Ischemia–Reperfusion Injury of the Intestine and Protective Strategies Against Injury REVIEW

C 2004)
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004), pp. 1359–1377 (°
REVIEW
Ischemia–Reperfusion Injury of the Intestine
and Protective Strategies Against Injury
ISMAIL HAMEED MALLICK, MRCS, MSc,* WENXUAN YANG, MD, PhD,* MARC C. WINSLET, MS, FRCS,*†
and ALEXANDER M. SEIFALIAN, MSc, PhD*
Ischemia–reperfusion injury of the intestine is a significant problem in abdominal aortic aneurysm
surgery, small bowel transplantation, cardiopulmonary bypass, strangulated hernias, and neonatal
necrotizing enterocolitis. It can also occur as a consequence of collapse of systemic circulation,
as in hypovolemic and septic shock. It is associated with a high morbidity and mortality. This
article is a comprehensive review of the current status of the molecular biology and the strategies to
prevent ischemia–reperfusion injury of the intestine. Various treatment modalities have successfully
been applied to attenuate reperfusion injury in animal models of reperfusion injury of the intestine.
Ischemic preconditioning has been found to be the most promising strategy against reperfusion injury
during the last few years, appearing to increase the tolerance of the intestine to reperfusion injury.
Although ischemic preconditioning has been shown to be beneficial in the human heart and the liver,
prospective controlled studies in humans involving ischemic preconditioning of the intestine are
lacking. Research focused on the application of novel drugs that can mimic the effects of ischemic
preconditioning to manipulate the cellular events during reperfusion injury of the intestine is required.
KEY WORDS: intestine; ischemia; intestinal transplantation; reperfusion injury.
Ischemia–reperfusion injury (IRI) of the intestine is an
important factor associated with a high morbidity and
mortality in both surgical and trauma patients (1). It is
of importance in situations such as the interruption of
blood flow to the gut as in abdominal aortic aneurysm
surgery, cardiopulmonary bypass, strangulated hernias,
neonatal necrotizing enterocolitis, and intestinal transplantation (2). IRI of the intestine also occurs in septic
and hypovolemic shock (3, 4).
Interruption of blood supply results in ischemic injury
which rapidly damages metabolically active tissues. Paradoxically, restoration of blood flow to the ischemic tissue
initiates a cascade of events that may lead to additional
Manuscript received April 1, 2004; accepted June 13, 2004.
From the *GI & Hepatobiliary Research Unit, University Department
of Surgery, Royal Free and University College Medical School, University College London, London NW3 2PF, UK., and †Department of Surgery, Royal Free Hampstead NHS Trust Hospital, London NW3 2QG, UK.
Address for reprint requests: A.M. Seifalian, PhD, University Department of Surgery, Royal Free and University College Medical School,
University College London, Rowland Hill Street, London NW3 2PF,
UK; [email protected].
cell injury known as reperfusion injury. This reperfusion
damage frequently exceeds the original ischemic insult
(5). On restoration of the blood supply, the molecular and
biochemical changes that occur during ischemia predispose to free radical-mediated damage. The reduction of
blood supply results in damage to the intestinal mucosa.
Among the internal organs, the intestine is probably the
most sensitive to IRI (6, 7). The intestine is composed of
labile cells that are easily injured by episodes of ischemia.
Subsequent reperfusion of the intestine results in further
damage to the mucosa (8). It has been shown that the
enterocytes that are located at the tips of the villi are more
sensitive to the effect of ischemia. Earlier studies attributed
this increased sensitivity of the enterocyte at the tips of the
villi to the location which is at the end of the distribution of
a central arteriole which can lead to lower oxygen tension
compared to the crypts (8, 9). However, a recent study in a
rat model demonstrated that the sensitivity of an enterocyte
is dependent on its state of differentiation (10).
There is substantial evidence that the mucosa of the
intestine becomes the site for the production of various
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
C 2004 Springer Science+Business Media, Inc.
0163-2116/04/0900-1359/0 °
1359
MALLICK ET AL.
acute-phase proteins (11, 12), gut hormones (13), and
cytokines (14, 15). These not only influence the intestine
but also may affect the function and integrity of distant organs. The initial site of abnormality in ischemia has been
emphasized on the cellular mitochondria, which is particularly important in producing adenosine triphosphate
(ATP) for organ recovery (16). The damage, however,
is dramatically magnified by a large number of events,
such as oxygen free radical formation, release of iron
storage, damage of the microvasculature of IR organs,
inflammatory cytokines, complement activation, and neutrophil infiltration at the site of injury (17). The enterocyte, in particular, has been shown to produce increased
amounts of cytokines during sepsis, endotoxemia, hemorrhagic shock, and trauma (18) .
SEARCH METHODS
All studies were identified by PubMed, ISIS, and
CAS searches with the following key words: intestine, ischemia, intestinal transplantation, reperfusion injury, xanthine oxidase, nitric oxide, endothelins, nuclear
factor- kappa B, neutrophils, cytokines, and preconditioning. All references were reviewed to retrieve additional
articles.
CONSEQUENCES OF INTESTINAL IRI
Alteration of Absorptive Function of the Intestine
Several studies have demonstrated an alteration of absorptive function of the intestine following IRI of the
intestine (19–21) . This could lead to deficient absorption of nutrients. In severe cases such as infarction of the
bowel, short-bowel syndrome can occur (22) . Sileri et
al. demonstrated in a rat model that IRI of the intestine
causes both acute and chronic alterations of intestinal absorptive function, which was associated with significant
mortality (23).
Bacterial Translocation
Bacterial translocation is the passage of viable bacteria from the gastrointestinal tract through the epithelial
mucosa to extraintestinal sites such as mesenteric lymph
nodes, liver, and spleen (24). Subsequently the bacteria can
disseminate throughout the body, producing sepsis, shock,
or multiple organ failure (MOF). Bacterial translocation
has been reported to occur in 44% of pediatric patients undergoing small bowel transplantation (25). The increase
in intestinal hyperpermeabilty occurring in IRI of the intestine is one of the factors causing bacterial translocation
(26–28). The other factors are overgrowth of bacteria in the
small intestine and differences in host immune defenses
(29).
