Physiol
portins, as well as genetic regulation of these mecha- Gastrointest Liver Physiol 2002;282:G211–G219.
nisms. Finally, optical devices such as confocal endo- 22. Thim L, Madsen F, Poulsen SS. Effect of trefoil factors on the
microscopy, optical coherence tomography, Mauna- viscoelastic properties of mucus gels. Eur J Clin Invest 2002;
Kea probe, and multiphoton microscopy will allow 32:519 –527.
23. Darling RL, Romero JJ, Dial EJ, et al. The effects of aspirin on
real-time assessment of cellular structures and molec-
gastric mucosal integrity, surface hydrophobic, and prostaglan-
ular imaging of specific targets involved in human din metabolism in cyclooxygenase knockout mice. Gastroenter-
gastric mucosal defense. ology 2004;127:94 –104.
24. Flemström G. Active alkalinization by amphibian gastric fundic
mucosa. Am J Physiol Endocrinol Metab Gastrointest Physiol
References
1977;233:E1–E12.
1. Hunter J. Digestion of the stomach after death. Philos Trans R 25. Rossmann H, Bachmann O, Vieillard-Baron D, et al. Na⫹/
Soc Lond 1772;62:447– 454. HCO3⫺ cotransport and expression of NBC1 and NBC2 in rabbit
2. Virchow R. Historisches, kritisches und positives zur lehre der gastric parietal and mucous cells. Gastroenterology 1999;116:
unterleibsaffektionen. Arch Pathol Anat 1853;5:281–375. 1389 –1398.
3. Vane JR. Inhibition of prostaglandin synthesis as a mechanism 26. Takeuchi K, Yagi K, Kato S, et al. Roles of prostaglandin E-re-
of action for aspirin-like drugs. Nat N Biol 1971;232:232–235. ceptor subtypes in gastric and duodenal bicarbonate secretion
4. Robert A, Nezamis JE, Lancaster C, et al. Cytoprotection by in rats. Gastroenterology 1997;113:1553–1559.
prostaglandins in rats. Prevention of gastric necrosis produced 27. Tanaka D, Tsutsumi S, Arai Y, et al. Genetic evidence for a
by alcohol, HCl, NaOH, hypertonic NaCl and thermal injury. Gas- protective role of heat shock factor 1 against irritant-induced
troenterology 1978;77:433– 443. gastric lesions. Mol Pharmacol 2007;71:985–993.
5. Robert A, Nezamis JE, Lancaster C, et al. Mild irritants prevent 28. Oyaka J, Otaka M, Matsuhashi T, et al. Over-expression of
gastric necrosis through “adaptive cytoprotection” mediated by 70-kDa heat shock protein confers protection against monochlo-
prostaglandins. Am J Physiol 1983;245:G113–G121.
ramine-induced gastric mucosal cell injury. Life Sci 2006;79:
6. Werther JL. The gastric mucosal barrier. Mt Sinai J Med 2000;
300 –305.
67:41–53.
29. Tarnawski A, Hollander D, Gergely H, et al. Comparison of
7. Montrose MH, Yasutada A, Takeuchi K, et al. Gastroduodenal
antacid, sucralfate, cimetidine and ranitidine in protection of the
mucosal defense. In: Johnson LR, ed. New York: Academic
gastric mucosa against ethanol injury. Am J Med 1985;79:19 –
Press, 2006:1259 –1291.
23.
8. Lichtenberger LM. Gastroduodenal mucosal defense. Curr Opin
30. Tarnawski A, Wang H, Tomikawa M, et al. Talcid triggers induction
Gastroenterol 1999;15:463– 472.
of heat shock proteins HSP-70 in gastric mucosa: a key to its
9. Tarnawski A. Cellular and molecular mechanisms of mucosal
mucosal protective action? Gastroenterology 1999;116:A331.
defense and repair. In: Yoshikawa T, Arakawa T. Bioregulation
31. Yang YH, Wu WK, Tai EK, et al. The cationic host defense
and its disorders in gastrointestinal tract. Tokyo, Japan: Black-
peptide rCRAMP promotes gastric ulcer healing in rats. J Phar-
well Science, 1998:3–17.
10. Ham M, Kaunitz JD. Gastroduodenal defense. Curr Opin Gastro- macol Exp Ther 2006;318:547–554.
enterol 2007;23:607– 616. 32. Taupin D, Podolsky DK. Trefoil factors initiators of mucosal
11. Szabo S. Mechanisms of gastric mucosal injury and protection. healing. Nat Rev Mol Cell Biol 2003;4:721.
J Clin Gastroenterol 1991;13:S21–S34. 33. Modlin IM, Kidd M, Lye KD, et al. Gastric stem cells: an update.
12. Kobayashi K, Arakawa T. Arachidonic acid cascade and gastric Keio J Med 2003;52:134 –137.
mucosal injury, protection, and healing: topics of this decade. 34. Leedham SJ, Brittan M, Preston SL, et al. The stomach periglan-
J Clin Gastroenterol 1995;21(Suppl 1):S12–S17. dular fibroblast sheath: all present and correct. Gut 2006;55:
13. Allen A. Structure of gastrointestinal mucus and the viscous and 295–296.
gel-forming properties of mucus. Br Med Bull 1978;34:28 –33. 35. Svanes K, Ito S, Takeuchi K, et al. Restitution of the surface
14. Allen A, Flemström G. Gastroduodenal mucus bicarbonate bar- epithelium of the in vitro frog gastric mucosa after damage with
rier: protection against acid and pepsin. Am J Physiol Cell hyperosmolar sodium chloride: morphologic and physiologic
Physiol 2005;288:C1–C19. characteristics. Gastroenterology 1982;82:1409 –1426.