1360
Injury to Distant Organs
IRI to the intestine results in production of molecules
such as hydrogen peroxide, superoxide, and inflammatory cytokines that may harm distant organs. This leads to
the development of systemic inflammatory response syndrome (SIRS), which can progress to MOF (30). Intestinal IRI also causes pulmonary infiltration of neutrophils,
which contributes to the development of acute respiratory
distress syndrome (ARDS) (31, 32). Hence, the gut has
been referred to as “the motor of MOF” (33).
MEDIATORS INVOLVED IN IRI
OF THE INTESTINE
Xanthine Oxidase and Oxygen Free Radicals
Xanthine oxidase (XO) is a highly versatile enzyme
which plays an important role in the catabolism of purines.
The intestine is the richest initial source of free radicals
that can cause further tissue damage (34). In mammals
xanthine dehydrogenase (XDH) can be converted to XO
either reversibly or irreversibly (35). During the ischemic
period, cellular ATP is catabolized to yield hypoxanthine
(36). The hypoxic stress also triggers the conversion of
XDH to the oxygen radical-producing XO. ATP depletion
results in loss of ATP-dependent ion channel regulation,
producing passive ion shifts across cell membranes: K+
and Mg2+ diffuse out, but Na+ , Ca2+ , and H2 O diffuse in,
causing cell swelling. Increased cellular Ca2+ is harmful,
one of its important consequences being the activation of a
calcium-dependent protease, which cleaves XDH to XO.
During reperfusion, molecular oxygen is reintroduced into
the tissue, where it reacts with hypoxanthine and XO to
produce a burst of oxygen free radicals—superoxide anion
(O−
2 ) and hydrogen peroxide (H2 O2 ) (37) (Figure 1).
Under physiological conditions, the damaging effects of
O−
2 are prevented by superoxide dismutase (SOD), which
converts O−
2 to H2 O2 (37). However, during reperfusion of
ischemic tissues, these natural defenses may be overcome.
Fig 1. Mechanism of xanthine oxidase-mediated free radical injury.
Modified from Granger et al. (6).
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
ISCHEMIA–REPERFUSION INJURY OF THE INTESTINE
O−
2 itself is a relatively low-energy radical, but it is responsible for production of the highly reactive and damaging
hydroxyl radical (OH• ). Typically OH• causes biological
damage by stimulating the free chain reaction known as
lipid peroxidation, in which OH• attacks the fatty acid
side chains of the membrane phospholipids and causes
organelle and cell disruption (38).
Endothelins (ETs)
ETs are potent peptide vasoconstictors derived from
the vascular endothelium (39). Among the three active
isoforms (ET-1, ET-2, and ET-3) , ET-1 is the most powerful endogenous vasoconstrictor substance known to date
(40, 41). The vasoconstrictive effects of ET-1 are predominantly mediated via ET-A receptors present on vascular
smooth muscle cells. The ET-B receptors mediate vasoconstriction (ET-B2 ) and vasodilatation (ET-B1 ) (42). ET
receptor antagonists provide significant protection against
IRI of the intestine (43–47). The major beneficial effects
of ET inhibition include a decrease in mucosal lesions, an
increase in blood flow, an increase in mucosal ATP, and a
decrease in leukocyte adhesion (48).
Heat Shock Proteins
Heat shock proteins (HSPs) are intracellular stress proteins that have been shown to accumulate after ischemia
(49).The concept of sublethal whole-body hyperthermia
conferring tolerance to ischemia and endotoxin exposure
is known as hyperthermic preconditioning and has been
associated with HSP accumulation (50, 51). In the rat intestine, tolerance to ischemic injury has been associated
with the production of various inducible HSPs: HSP-70
(52), HSP-72 (53), and HSP-73 (54). The exact mechanism whereby HSPs confer protection of the intestine
against IRI is not known. Stojadinovic and coworkers
showed that a probable mechanism involves the inhibition
of leukotriene-B4 production and subsequent prevention
of neutrophilic activation and chemotaxis (53).
Polymorphonuclear Neutrophils (PMNs)
There is a growing body of evidence that incriminates
PMNs in the pathophysiology of IRI (55–58). In a model of
small bowel transplantation, Massberg et al. showed that
the manifestation of IRI is primarily due to leukocyte–
endothelial cell interactions in the submucosal venules of
the transplanted intestine (59).
PMN-depleted blood in the reperfusate has been shown
to ameliorate IRI in the human small bowel (60). Intravital microscopic studies of tissues exposed to IRI reveal an
acute inflammatory response that is characterized by increased adhesion and emigration of PMNs in postcapillary
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
venules and enhanced protein efflux (61). Indirectly, the
assessment of neutrophil migration into the small intestine is performed by measuring myeloperoxidase (MPO)
activity in the small intestine (62).
The emigration of PMNs from the postcapillary venules
to areas of inflammation is a complex and highly coordinated three-step process, namely, rolling, adhesion, and
extravasation (63) (Figure 2). First, the leukocytes roll
along the endothelial cell surface. This initial reaction is
mediated by the upregulation of adhesion molecules such
as P-selectin and E-selectin on the surface of the endothelium and their interaction with L-selectin, which is expressed constitutively by the leukocytes. This weak leukocyte tethering to the endothelium brings the leukocyte
into close proximity to chemoattractants (e.g., leukotriene
B4 , C5a, and PAF) and chemokines (64). Chemokines
bind to receptors on leukocytes to activate β2 integrins
(CD11a, CD11b, and CD 11c/CD18). The activated integrins bind to the adhesion molecules such as intercellular adhesion molecule (ICAM-1) and vascular cellular
adhesion molecule (VCAM-1) expressed on endothelial
cells. This interaction strengthens the adhesion between
the leukocyte and the endothelial cells, thereby allowing
extravasation of the leukocyte out of the circulation and
into the interstitium (65). Riaz et al. showed that following
IRI of the mouse intestine, both P- and E-selectins were
expressed (66). Pretreatment with anti-P-selectin antibody
reduced PMN rolling and adhesion, thereby attenuating
the injury (66). PMNs can cause tissue damage in several ways including secretion of proteolytic enzymes such
as elastase from cytoplasmic granules (67), production of
free radicals via a respiratory burst (68), and physical impairment of the microcirculation and thereby extension
of ischemia (69). Nalini and colleagues showed that it is
PMNs and not XO that are the initial source of free radicals
in a rat model of IRI of the intestine (70).
Nitric Oxide (NO)
NO is a free radical and a highly reactive substance.
NO is synthesized from L-arginine by a family of enzymes
known as NO synthases (NOS). Three isoforms of NOS
have been identified and cloned: endothelial NOS (eNOS),
neuronal NOS (nNOS), and inducible NOS (iNOS) (71).
Whereas eNOS and nNOS are constitutively expressed,
iNOS is only expressed in response to cytokines and
growth factors. nNOS is the predominant NOS in the normal intestine and this suppresses the expression of iNOS
(72). Possible sources of NO in the intestine include intrinsic intestinal tissue (epithelium, mast cells, smooth
muscle, and neural plexus) and leukocytes (PMNs and
monocytes) (75).
1361
MALLICK ET AL.
Fig 2. Sequence of events in emigration of neutrophils. First, the endothelial selectins are upregulated and interact with the
oligosaccharides on the surface of leukocytes. This then brings the leukocytes into close proximity to chemoattractants such
as chemokines, which activates the integrins. Integrins (CD11/CD18) bind to the endothelial receptors (ICAM-1), resulting
in strong adhesion, followed by extravasation of the neutrophil. Reproduced from Collard and Gelman (2).
The role of NO in IRI is still a matter of considerable
controversy. NO plays a dichotomous role as both a cytotoxic and a cytoprotective molecule in intestinal IRI,
whereas inhibition of NO causes tissue dysfunction in certain models of IRI and inhibition of NO provides benefit
in others. The constitutive forms of NO synthase (nNOS
and eNOS) are critical to normal physiology and inhibition of these enzymes causes tissue damage (76). The
induction of iNOS produces large amounts of NO and peroxynitrite, leading to tissue injury, and therefore specific
inhibition of this enzyme is considered beneficial (76).
Additionally, iNOS knockout mice exhibitedsignificant
resistance to mucosal barrier dysfunction and bacterial
translocation after intestinal IRI, further supporting a role
for iNOSas an important mediator of reperfusion injury in
the intestine (77). On the contrary, Cuzzocrea et al. provided evidence that the production of peroxynitrate was
not related to the induction of iNOS, whereas it was associated with the inhibition of eNOS (78). In a recent
study, Wu et al. demonstrated that the formation of iNOS
after IRI of the intestine enhanced mucosal apoptosis,
leading to IRI of the intestine (79). Virlos et al. showed
that pulmonary injury following intestinal IRI was associated with decreased eNOS expression in the lung (80).
Expression of iNOS occurred during the later stages of
reperfusion, with overproduction of NO and nitrosylation
1362
of protein tyrosine residues, thus aggravating pulmonary
injury (80).
NO has many beneficial effects in the intestine such
as scavenging of oxygen free radicals, maintenance of
normal vascular permeability, inhibition of smooth muscle proliferation, reduction of leukocyte adherence to the
mesenteric endothelium, prevention of mast cell activation, and inhibition of platelet aggregation (81–85).
Under physiological conditions, NO will predominate
in the microcirculation (86). However, during the early
phase of reperfusion in IRI there is an accumulation of
O2 − . This limits the accumulation of NO and prevent
any of its beneficial effects. Hence there is a tendency for
PMNs to adhere to the endothelium and for platelets to aggregate (87). In the absence of NO, the O2 − produced will
undergo spontaneous dismutation to form H2 O2 . H2 O2
formed in this way can promote the activation of phospholipase A2 (PLA2) and lead to the accumulation of proinflammatory mediators such as platelet activating factor
(PAF) and leukotrienes (LTB4 ) (summarized in Figure 3).
These changes can lead to activation of transcription nuclear factor kappa B (NF–κB), which is a primary regulator of cytokine and adhesion molecule gene expression. Thus, the loss of endogenous NO production has
disastrous consequences resulting in dysfunction of the
endothelium and PMN-mediated tissue injury.
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
ISCHEMIA–REPERFUSION INJURY OF THE INTESTINE
Fig 3. Role of nitric oxide in ischemia reperfusion injury.
Heme Oxygenase (HO)
HO-1 expression in various models of IRI of the intestine has been shown to offer cytoprotection during the
cascade of events in IRI (88–91). HO-1 is the rate-limiting
enzyme in the conversion of heme into carbon monoxide
(CO), biliverdin (which is rapidly converted to bilirubin),
and free iron (Fe2+ ) (92). The HO-1 system is thought to
play a pivotal role in the maintenance of antioxidant and
oxidant homeostasis during cellular injury (93). Three isoforms of HO have been identified so far: inducible HO-1;
constitutively expressed HO-2; and HO-3, which is related
to HO-2 but is less well characterized. The HO-1 system
exerts four major beneficial effects: (a) antioxidant function, (b) maintenance of the microcirculation, (c) antiapoptosis, and (d) anti-inflammatory function (93). The
antioxidant function relies on heme degradation, production of bilirubin (94), and formation of ferritin via Fe2+
(95). The production of CO, with its vasodilatory and antiplatelet properties, maintains the microcirculation and
may be involved in the anti-apoptotic and cell arrest mechanisms (93). The HO-1 system exerts anti-inflammatory
effects via modulation of endothelial adhesion molecules
and chemoattractant factors (96). Vachharajani et al. provided evidence that HO-1 and biliverdin modulate leukoDigestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
cyte recruitment by altering the expression of P-and Eselectins in the intestine, lungs, liver, and kidneys (97).
It has also been shown that HO-1 inhibits the NO system by downregulating iNOS, which contains a heme–iron
moiety (98).
Nakao et al. showed that inhalation of CO improved
graft function and survival in a rat model of small bowel
transplantation (90). Exogenous administration of bilirubin has shown to reduce intestinal injury in rat models of
IRI (99, 100). Doxorubicin has been shown to induce HO1 and attenuate lung injury following IRI of the intestine
(88). Squiers et al. have shown that HO-1 overexpression
in small bowel donors may decrease IRI and improve survival of both the animal and the transplanted bowel (91).