15. Atuma C, Strugala V, Allen A, et al. The adherent gastric mucus 36. Lacy ER, Ito S. Rapid epithelial restitution of the rat gastric
gel layer: thickness and physical state in vivo. Am J Physiol mucosa after ethanol injury. Lab Invest 1984;51:573–583.
Gastrointest Liver Physiol 2001;280:G922–G929. 37. Tarnawski A, Hollander D, Stachura J, et al. Prostaglandin pro-
16. Hills BA, Butler BD, Lichtenberger LM. Gastric mucosal barrier: tection of the gastric mucosa against alcohol injury—a dynamic
hydrophobic lining to the lumen of the stomach. Am J Physiol time-related process. The role of mucosal proliferative zone.
1983:244:G561–G568. Gastroenterology 1985;88:334 –359.
17. Jordan N, Newton J, Pearson J, et al. A novel method for the 38. Tarnawski A, Stachura J, Durbin T, et al. Increased expression of
visualization of the in situ mucus layer in rat and man. Clin Sci epidermal growth factor receptor during gastric ulcer healing in
(Lond) 1998;95:97–106. rats. Gastroenterology 1992;102:695– 698.
56 LAINE, TAKEUCHI, AND TARNAWSKI GASTROENTEROLOGY Vol. 135, No. 1
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

39. Nguyen T, Chai J, Tanigawa T, et al. Novel roles of local IGF-1 rat stomachs. Aliment Pharmacol Ther 2000;14(Suppl
activation in rat gastric ulcer healing: promotes actin polymer- 1):18 –25.
ization, cell proliferation, reepithelialization and induces COX-2 60. Takeuchi K, Komoike Y, Takeeda M, et al. Gastric mucosal
in a PI3K-dependent manner. Am J Pathol 2007;170:1219 – ulcerogenic responses following barrier disruption in knockout
1228. mice lacking prostaglandin EP1 receptors. Aliment Pharmacol
40. Pai R, Soreghan BA, Szabo IL, et al. Prostaglandin E2 transac- Ther 2002;16:74 – 82.
tivates EGF receptor: a novel mechanism for promoting colon 61. Takahashi S, Takeuchi K, Okabe S. EP4 receptor mediation of
cancer growth and gastrointestinal hypertrophy. Nat Med 2002; prostaglandin E2-stimulated mucus secretion by rabbit gastric
8:289 –293. epithelial cells. Biochem Pharmacol 1999;58:1997–2002.
41. Chiou SK, Tanigawa T, Akahoshi T, et al. Survivin—a novel 62. Kato S, Aihara E, Yoshii K, et al. Dual action of prostaglandin E2
target for indomethacin-induced gastric injury. Gastroenterology on gastric acid secretion through different EP receptor subtypes
2005;128:63–73. in the rat. Am J Physiol 2005;89:G64 –G69.
42. Gannon B, Browning J, O’Brien P, et al. Mucosal microvascular 63. Tanaka S, Tache Y, Kaneko H, et al. Central vagal activation
architecture of the fundus and body of the human stomach. increases mucous gel thickness and surface cell intracellular
Gastroenterology 1984;86:866 – 875. pH in rat stomach. Gastroenterology 1997;122:409 – 417.
43. Guth PH. Current concepts in gastric microcirculatory patho- 64. Chatzaki E, Lambropoulou M, Constantinidis TC, et al. Cortico-
physiology. Yale J Biol Med 1992;65:677– 688. tropin-releasing factor (CRF) receptor type 2 in the human stom-
44. Peskar BM. Neural aspects of prostaglandin involvement in ach: protective biological role by inhibition of apoptosis. J Cell
gastric mucosal defense. J Physiol Pharmacol 2001;52:555– Physiol 2006;209:905–911.
568. 65. Kaneko H, Taché Y, Kusugami K. Importance of medullary thy-
45. Holzer P. Neural regulation of gastrointestinal blood flow. In: rotropin-releasing hormone in brain-gut circuits regulating gas-
Johnson LR, ed. Physiology of the gastrointestinal tract. 4th ed. tric integrity: preclinical studies. J Gastroenterol 2002;37(Suppl
New York: Academic Press, 2006:817– 839. 14):128 –132.
46. Fiorucci S, Antonelli E, Distrutti E, et al. Inhibition of hydrogen 66. Gyires K, Müllner K, Rónai AZ. Activation of central opioid re-
sulfide generation contributes to gastric injury caused by anti- ceptors may induce gastric mucosal defense in the rat. J Physiol
inflammatory nonsteroidal drugs. Gastroenterology 2005;129: Paris 2001;95:189 –196.
1210 –1224.
67. Mózsik G, Karádi O, Király A, et al. The key-role of vagal nerve
47. Fiorucci S, Distrutti E, Cirino G, et al. The emerging roles of
and adrenals in the cytoprotection and general gastric mucosal
hydrogen sulfide in the gastrointestinal tract and liver. Gastro-
integrity. J Physiol Paris 2001;95:229 –237.
enterology 2006;131:259 –271
68. Szabo S, Trier J. Pathogenesis of acute gastric mucosal injury:
48. Tarnawski A, Sarfeh IJ, Lu S-Y, et al. Quality of ulcer healing:
sulfhydryls as a protector, adrenal cortex as a modulator, and
evidence for impaired restoration of sensory CGRP nerves in the
vascular endothelium as a target. In: Allen A, ed. Mechanisms
scar of experimental gastric ulcer. Eur J Gastroenterol and
of mucosal protection in the upper gastrointestinal tract. New
Hepatol 1993;S81–S85.