Recently Attuwaybi et al. demonstrated that subcutaneous
administration of hemin (HO-1 inducer) significantly reduced IRI of the intestine (89).
Pro-inflammatory Proteins
The precise chemical steps connecting the generation
of free radicals and secretion of inflammatory mediators by endothelial cells are not well understood. It appears that NO is a key factor involved in the synthesis
1363
MALLICK ET AL.
Fig 4. The relationship between oxidants and activation of NF-κB. The transcription
factor NF-κB is normally found in the cytoplasm of the cell in an inactive form as p50/p65/
I-κB complex. Stimulation of the cells with cytokines, oxidants, or bacterial products
leads to an increase in reactive oxygen metabolites, which then activates I-κB kinase.
This kinase phosphorylates the inhibitory complex, which is then ubiquitinated. Next,
proteolysis occurs by the action of the 26S proteasome complex and nuclear translocation
of the active NF-κB.
of pro-inflammatory proteins (101). NO may be exerting its effects at the transcription level, probably by the
regulation of NF-κB, which regulates the expression of
several genes that are involved in the process of IRI (102)
(Figure 4). NF-κB belongs to a family of dimeric transcription factor complexes which is normally found in the
cytoplasm of unstimulated cells in their inactive forms,
i.e., as the p50/p65/I-κB ternary complex or the p65/p105
inactive binary complex. Stimulation of cells with certain cytokines, oxidants, or inflammatory mediators such
as PAF and LTB4 results in phosphorylation and ubiquitination of the I-κB or p105 subunit (103, 104). This
results in translocation of the subunit to the nucleus to activate transcription of its target genes. NF-κB induces the
expression of adhesion molecules (ICAM-1, VCAM-1,
P-selectin, E-selectin), tumor necrosis factor (TNF-α and
-β), interleukins, chemokines, and enzymes (inducible nitric oxide synthase and cyclooxygenase-2) (105, 106). It
appears that in IRI, reoxygenation is the key stimulus
in the regulation of NF-κB. By maintaining the interaction between IκBα and NF-κB, upregulation of adhesion
molecules is inhibited (107). NO appears to prevent the
formation of substances that activate NF-κB as well as
to enhance the activity of proteins that inhibit NF-κB.
Previous studies suggest that NO may induce the de
novo synthesis or stabilize I-κBα, thereby limiting NF-κB
activation (108).
1364
Complement
Several studies have suggested that the complement system is a major candidate in the pathogenesis of IRI of the
intestine (109–112). P-Selectin has been shown to mediate
IRI of the intestine via complement (C5b-9) deposition in
the intestinal mucosa (113). Activation of the complement
cascade leads to a series of potent inflammatory events
including expression of the ICAM-1 gene and upregulation of cytokines such as TNF-α and interleukin (IL-1α)
(112). In a recent study it was shown by Montalto et al.
that complement mediates upregulation of intestinal iNOS
and diminishes protein levels of mitochondrial SOD and
the activity of Cu/Zn SOD (114).
Iron
During ischemia, metabolites such as lactic acid accumulate and the intracellular pH falls. This decreases
the stability of lysosomal membranes, activates lysosomal
lytic enzymes, inhibits the binding of transitional metals
such as iron to their carrier proteins (e.g., transferrin and
ferritin), and results in increased free intracellular iron
(Fe2+ ), which accelerates in vivo free radical formation in
the following (Haber–Weiss) reaction: Fe2+ + H2 O2 →
Fe3+ + OH− + H• (115). Using chelating agents of iron
such as desferrioxamine has proved to be effective in reducing IRI because these agents prevent the Haber–Weiss
reaction and hydroxyl radical formation (116).
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
ISCHEMIA–REPERFUSION INJURY OF THE INTESTINE
TABLE 1. A LIST OF STUDIES IN WHICH ISCHEMIC PRECONDITIONING WAS PERFORMED AGAINST REPERFUSION INJURY IN THE INTESTINE
First author (Ref. No.)
Species
Aksoyek (26)
Rats
Cinel (142)
Ferencz (138)
Rats
Dogs
McCallion (133)
Rats
Sola (126)
Rats
Tamion (139)
Rats
Unal (132)
Rats
Vlasov (128)
Rats
Wu (140)
Rats
IPC protocol
Mechanism
10 min of ischemia &
10 min of reperfusion
10 min of ischemia
4 cycles of 5 min of
ischemia & 10 min of
reperfusion
3 cycles of 2 min of
ischemia & 5 min of
reperfusion
10 min of ischemia & 5
min of reperfusion
↓ iNOS expression
4 cycles of 1 min of
ischemia & 10 min of
reperfusion
2 cycles of 5 min of
ischemia & 5 min of
reperfusion
10 min of ischemia &
10 min of reperfusion
20 min of ischemia & 5
min of reperfusion
Expression of HO-1
Anti-apoptosis
Oxidative stress
Outcome
↓tissue damage & bacterial
translocation
↓number of apoptotic cells
↑GSH & SOD
↑adenosine
Protects against mucosal
barrier dysfunction
↑NO synthesis
↓tissue damage in this
model of intestinal
transplantation
↓fluid requirements,
↓pulmonary edema,
↓lactate & TNF-α
*↓MDA, ↓MPO, ↓tissue
damage
↓neutrophil infiltration,
↓oxidative stress and
anti-apoptosis
↑NO synthesis
Anti-apoptosis
↓tissue damage
↓reactive oxygen species,
↓lipid peroxidation,
↓cytochrome c, &
↓caspase-6
Note. GSH, glutathione; HO-1, heme oxygenase-1; iNOS, inducible nitric oxide synthase; MDA, malonyldialdehyde; MPO, myeloperoxidase; NO, nitric oxide; SOD, superoxide dismutase.
*By-product of lipid peroxidation.