York: Raven Press, 1984:287–293.
49. Holzer P. Role of visceral afferent neurons in mucosal inflam-
69. Szabo S. Peptides, sulfhydryls, and glucocorticoids in gastric
mation and defense. Curr Opin Phamacol 2007;7:563–569.
mucosal defense: coincidence or connection? Gastroenterology
50. Stroff T, Plate S, Respondek M, et al. Protection by gastrin in the
1984;87:228 –229.
rat stomach involves afferent neurons, calcitonin gene-related
70. Brzozowski T, Konturek PC, Drozdowicz D, et al. Role of central
peptide, and nitric oxide. Gastroenterology 1995;109:89 –97.
and peripheral ghrelin in the mechanism of gastric mucosal
51. Atay S, Tarnawski AS, Dubois A. Eicosanoids and the stomach.
defence. Inflammopharmacology 2005;13:45– 62.
Prostaglandins Other Lipid Mediat 2000;1:105–124.
52. Halter F, Schmassmann A, Peskar BM, et al. Cyclooxygenase-2 71. Filaretova LP, Filaretov AA, Makara GB. Corticosterone increase
implications on maintenance of gastric mucosal integrity and inhibits stress-induced gastric erosions in rats. Am J Physiol
ulcer healing: controversial issues and perspectives. Gut 2001; Gastrointest Liver Physiol 1998;274:G1024 –G1030.
49:443– 453. 72. Filaretova L, Bagaeva T, Podvigina T, et al. Various ulcerogenic
53. Brzozowski T, Konturek PC, Konturek SJ, et al. Role of prosta- stimuli are potentiated by glucocorticoid deficiency in rats.
glandins in gastroprotection and gastric adaptation. J Physiol J Physiol Paris 2001;95:59 – 65.
Pharmacol 2005;56(Suppl 5):33–55. 73. Filaretova L, Podvigina T, Bagaeva T, et al. Gastroprotective role
54. Lichtenberger LM, Graziani LA, Dial EJ, et al. Role of surface- of glucocorticoid hormones. J Pharmacol Sci 2007;104:195–
active phospholipids in gastric cytoprotection. Science 1983; 201.
219:1327–1329. 74. Kaufman DW, Kelly JP, Rosenberg L, et al. Recent patterns of
55. Redfern JS, Lee E, Feldman M. Effect of immunization with medication use in the ambulatory adult population of the United
prostaglandin metabolites on gastrointestinal ulceration. Am J States: the Slone survey. JAMA 2002;287:337–344
Physiol 1988;255:G723–G730. 75. Paulose-Ram R, Hirsch R, Dillon C, et al. Frequent monthly use
56. Redfern JS, Feldman M. Role of endogenous prostaglandins of selected non-prescription and prescription non-narcotic anal-
in preventing gastrointestinal ulceration: induction of ulcers gesics among U.S. adults. Pharmacoepidemiol Drug Saf 2005;
by antibodies to prostaglandins. Gastroenterology 1989;96: 14:257–266.
596 – 605. 76. Laine L. Nonsteroidal anti-inflammatory drug gastropathy. Gas-
57. Narumiya S, Sugimoto Y, Ushikubi F. Prostanoid receptors: trointest Endosc Clin N Am 1996;6:489 –504.
structures, properties, and functions. Physiol Rev 1999;79: 77. Baskin WN, Ivey KJ, Krause WJ, et al. Aspirin-induced ultrastruc-
1193–1226. tural changes in human gastric mucosa. Correlation with poten-
58. Takeuchi K, Ukawa H, Kato S, et al. Impaired duodenal bicar- tial difference. Ann Intern Med 1976;85:299 –303.
bonate secretion and mucosal integrity in mice lacking prosta- 78. Strom BL, Berlin JA, Kinman JL, et al. Parenteral ketorolac and
glandin E receptor subtype EP3. Gastroenterology 1999;117: risk of gastrointestinal and operative site bleeding. A postmar-
1128 –1135. keting surveillance study. JAMA 1996;275:376 –382.
59. Araki H, Yagi K, Suzuki K, et al. Roles of prostaglandin E 79. Laine L, Harper S, Simon T, et al. A randomized trial comparing
receptor subtypes in cytoprotective action of prostaglandin E2 in the effect of rofecoxib, a COX-2 specific inhibitor, to ibuprofen on
July 2008 GASTRIC MUCOSAL DEFENSE AND CYTOPROTECTION 57

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
the gastroduodenal mucosa of osteoarthritis patients. 98. Vane JR, Botting RM. A better understanding of anti-inflamma-
Gastroenterology 1999;117:776 –783. tory drugs based on isoforms of cyclooxygenase (COX-1 and
80. Rostom A, Muir K, Dube C, et al. Gastrointestinal safety of COX-2). Adv Prostaglandin Thromboxane Leukot Res 1995;23:
cyclooxygenase-2 inhibitors: a Cochrane Collaboration System- 41– 48.
atic Review. Clin Gastroenterol Hepatol 2007;5:818 – 828 99. Wallace JL, McKnight W, Reuter BK, et al. NSAID-induced gastric
81. Lancaster-Smith MJ, Jaderberg ME, Jackson DA. Ranitidine in damage in rats: requirement for inhibition of both cyclooxygen-
the treatment of non-steroidal anti-inflammatory drug associ- ase 1 and 2. Gastroenterology 2000;119:706 –714.
ated gastric and duodenal ulcers. Gut 1991;32:252–255. 100. Tanaka A, Araki H, Hase S, et al. Inhibition of both COX-1 and
82. Mizuno H, Sakamoto C, Matsuda K, et al. Induction of cycloox- COX-2 is required for development of gastric damage in re-
ygenase 2 in gastric mucosal lesions and its inhibition by the sponse to nonsteroidal antiinflammatory drugs. J Physiol Paris
specific antagonist delays healing in mice. Gastroenterology 2001;95:21–27.