PROTECTIVE STRATEGIES AGAINST
ISCHEMIA–REPERFUSION INJURY
OF THE INTESTINE
The mechanisms of IRI of the intestine and the ways
to limit these have been under extensive research for almost a decade. Several therapeutic modalities have been
used successfully to attenuate reperfusion injury in animal models of IRI of the intestine. Potential strategies
to overcome IRI include several modalities: (a) ischemic
preconditioning (IPC), (b) antioxidants, (c) NO supplementation, (d) anticomplement therapy, (e) antileukocyte
therapy, (f) perfluorocarbons, (g) enteral feeding, (h) glutamine supplementation, and (i) glycine supplementation.
Ischemic Preconditioning (IPC)
IPC refers to a phenomenon whereby exposure of a
tissue to brief periods of ischemia protects them from the
deleterious effects of prolonged IRI (117). This fascinating
phenomenon was first described in 1986 by Murry et al.
in the canine heart (118). Subsequently beneficial effects
have been demonstrated in the liver (119), skeletal muscle
(120), brain (121), spinal cord (122), kidney (123), lung
(124), retina (125), and intestine (26, 126–128) in various
animal models. There is compelling evidence that IPC is
beneficial in the human heart (129) and the liver (130).
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
IPC of the intestine was first described by Hotter et al.
in 1996 (131) and subsequent studies have confirmed this
phenomenon (Table 1). During this procedure, the small
bowel is rendered ischemic by placement of an occlusion vascular clamp around the superior mesenteric artery.
After an ischemic interval of 5–20 min, the clamp is removed and the small intestine is reperfused for 5–15 min
before the prolonged ischemic insult. However, the ideal
IPC protocol including the time of short ischemia and
number of cycle(s) is still not established. Some studies have reported that more than one cycle of ischemia
and reperfusion is required to achieve this beneficial
effect (132, 133).
The effects of IPC can be differentiated into two
phases characterized by different time frames and mechanisms: (a) an early phase (early or classic preconditioning)
that immediately follows the transient ischemia and lasts
2–3 hr and (2) a late phase (late preconditioning), which
begins 12–24 hr from the transient ischemia and lasts
for about 3–4 days (134–136). Whereas early IPC occurs within minutes and involves the direct modulation of
specific cell functions (134), the late phase requires the
simultaneous activation of multiple stress-response genes
with de novo synthesis of several proteins (135). Despite
these differences, both phases of preconditioning can be
initiated by the same stimuli and share the same intracellular signal pathways to a certain extent (135). Because of
1365
MALLICK ET AL.
its sustained duration, late preconditioning might be particularly relevant for improving the results of small bowel
transplantation. However, the present knowledge of the
mechanisms responsible is quite preliminary. Recently,
Ferencz et al. demonstrated that synthesis of proteins such
as SOD and GSH was instrumental in the protection afforded by delayed IPC in a canine model of small bowel
autotransplantation (137).
The exact mechanism by which IPC confers protection
in the intestine is unclear. Several mediators have been
advocated to play a crucial role in this protective phenomenon including adenosine (132, 133), NO (126), oxidative stress (138), HO-1 (139), and anti-apoptosis (140).
Aksoyek et al. showed that IPC of the intestine decreases the translocation of bacteria from the intestine in a
rat model of IRI (26). In a rat model of intestinal transplantation, Sola et al. identified a protective pathway initiated
by IPC that involved the activation of NO synthesis (126).
This increase in NO synthesis was short-lived and detected
at the end of preconditioning. The use of L-NAME eliminated this protection.
Davis et al. showed that IPC completely prevented the
expression of P-selectin in the ischemic intestine and abolished the IRI-induced leukocyte rolling (127). These powerful anti-inflammatory actions of IPC were thought to be
triggered by stimulation of adenosine A1 receptors during
the period of preconditioning.
The protective effect of IPC has also been associated
with a decrease in xanthine accumulation and a diminished
conversion of XDH to XO (141). There is evidence that
IPC reduces apoptosis in the intestinal epithelial cells and
this is attributed to significant expression of Bcl-2 (142).
Tamion et al., in a rat model of hemorrhagic shock followed by resuscitation, showed that IPC reduced fluid requirements, lung edema, lactate, and TNF-α production
and induced a more than fivefold increase in intestinal
HO-1 expression (139). These effects were abolished by
the HO-1 inhibitor tin protoporphyrin (Sn-PP).
Antioxidants
The human body is endowed with many natural antioxidants, which, unfortunately, are not capable of protection against the attack of oxidant formation during an
IRI. It has been shown in many animal studies that supplementation of these antioxidants is helpful in reducing IRI-induced tissue damage (Table 2). The agents that
have been used are allopurinol (143), SOD (144), iron
chelator (e.g., desferrioxamine) (145), N -acetyl cysteine
(NAC) (146), ethanol (147), ascorbic acid (148), tocopherol (149), pentoxifylline (150, 151), captopril (45), and
verapamil (152).
1366
Invtraneous SOD mimetic (M40401), administered
15 min prior to reperfusion in a model of IRI, preserved
the architecture of the ileum, reduced the expression of
cytokines, and abolished the expression of P-selectin and
ICAM-1 (144). In some cases two antioxidants are administered simultaneously (e.g., SOD and catalase) in an
attempt to block the oxidants at more than one stage of
formation (153). Kojima et al. showed that an intraduodenal infusion of the antioxidants glutathione, rebamipide,
and dimethylsulfoxide attenuated the increase in apoptosis
in the intestinal mucosa after IRI (154). A newly developed free radical scavenger, edaravone, has been shown
to be effective in reducing IRI of the intestine through its
anticytokine and antichemokine effects (155).
NO Supplementation
Supplementation of NO has been shown to attenuate IRI
in the intestine in cats and dogs (156, 157). Kalia et al.
showed that a spontaneous NO donor (FK-409) reduced
both intestinal and lung damage after IRI of the intestine in rats (158). It was also shown that inhaled NO at a
low concentration exerts profound anti-leukocyte adherence effects in cat and dog IRI (159). Whereas the use of
selective inhibitors of iNOS has been shown to be beneficial, the use of nonselective inhibitors has been shown
to be deleterious in intestinal IRI (160). Recently, Naito
et al. demonstrated that the administration of a selective
iNOS inhibitor, ONO-1714, attenuated IRI of the intestine (161). Intraluminal administration of nitroglycerine,
a potent exogenous NO donor, prevented the increase in
intestinal permeability associated with IRI of the intestine
(162).