1997;112:387–397. 101. Tanaka A, Araki H, Hase S, et al. Roles of COX-1 and COX-2
83. Ukawa H, Yamakuni H, Kato S, et al. Effects of cyclooxygen- inhibition in nonsteroidal antiinflammatory drug-induced gastric
ase-2 selective and nitric oxide-releasing nonsteroidal antiin- damage in rats: relation to functional responses. Aliment Phar-
flammatory drugs on mucosal ulcerogenic and healing re- macol Ther 2002;16:90 –101.
sponses of the stomach. Dig Dis Sci 1998;43:2003–2011. 102. Filaretova L, Komoike Y, Tanaka A, et al. COX-2 inhibitor induces
84. Schmassmann A, Peskar BM, Stettler C, et al. Effects of inhi- gastric mucosal damage in adrenalectomized rats. Inflammo-
bition of prostaglandin endoperoxide synthase-2 in chronic gas- pharmacology 2002;10:413– 422.
tro-intestinal ulcer models in rats. Br J Pharm 1998;123:795– 103. Kato S, Ogawa Y, Kanatsu K, et al. Ulcerogenic influence of
804. selective cyclooxygenase (COX)-2 inhibitors in the rat stomach
85. Chan FKL, Wong VWS, Suen BY, et al. Effect of celecoxib on the with adjuvant-induced arthritis. J Pharmacol Exp Ther 2002;
healing of complicated gastric ulcers: a prospective, double- 303:503–509.
blinded, randomized trial. Gastroenterology 2005;128(Suppl 2): 104. Takeuchi K, Ueshima K, Hironaka Y, et al. Oxygen free radicals
A24 –A25. and lipid peroxidation in the pathogenesis of gastric mucosal
86. Hernandez-Diaz S, Rodriguez LA. Association between nonste- lesions induced by indomethacin in rats. Digestion 1991;49:
roidal anti-inflammatory drugs and upper gastrointestinal tract 175–184.
bleeding/perforation: an overview of epidemiologic studies pub-
105. Asako H, Kubes P, Wallace JL, et al. Indomethacin-induced
lished in the 1990s. Arch Int Med 2000;160:2093–2099.
leukocyte adhesion in mesenteric venules; role of lipoxygenase
87. Silverstein FE, Graham DY, Senior JR, et al. Misoprostol reduces
products. Am J Physiol 1992;262:G903–G908.
serious gastrointestinal complications in patients with rheuma-
106. Wallace JL, Keeman CM, Granger DN. Gastric ulceration in-
toid arthritis receiving nonsteroidal anti-inflammatory drugs. A
duced by non-steroidal anti-inflammatory drugs is a neutrophil-
randomized, double-blind, placebo-controlled trial. Ann Intern
dependent process. Am J Physiol 1995;259:G462–G467.
Med 1995;123:241–249.
107. Takeuchi K, Tanaka A, Hayashi Y, et al. COX inhibition and
88. Silverstein F, Faich G, Goldstein JL, et al. Gastrointestinal tox-
NSAID-induced gastric damage: the role in various pathogenic
icity with celecoxib vs nonsteroidal anti-inflammatory drugs for
events. Curr Top Med Chem 2005;5:371–381.
osteoarthritis and rheumatoid arthritis. The CLASS study: a
108. Mersereau WA, Hinchey EJ. Prevention of phenylbutazone ulcer
randomized controlled trial. JAMA 2000;284:1247–1255.
in the rat by glucose: role of a glycoprivic receptor system. Am J
89. FDA Arthritis Advisory Committee meeting. February 7, 2001.
Physiol 1982;242:G429 –G432.
Available at: http://www.fda.gov/ohrms/dockets/ac/01/briefing/
109. Takuechi K, Ueki S, Okabe S. Importance of gastric motility in
3677b1.htm. Accessed June 15, 2008.
the pathogenesis of indomethacin-induced gastric lesions in
90. Bombardier C, Laine L, Reicin A, et al. Comparison of upper
gastrointestinal toxicity of rofecoxib and naproxen in patients rats. Dig Dis Sci 1986;31:1114 –1121.
with rheumatoid arthritis. N Engl J Med 2000;343:1520 –1528. 110. Takeeda M, Yamato M, Kato S, et al. Cyclooxygenase
91. Schnitzer TJ, Burmester GR, Mysler E, et al. Comparison of isozymes involved in adaptive functional responses in rat
lumiracoxib with naproxen and ibuprofen in the Therapeutic stomachs following barrier disruption. J Pharmacol Exp Ther
Arthritis Research and Gastrointestinal Event Trial (TARGET), 2003;307:713–719.
reduction in ulcer complications: randomised controlled trial. 111. Husain SS, Szabo IL, Pai R, et al. MAP (ERK-2) kinase—a key
Lancet 2004;364:665– 674. target for NSAIDs-induced inhibition of gastric cancer cell pro-
92. Wallace JL, Granger DN. Pathogenesis of NSAID gastropathy; liferation and growth. Life Sci 2001;69:3045–3054.
are neutrophils the culprits? Trend Pharmacol Sci 1992;3:129 – 112. 85. Bias P, Buchner A, Klesser B, et al. The gastrointestinal
131. tolerability of the LOX/COX inhibitor, licofelone, is similar to
93. Feng L, Sun W, Xia Y, et al. Cloning two isoforms of rat cyclo- placebo and superior to naproxen therapy in healthy volunteers:
oxygenase: differential regulation of their expression. Arch Bio- results from a randomized, controlled trial. Am J Gastroenterol
chem Biophys 1993;307:361–368. 2004;99:611– 618.