Anticomplement Therapy
IRI is significantly reduced by complement inhibition
or complement depletion or in complement-deficient animals (2). Administration of a complement (C5a) receptor
antagonist has been shown to protect the intestine against
IRI in a murine model (110). Arumugam et al. showed that
administration of an orally active C5a receptor antagonist
was highly effective in reducing damage to the intestine in
a rat model (163). The anti-C5 antibody inhibited TNF-α
expression and tissue injury. Zhao et al. showed that the
use of an antimurine C5 monoclonal antibody decreased
neutrophilic infiltration of both the intestine and the lungs
(164). Williams et al. showed that the administration of a
complement inhibitor of soluble complement receptor 1
(sCR-1) attenuated intestinal mucosal injury, reduced intestinal vascular hyperpermeabilty, and increased survival
in a rat model (165). It has been demonstrated recently
that the administration of human C1 esterase inhibitor has
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
ISCHEMIA–REPERFUSION INJURY OF THE INTESTINE
TABLE 2. A LIST OF ANTIOXIDANTS USED AGAINST REPERFUSION INJURY IN THE INTESTINE
Agent
First author (Ref. No.) Species
Mechanism
Allopurinol
Ferrer (205)
Rats
Anti-thrombin III
Ozden (206)
Rats
Oxidative stress,
scavenging free
radicals
Lipid peroxidation
Bilirubin
Hammerman (100)
Rats
Lipid peroxidation
Edaravone
Ceran (99)
Tomatsuri (155)
Rats
Rats
Antioxidant
Anti-cytokine &
-chemokine
Captopril
Buyukgebiz (45)
Rats
Free radical scavenger
Cyclosporine
Desferrioxamine
Epidermal growth factor (EGF)
Puglisi (207)
Balogh (145)
Berlanga (208)
Rats
Rats
Rats
Free radical scavenger
Lipid peroxidation
Lipid peroxidation
Glutamine
Harward (199)
Rats
Lipid peroxidation
Melatonin
Ustundag (209)
Rats
Oxidative stress
Kazez (210)
Rats
Lipid peroxidation
N -Acetyl cysteine
Sun (169)
Rats
Neutrophil infiltration
Nitroglycerine
Dun (211)
Rats
Lipid peroxidation
Pyruvate
Cicalese (212)
Rats
Free radical scavenger
Rapamycin
SOD
Puglisi (207)
Riaz (173)
Rats
Rats
Free radical scavenger
Oxidative stress
Trimetazidine
Tetik (213)
Rats
Free radical scavenger
Verapamil
Vitamins C & E
Mocan (152)
Gunel (149)
Rats
Rabbits
Oxidative stress
Lipid peroxidation
Outcome
↓tissue damage,
↓amylase, ↓MDA,
↓GSH, & ↓MPO
*↓MDA, ↓ MPO, ↓tissue
damage
↓blood and intestinal
thiobarbituric acid &
tissue damage
↓tissue damage
↓tissue damage, ↓MPO,
↓TBA-reactive
substances, ↓CINC-1
protein, & ↓CINC-1
mRNA
*↓MDA, ↓ MPO, ↓tissue
damage
↓tissue damage
*↓MDA
*↓MDA, ↓ MPO, ↓tissue
damage
↑GSH, ↓conjugated diene
(a by-product of lipid
peroxidation)
↑SOD, ↑glutathione
peroxidase
*↓MDA & ↓tissue
damage
↓MPO & ↓endothelial
permeability
↓tissue damage, ↓MDA,
& ↓LDH
↓tissue damage, ↓free
radical & neutrophil
infiltration
↓tissue damage
↓neutrophil rolling &
adhesion
*↓MDA, ↓ MPO, ↓tissue
damage
↓tissue damage
*↓MDA
Note. CINC, cytokine-induced neutrophil chemoattractant; GSH, glutathione; HO-1, heme oxygenase-1; iNOS, inducible nitric oxide
synthase; LDH, lactate dehydrogenase; MDA, malonyldialdehyde; MPO, myloperoxidase; NO, nitric oxide; RNA, ribonucleic acid; SOD,
superoxide dismutase; TBA, thiobarbituric acid.
*An indicator of lipid peroxidation.
been shown to attenuate reperfusion injury in a mouse
model of intestinal IRI (166).
Antileukocyte Therapy
There are broadly three therapeutic strategies to attenuate leukocyte-mediated IRI: (a) inhibition of leukocyte
activation, (b) inhibition of leukocyte adhesion molecule
synthesis, and (c) inhibition of leukocyte–endothelial
adhesion (2).
The activation of leukocytes in IRI is aided by the release of inflammatory mediators such as leukotriene-B4 ,
(LT-B4) platelet activating factor (PAF), and tumor necrosis factor-α (TNF- α) (65). Studies have shown that the use
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
of LT-B4 antagonists (167), PAF antagonists (168, 169),
and TNF-α inhibitors (170) limits IRI-induced leukocyte
activation.
The second strategy to attenuate leukocyte-mediated
IRI has been to inhibit leukocyte adhesion molecule synthesis. Transcription factors regulating leukocyte adhesion
molecule synthesis, such as NF-κB, are the targets for
BAY11-7085 (I-κ B inhibitor) (171) and α melanocyte
stimulating hormone (172).
The third strategy is to limit leukocyte–endothelial adhesion. Riaz et al. showed that the use of allopurinol
and SOD reduced leukocyte rolling and adhesion in a
dose-dependent manner (173). The use of monoclonal
1367
MALLICK ET AL.
antibodies directed against adhesion molecules reduces
the severity of IRI in the intestine. One restraining factor
that has limited the clinical use of monoclonal antibodies
against leukocyte–endothelial interaction is the possibility of an immune reaction (174). Pretreatment with an
anti-P-selectin antibody has been shown to reduce leukocyte adhesion by about 85%, and IRI-induced leukocyte
adhesion was significantly reduced in leukocyte function
antigen-1 (LFA)-deficient mice (66). However, Carmody
et al. demonstrated that eliminating P-selectin by using Pselectin knockout mice attenuated reperfusion injury, and
this beneficial effect was found to be independent of neutrophil adhesion (175). The administration of anti-LFA-1
antibodies has been shown to be effective in improving
allograft survival in small bowel transplantation (176). A
nonexhaustive list of antileukocyte and anti-inflammatory
agents used in IRI of the intestine is shown in Table 3.
Perfluorocarbons (PFCs)
PFCs are hydrocarbon molecules in which hydrogen
atoms are replaced by fluorine. They can dissolve 20–25
times more oxygen than plasma at room temperature. The
release of oxygen is facilitated by a low oxygen-binding
constant, causing a linear relationship between oxygen
saturation and partial pressure. This results in release of
oxygen to the tissues. Several studies have shown that
PFCs attenuate IRI of the intestine (177–179).
Intraluminal administration of oxygenated PFCs into
the intestine has been shown to preserve mucosal function and integrity after IRI of the intestine (180). Ohara
et al. showed that peritoneal lavage with PFCs reduced
intestinal and pulmonary damage following IRI of the intestine (181), whereas Vejchapipat et al. demonstrated that
extraluminal administration of PFCs did not protect the intestine from IRI in a rat model (182). The small intestine,
when placed in a bag containing PFCs, attenuated the expression of cytokines and decreased the intestinal damage
in a rat model of IRI (183). Partial liquid ventilation with
PFCs has been shown to reduced remote organ injury by
reducing the lung damage (184).
Enteral Feeding
Enteral nutrition has been shown to reduce the death rate
and remote organ injury in a mouse model (185). Enteral
feeding maintains the mucosal immunity and the resistance to infections. Fukatsu et al. showed that 5 days of gut
“starvation” with parenteral nutrition increases ICAM-1
and P-selectin expression, thereby attracting PMNs to
the intestine, compared with enterally fed animals (186).
These ICAM-1 changes were reversible with reinstitution
of enteral feeding. Five days of refeeding the parenterally
1368
fed mice with chow returned ICAM-1 levels to normal.
Furthermore, lack of enteral feeding has shown to alter
the levels of IL-4 and IL-10 in the lamina propria (187).
IL-4 and IL-10 inhibit endothelial expression of ICAM-1.
Glutamine Supplementation
Glutamine, considered to be a nonessential amino acid,
appears to be a conditionally essential nutrient during serious injury (188). Glutamine is the most abundant amino
acid in the plasma and skeletal muscle under normal physiological conditions, but the levels fall substantially after
injury or surgery (189). Several studies demonstrate that Lglutamine- and glutamine dipeptide (glycyl-glutamine and
alanyl-glutamine)-supplemented parenteral nutrition and
L-glutamine-supplemented enteral feeds decrease bacterial translocation, diminish experimental colitis, and enhance mucosal adaptation with refeeding or following
bowel resection (190, 191). In a porcine model of segmental small bowel autotransplantation, Li et al. showed
that glycyl-glutamine-enriched parenteral nutrition significantly increased graft mucosal glutamine and protein
levels and decreased intestinal permeability and bacterial translocation (192). Of interest, in a rat small bowel
transplantation model, significantly improved jejunal graft
maltose absorption and posttransplantation mortality were
demonstrated when harvested grafts were cold-preserved
in 1% L-glutamine versus saline alone or in University of
Wisconsin preservation solutions (193).
Intravenous infusion of L-glutamine has been demonstrated to increase the cytoprotective HO-1 and the antioxidants GSH and SOD, with a concomitant decrease in
TNF-α in IRI of the intestine (194). Glutamine-enriched
tube feeding was shown by Zhang et al. to enhance transplanted small bowel growth, nutrient absorption, and mucosal barrier function (195). Ikeda et al., in a mouse model
of IRI of the intestine, showed that supplementation of
TPN with glutamine partially supports mucosal immunity
by preserving respiratory and intestinal IgA levels, maintaining the proper IgA-stimulating cytokine milieu within
the intestine, and reducing intestinal ICAM-1 expression
and neutrophil adhesion (196). Wischmeyer provided evidence that glutamine enhances heat shock protein (HSP)
expression and thereby confers protection against a variety
of stressful stimuli (197, 198). Another probable mechanism is that glutamine given intravenously increased mucosal glutathione (GSH) levels and attenuated IRI-induced
cell membrane lipid peroxidation (199).
Glycine Supplementation
L-Glycine, a nonessential amino acid, protects the
intestine from IRI (27, 200, 201). Glycine has an
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
ISCHEMIA–REPERFUSION INJURY OF THE INTESTINE
TABLE 3. A LIST OF ANTILEUKOCYTE AND ANTI-INFLAMMATORY AGENTS USED AGAINST REPERFUSION INJURY IN THE
INTESTINE
Agent
First author (Ref. No.)