94. Vane JR, Botting RM. New insights into the mode of action of 113. Fiorucci S, de Lima OC Jr, Mencarelli A, et al. Cyclooxygenase-
anti-inflammatory drugs. Inflamm Res 1995;44:1–10. 2-derived lipoxin A4 increases gastric resistance to aspirin-
95. O’Neill GP, Ford-Hutchinson AW. Expression of mRNA for cyclo- induced damage. Gastroenterology 2002;123:1598 –1606.
oxygenase-1 and cyclooxygenase-2 in human tissues. FEBS Lett 114. Fiorucci S, Santucci L, Wallace JL, et al. Interaction of a selec-
1993;330:156 –160. tive cyclooxygenase-2 inhibitor with aspirin and NO-releasing
96. Kargman S, Charlesion S, Cartwright M, et al. Characterization of aspirin in the human gastric mucosa. Proc Natl Acad Sci U S A
prostaglandin G/H synthase 1 and 2 in rat, dog, monkey, and 2003;100:10937–10941.
human gastrointestinal tracts. Gastroenterology 1993;111:445– 115. Laine L, Maller ES, Yu C, et al. Ulcer formation with low-dose
454. enteric-coated aspirin and the effect of COX-2 selective inhibition:
97. Whittle BJR. Temporal relationship between cyclooxygenase in- a double-blind trial. Gastroenterology 2004;127:395– 402.
hibition, as measured by prostacyclin biosynthesis, and the 116. Cook DJ, Fuller HD, Guyatt GH, et al. Risk factors for gastroin-
gastrointestinal damage induced by indomethacin in the rat. testinal bleeding in critically ill patients. Canadian Critical Care
Gastroenterology 1981;80:94 –98. Trials Group. N Engl J Med 1994;330:377–381.
58 LAINE, TAKEUCHI, AND TARNAWSKI GASTROENTEROLOGY Vol. 135, No. 1
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

117. Cook D, Heyland D, Griffith L, et al. Risk factors for clinically 135. Kamada T, Sato N, Kawano S, et al. Gastric mucosal hemodynam-
important upper gastrointestinal bleeding in patients requiring ics after thermal or head injury. A clinical application of reflectance
mechanical ventilation. Canadian Critical Care Trials Group. Crit spectrophotometry. Gastroenterology 1982;83:535–540.
Care Med 1999;27:2812–2817. 136. Leung FW, Itoh M, Hirabayashi K, et al. Role of blood flow in
118. Lucas CE, Sugawa C, Riddle J, et al. Natural history and surgical gastric and duodenal mucosal injury in the rat. Gastroenterology
dilemma of “stress” gastric bleeding. Arch Surg 1971;102: 1985;88:281–289.
266 –273. 137. Yasue N, Guth PH. Role of exogenous acid and retransfusion in
119. Czaja AJ, McAlhany JC, Pruitt BA Jr. Acute gastroduodenal dis- hemorrhagic shock-induced gastric lesions in the rat. Gastroen-
ease after thermal injury. An endoscopic evaluation of incidence terology 1988;94(Suppl 1):1135–1143.
and natural history. N Engl J Med 1974;291:925–929. 138. Ritchie WP Jr. Acute gastric mucosal damage induced by bile salts,
120. Peura DA, Johnson LF. Cimetidine for prevention and treatment acid, and ischemia. Gastroenterology 1975;68:699 –707.
of gastroduodenal mucosal lesions in patients in an intensive 139. Maricic N, Ehrlich K, Gretzer B, et al. Selective cyclo-oxygen-
care unit. Ann Int Med 1985;103:173–177. ase-2 inhibitors aggravate ischaemia-reperfusion injury in the
rat stomach. Br J Pharmacol 1999;128:1659 –1666.
121. Reusser P, Gyr K, Scheidegger D, et al. Prospective endoscopic
140. Schmedtje JF, Ji YS, Liu WL, et al. Hypoxia induces cyclooxy-
study of stress erosions and ulcers in critically ill neurosurgical
genase-2 via the NF-␬B p65 transcription factor in human vas-
patients: current incidence and effect of acid-reducing prophy-
cular endothelial cells. J Biol Chem 1997;272:601– 608.
laxis. Crit Care Med 1990;18:270 –274.
141. Björne H, Govoni M, Törnberg DC, et al. Intragastric nitric oxide
122. Spirt MJ. Stress-related mucosal disease: risk factors and pro-
is abolished in intubated patients and restored by nitrite. Crit
phylactic therapy. Clin Ther 2004;26:197–213.
Care Med 2005;33:1722–1727.
123. Cook DJ, Reeve BK, Guyatt GH, et al. Stress ulcer prophylaxis in
142. Michida T, Kawano S, Masuda E, et al. Endothelin-1 in the
critically ill patients. Resolving discordant meta-analyses. JAMA gastric mucosa in stress ulcers of critically ill patients. Am
1996;275:308 –314. Gastroenterol 1997;92:1177–1181.
124. Messori A, Trippoli S, Vaiani M, et al. Bleeding and pneumonia 143. Naito Y, Yoshikawa T, Matsuyama K, et al. Neutrophils, lipid
in intensive care patients given ranitidine and sucralfate for peroxidation, and nitric oxide in gastric reperfusion injury in rats.
prevention of stress ulcer: meta-analysis of randomised con- Free Radic Biol Med 1998;24:494 –502.
trolled trials. BMJ 2000;321:1103–1106. 144. Wada K, Kamisaki Y, Ohkura T, et al. Direct measurement of
125. Ruiz-Santana S, Ortiz E, Gonzalez B, et al. Stress-induced gas- nitric oxide release in gastric mucosa during ischemia-reperfu-
troduodenal lesions and total parenteral nutrition in critically ill sion in rats. Am J Physiol 1998;274:G465–G471.
patients: frequency, complications, and the value of prophylac- 145. Kobata A, Kotani T, Komatsu Y, et al. Dual action of nitric oxide
tic treatment. A prospective, randomized study. Crit Care Med in the pathogenesis of ischemia/reperfusion-induced mucosal
1991;19:887– 891. injury in mouse stomach. Digestion 2007;75:188 –197.
126. Apte NM, Karnad DR, Medhekar TP, et al. Gastric colonization 146. Skillman JJ, Gould SA, Chung RS, et al. The gastric mucosal
and pneumonia in intubated critically ill patients receiving barrier: clinical and experimental studies in critically ill and
stress ulcer prophylaxis: a randomized, controlled trial. Crit normal man, and in the rabbit. Ann Surg 1970;172:564 –584.
Care Med 1992;20:590 –593. 147. Fiddian-Green RG, McGough E, Pittenger G, et al. Predictive value
127. Metz CA, Livingston DH, Smith JS, et al. Impact of multiple risk of intramural pH and other risk factors for massive bleeding from
factors and ranitidine prophylaxis on the development of stress- stress ulceration. Gastroenterology 1983;85:613– 620.
related upper gastrointestinal bleeding: a prospective, multi- 148. Maynard N, Bihari D, Beale R, et al. Assessment of splanchnic
center, double-blind, randomized trial. The Ranitidine Head In- oxygenation by gastric tonometry in patients with acute circula-
jury Study Group. Crit Care Med 1993;21:1844 –1849. tory failure. JAMA 1993;270:1203–1210.
128. Hanisch EW, Encke A, Naujoks F, et al. A randomized, double- 149. Maynard ND, Bihari DJ, Dalton RN, et al. Increasing splanchnic
blind trial for stress ulcer prophylaxis shows no evidence of blood flow in the critically iII. Chest 1995;108:1648 –1654.
increased pneumonia. Am J Surg 1998;176:453– 457. 150. Tanaka A, Hatazawa R, Takahira Y, et al. Preconditioning
129. Martin LF, Booth FV, Karlstadt RG, et al. Continuous intrave- stress prevents cold restraint stress-induced gastric lesions
nous cimetidine decreases stress-related upper gastrointesti- in rats: roles of COX-1, COX-2 and PLA2. Dig Dis Sci 2007;
52:478 – 487.
nal hemorrhage without promoting pneumonia. Crit Care Med
151. Pilotto A. Aging and the gastrointestinal tract. Ital J Gastroen-
1993;21:19 –30.
terol Hepatol 1999;31:137–153.
130. Pinilla JC, Oleniuk FH, Reed D, et al. Does antacid prophylaxis
152. Lee M, Feldman M. The aging stomach: implications for NSAID
prevent upper gastrointestinal bleeding in critically ill patients?
gastropathy. Gut 1997;41:425– 426.
Crit Care Med 1985;13:646 – 650.
153. Cryer B, Redfern JS, Goldschmiedt M, et al. Effect of aging on
131. Groll A, Simon JB, Wigle RD, et al. Cimetidine prophylaxis for
gastric and duodenal mucosal prostaglandin concentrations in
gastrointestinal bleeding in an intensive care unit. Gut 1986;
humans. Gastroenterology 1992;102:1118 –1123.
27:135–140. 154. Vogiagis D, Gllare EM, Misajon A, et al. Cyclooxygenase-1 and
132. Halloran LG, Zfass AM, Gayle WE, et al. Prevention of acute an alternatively spliced mRNA in the rat stomach: effects of
gastrointestinal complications after severe head injury: a con- aging and ulcers. Am J Physiol Gastrointest Liver Physiol 2000;
trolled trial of cimetidine prophylaxis. Am J Surg 1980;139:44 – 278:G820 –G827.
48. 155. Gronbech JE, Lacy ER. Role of gastric blood flow in impaired
133. Cook DJ, Griffith LE, Walter SD, et al. Canadian Critical Care defense and repair of aged rat stomachs. Am J Physiol 1995;
Trials Group. The attributable mortality and length of intensive 269:G737–G744.
care unit stay of clinically important gastrointestinal bleeding in 156. Miyake H, Inaba N, Kato S, et al. Increased susceptibility of rat
critically ill patients. Crit Care 2001;5:368 –375. gastric mucosa to ulcerogenic stimulation with aging. Role of
134. Zandstra DF, Stoutenbeek CP. The virtual absence of stress- capsaicin-sensitive sensory neurons. Dig Dis Sci 1996;41:339 –
ulceration related bleeding in ICU patients receiving prolonged 345.
mechanical ventilation without any prophylaxis. A prospective 157. Goto H, Tachi K, Hisanaga Y, et al. Exacerbatory mechanism
cohort study. Intensive Care Med 1994;20:335–340. responsible for water immersion stress-induced gastric lesions
July 2008 GASTRIC MUCOSAL DEFENSE AND CYTOPROTECTION 59

GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN
in aged rats compared with young rats. Clin Exp Pharmacol 177. Takeuchi K, Araki H, Komoike Y, et al. Adaptive gastric cytopro-
Physiol 2001;28:659 – 662. tection is mediated by prostaglandin EP1 receptors: a study
158. Uchida M, Kawano O, Misaki N, et al. Healing of acetic acid using rats and knockout mice. J Pharmacol Exp Ther 2001;297:
induced gastric ulcer and gastric mucosal PGI2 level in rats. Dig 1160 –1165.
Dis Sci 1990;35:80 – 85. 178. Rostom A, Dube C, Wells G, et al. Prevention of NSAID-induced
159. Tarnawski A, Pai R, Deng X, et al. Aging gastropathy—novel gastroduodenal ulcers. Cochrane Database Syst Rev 2007:3.
mechanisms: hypoxia, upregulation of multifunctional phospha- 179. Laine L, Margolis J, Brown K. Use of GI protective agents and
tase PTEN and proapoptotic factors. Gastroenterology 2007; COX-2 selective inhibitors in 14,394,624 US patients taking
133:1938 –1947. NSAIDs. Gastroenterology 2004;126(Suppl 2):A36 –A37
160. Sarfeh IJ, Tabak C, Eugene J, et al. Clinical significance of 180. Yeomans ND, Tulassay Z, Juhasz L, et al. A comparison of
erosive gastritis in patients with alcoholic liver disease and omeprazole with ranitidine for ulcers associated with nonste-
upper gastrointestinal hemorrhage. Ann Surg 1981;194:149 – roidal antiinflammatory drugs. N Engl J Med 1998;338:719 –
151. 726.
161. Auroux J, Lamarque D, Roudot-Thoraval F, et al. Gastroduodenal 181. Graham DY, Agrawal NM, Campbell DR, et al. Ulcer prevention
ulcer and erosions are related to portal hypertensive gastropa- in long-term users of nonsteroidal anti-inflammatory drugs: re-
thy and recent alcohol intake in cirrhotic patients. Dig Dis Sci sults of a double-blind, randomized, multicenter, active- and
2003;48:1118 –1123. placebo-controlled study of misoprostol vs lansoprazole. Arch
162. Sarfeh IJ, Malki A, Tarnawski A, et al. Portal hypertension and Intern Med 2002;162:169 –175.
gastric mucosal injury in rats: effects of alcohol. Gastroenterol- 182. Chan FK, Chung SC, Suen BY, et al. Preventing recurrent upper
ogy 1983;84:987–993. gastrointestinal bleeding in patients with Helicobacter pylori
163. Sarfeh IJ, Tarnawski A. Portal hypertensive gastropathy. Gastro- infection who are taking low-dose aspirin or naproxen. N Engl
enterology 1987;93:1129 –1131. J Med 2001;344:967–973.
164. Kawanaka H, Jones MK, Szabo IL, et al. Activation of eNOS in 183. Lai KC, Lam SK, Chu KM, et al. Lansoprazole for the prevention
rat portal hypertensive gastric mucosa is mediated by TNF-␣ via of recurrences of ulcer complications from long-term low-dose
the PI e-kinase-Akt signaling pathway. Hepatology 2002;35: aspirin use. N Engl J Med 2002;346:2033–2038.
393– 402. 184. Laine L, Curtis SP, Cryer B, et al. Assessment of upper gastro-
165. Ichikawa YA, Tarnawski IJ, Sarfeh T, et al. Distorted microan- intestinal safety of etoricoxib and diclofenac in patients with
gioarchitecture and impaired angiogenesis in gastric mucosa of osteoarthritis and rheumatoid arthritis in the Multinational Etori-
portal hypertensive rat. Gastroenterology 1994;106:702–708. coxib and Diclofenac Arthritis Long-term (MEDAL) programme: a
166. Kawanaka H, Tomikawa M, Jones MK, et al. Portal hypertensive randomised comparison. Lancet 2007;369:465– 473.
gastric mucosa has reduced activation of MAP kinase (ERK2) in 185. Chan FK, Hung LC, Suen BY, et al. Celecoxib versus diclofenac
response to alcohol injury: a key to impaired healing? FASEB J and omeprazole in reducing the risk of recurrent ulcer bleeding
2001;15:574 –576. in patients with arthritis. N Engl J Med 2002;347:2104 –2110.
167. Tomikawa M, Akiba Y, Kaunitz JD, et al. New insight into impair- 186. Lai KC, Chu KM, Hui WM, et al. Celecoxib compared with
ment of mucosal defense in portal hypertensive gastric mu- lansoprazole and naproxen to prevent gastrointestinal ulcer
cosa. J Gastrointest Surg 2000;4:458 – 463. complications. Am J Med 2005;118:1271–1278.
168. Tarnawski A, Sarfeh IJ, Stachura J, et al. Microvascular abnor- 187. Chan FKL, Hung LCT, Suen BY, et al. Celecoxib versus diclofenac
malities of the portal hypertensive gastric mucosa. Hepatology plus omeprazole in high-risk arthritis patients: results of a random-
1988;8:1488 –1494. ized double-blind trial. Gastroenterology 2004;127:1038 –1043.
169. Sarfeh IJ, Soliman H, Waxman K, et al. Impaired oxygenation of 188. Chan FK, Wong VW, Suen BY, et al. Combination of a cyclo-
gastric mucosa in portal hypertension: the basis for increased oxygenase-2 inhibitor and a proton-pump inhibitor for prevention
susceptibility to injury. Dig Dis Sci 1889;34:225–228. of recurrent ulcer bleeding in patients at very high risk: a double-
170. Tarnawski A, Hollander D. Cytoprotection of gastric and duode- blind, randomised trial. Lancet 2007;369:1621–1626.
nal mucosa. R Soc Med Curr Med Lit (Gastroenterol) 1987;6: 189. Kantorova I, Svoboda P, Scheer P, et al. Stress ulcer prophy-
3–11. laxis in critically ill patients: a randomized controlled trial. Hepa-
171. Tarnawski A, Stachura J, Ivey KJ, et al. Ethanol-induced duode- togastroenterology 2004;51:757–761.
nal lesions in man. Protective effect of prostaglandin. Prosta- 190. Cook D, Guyatt G, Marshall J, et al. A comparison of sucralfate
glandins 1981;21(Suppl):147–153. and ranitidine for the prevention of upper gastrointestinal bleed-
172. Tarnawski A, Stachura J, Hollander D, et al. Cellular aspects of ing in patients requiring mechanical ventilation. Canadian Crit-
alcohol-induced injury and prostaglandin protection of the hu- ical Care Trials Group. N Engl J Med 1998;338:791–797.
man gastric mucosa. J Clin Gastroenterol 1988;19(Suppl 1): 191. Heyland D, Mandell LA. Gastric colonization by gram-negative
S35. bacilli and nosocomial pneumonia in the intensive care unit
173. Arakawa T, Higuchi K, Fujiwara Y, et al. Fifteenth anniversary of patient. Evidence for causation. Chest 1992;101:187–193.
rebamipide: looking ahead to the new mechanisms and new 192. Bonten MJ, Gaillard CA, de Leeuw PW, et al. Role of colonization
applications. Dig Dis Sci 2005;50(Suppl 1):S3–S11. of the upper intestinal tract in the pathogenesis of ventilator-
174. Tarnawski A, Hollander D, Gergely H. Antacids—new perspec- associated pneumonia. Clin Infect Dis 1997;24:309 –319.
tives in cytoprotection. Scand J Gastroenterol 1990;25(Suppl 193. Bonten MJ, Gaillard CA, van der Geest S, et al. The role of
174):9 –14. intragastric acidity and stress ulcus prophylaxis on colonization
175. Tarnawski A, Tomikawa M, Ohta M, et al. Antacid Talcid acti- and infection in mechanically ventilated ICU patients. A strati-
vates in gastric mucosa genes encoding for EGF and its recep- fied, randomized, double-blind study of sucralfate versus antac-
tor. The molecular basis for its ulcer healing action. J Physiol ids. Am J Respir Crit Care Med 1995;152:1825–1834.
(Pairs) 2000;94:93–98. 194. Torres A, El-Ebiary M, Soler N, et al. Stomach as a source of
176. Lambrecht N, Trautmann M, Korolkiewicz R, et al. Role of eico- colonization of the respiratory tract during mechanical ventila-
sanoids, nitric oxide, and afferent neurons in antacid induced tion: association with ventilator-associated pneumonia. Eur Re-
protection in the rat stomach. Gut 1993;34:329 –337. spir J 1996;9:1729 –1735.
60 LAINE, TAKEUCHI, AND TARNAWSKI GASTROENTEROLOGY Vol. 135, No. 1
GASTROENTEROLOGY
BASIC AND CLINICAL
REVIEWS IN

195. Faisy C, Guerot E, Diehl JL, et al. Clinically significant 201. Martin LF, Booth FV, Reines HD, et al. Stress ulcers and organ
gastrointestinal bleeding in critically ill patients with and failure in intubated patients in surgical intensive care units. Ann
without stress-ulcer prophylaxis. Intensive Care Med 2003; Surg 1992;215:332–337.
29:1306 –1313. 202. Martin LF. Stress ulcers are common after aortic surgery. En-
196. Allen ME, Kopp BJ, Erstad BL. Stress ulcer prophylaxis in the doscopic evaluation of prophylactic therapy. Am Surg 1994;60:
postoperative period. Am J Health Syst Pharm 2004;61:588 –596. 169 –174.
197. Raff T, Germann G, Hartmann B. The value of early enteral 203. Kahn JM, Doctor JN, Rubenfeld GD. Stress ulcer prophylaxis in
nutrition in the prophylaxis of stress ulceration in the severely mechanically ventilated patients: integrating evidence and judg-
burned patient. Burns 1997;23:313–318. ment using a decision analysis. Intensive Care Med 2006;32:
198. Bonten MJ, Gaillard CA, van Tiel FH, et al. Continuous enteral 1151–1158.
feeding counteracts preventive measures for gastric coloniza- 204. Singh G, Fort JG, Goldstein JL, et al. Celecoxib versus naproxen
tion in intensive care unit patients. Crit Care Med 1994;22: and diclofenac in osteoarthritis patients: SUCCESS-I Study.
939 –944. Am J Med 2006;119:255–266.
199. Conrad SA, Gabrielli A, Margolis B, et al. Randomized, double-
blind comparison of immediate-release omeprazole oral sus-
pension versus intravenous cimetidine for the prevention of
upper gastrointestinal bleeding in critically ill patients. Crit Care Received January 23, 2008. Accepted May 5, 2008.
Med 2005;33:760 –765. Address requests for reprints to: Loren Laine, Room 12-137, L. A.
200. Zinner MJ, Rypins EB, Martin LR, et al. Misoprostol versus County and U.S.C. Medical Center, 1200 N State St, Los Angeles,
antacid titration for preventing stress ulcers in postoperative California 90033. e-mail: llaine@usc.edu.; fax: (323) 226-7573
surgical ICU patients. Ann Surg 1989;210:590 –595. Supported in part by a VA Merit Review (to A.T.).

Potrebbero piacerti anche