Species
α-Melanocyte
Anti-IL-8 antibody
Hassoun (172)
Tsuruma (214)
Rats
Rats
Anti-TNF antibody
Koksoy (31)
Rats
BAY-11-7095
Zou (171)
Rats
Inhibition of NF-κB
Inhibition of IL-8
expression
Inhibition of TNF
expression
Inhibition of I-κB
BN 52021
CP-105,696
Droy-Lefaix (215)
Souza (216)
Rats
Rats
PAF antagonist
Inhibition of leukotrienes
CINC Ab
Yagihashi (217)
Rats
Cyclic peptide AcF
(OpdChaWR)
Arumugam (163)
Rats
Inhibition of TNF
expression
Complement antagonist
Ethanol
Yamaguchi (147)
Mice
Fetal bovine serum
FK-409
Chen (218)
Kalia (158)
Rats
Rats
FR-167653
Kawata (156)
Yamamoto (7)
Dogs
Rats
Takeyoshi (9)
Dogs
GPI 6150
Mazzon (219)
Rats
Glycine
Lee (201)
Rats
Iijima (27)
Mice
Heparin binding EGF
Xia (220)
Rats
Inhibition of neutrophil
infiltration
Hepatocyte growth
factor
Hyperbaric oxygen
IL-10
Kuenzler (221)
Rats
Inhibition of TNF
Yang (222)
Lane (223)
Rats
Mice
Inhibition of TNF
Cytokine attenuation
Inosine
Dowdall (224)
Rats
Cytokine attenuation
Stroh (225)
Rats
Inhibition of neutrophil
infiltration
Lipocortin-1
Cuzzocrea (226)
Rats
Inhibition of neutrophil
infiltration
L-Arginine
Warnecke (227)
Rats
Inhibition of TNF-α &
IL-6
L-Propionyl
Carnitine
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
Mechanism
Inhibition of neutrophil
infiltration
Inhibition of IL-8
NO donor
NO donor
Inhibition of TNF-α &
IL-1
Inhibition of TNF-α &
IL-1
P-Selectin & ICAM-1
blocker
Inhibition of neutrophil
infiltration
Inhibition of IL-6
Outcome
↓tissue damage & ↓MPO
↓tissue damage & TNF-α
↓neutrophil sequestration
↓tissue damage >30 min
of reperfusion &
↑tissue damage >6hr
reperfusion, ↓MPO,
↓IL-6, & ↓ICAM-1
↓tissue damage
↓tissue damage,
↓vascular permeability,
& ↓TNF-α in lungs &
intestine
↓tissue damage &
↓TNF-α
↓tissue damage, ↓TNF-α
& inhibition of
IRI-induced
neutropenia
↓leukocyte rolling &
adhesion
↓IL-8
Maintains WCC,
↓villi–leukocyte
adhesion, ↓ lung
damage, & ↑survival
↓tissue damage
↓tissue damage, ↓TNF,
↓IL-1, & ↓AST
↓tissue damage
Improves mean BP,
↓tissue damage,
↓MPO, & ↑ survival
↑mucosal protein &
DNA, ↓MPO
↓tissue damage,
↓bacterial
translocation, & ↑
survival
↓expression of P/ Eselectins, VCAM-1, &
ICAM-1
↓caspases
↓TNF-α & lung MPO
↓tissue damage, ↓TNF-α,
↓lung MPO, & ↓IL-6
↓tissue damage, ↓ gut
permeability, & ↓lung
MPO
↑survival, maintains
mean arterial pressure,
↓tissue MPO
Maintains blood pressure,
↓tissue MPO, &
↓tissue damage
↓tissue damage, ↓TNF-α,
↓hyaluronic acid, &
↓IL-6
1369
MALLICK ET AL.
TABLE 3.
Agent
First author (Ref. No.) Species
CONTINUE.
Mechanism
Outcome
Lexipafant
Sun (169)
Rats
PAF antagonist
M40401
Cuzzocrea (144)
Rats
Magnolol
Methyl prednisolone
Loong (228)
Warnecke (227)
Rats
Rats
P-Selectin blocker &
inhibition of ICAM-1
Inhibition of IL-6
Inhibition of TNF & IL-6
P-Selectin
Glycoprotein ligand
Farmer (229)
Rats
P-Selectin blocker
Pentoxifylline
Sener (150)
Rats
Pirfenidone
Arumugam (230)
Rats
Inhibition of neutrophil
infiltration
Inhibition of TNF-α
Rolipram
Souza (170)
Rats
Inhibition of TNF-α
UK 74505
Souza (168)
Rats
PAF antagonist
↓endothelial permeability
& ↓MPO
Improves blood pressure,
↓MDA, & ↓MPO
↓tissue damage &↓ IL-6
↓tissue damage, ↓TNF-α,
↓hyaluronic acid, & ↓
IL-6
↓tissue damage, ↓ IL-11,
IL-6, interferons, &
↓MPO
↓MDA, ↓MPO, &
↑glutathione
↓tissue damage &
↓TNF-α
↓intestinal & lung
permeability,
↓recruitment of
neutrophils, ↓ IL-10,
↓TNF-α in both lungs
& intestine
↓TNF-α, ↓MPO, ↓tissue
damage & mucosal
permeability
Note. Ab, antibody; AST, aspartate aminotransferase; BP, blood pressure; CINC, cytokine-induced neutrophil chemoattractant; EGF, epidermal growth factor; GSH, glutathione; HO-1, heme oxygenase-1; ICAM, MDA-malonyldialdehyde;
intercellular malonyldialdehyde; MPO, myelooperoxidase; NF-κB, nuclear factor kappa −B; NO, nitric oxide; PAF, platelet
activating factor; SOD, superoxide dismutase; TNF, tumor necrosis factor; VCAM; vascular cellular adhesion molecule;
WCC, white cell count.
impressive catalog of functions such as anti-inflammatory,
immunomodulatory, and direct cytoprotective functions
(200–202). Iijima et al. showed that the administration of
20% glycine attenuated mucosal damage, preserved the
intestinal protein content, reduced bacterial translocation,
and improved the overall survival in a murine model (27).
Glycine acts on inflammatory cells such as macrophages to
suppress the activation of transcription factors and the formation of free radicals and inflammatory cytokines (203).
Recently, Jacob et al. demonstrated that supplementation
of glycine caused a downregulation of pro-apoptotic bax
and caspase-3, whereas bcl-2 was upregulated (204).
CONCLUSION AND FUTURE WORK
IRI of the intestine is a complex, multifactorial pathophysiological process that involves the actions of oxygenderived free radicals, cytokines, nitric oxide, and PMNs.
Despite the complexity, attenuation of the reperfusion injury through ischemic and pharmacological preconditioning has been successfully applied to animal models of intestinal IRI. IPC has been found to be the most promising
strategy against IRI during the last few years, appearing to
increase the tolerance of the intestine to reperfusion injury.
However, although IPC has been shown to be beneficial
in the human heart and the liver, prospectively controlled
1370
studies in humans involving IPC of the intestine are lacking. Research focused on the application of novel drugs
that can mimic the effects of IPC to manipulate the cellular
events during IRI of the intestine is required.
REFERENCES
1. Koike K, Moore FA, Moore EE, Read RA, Carl VS, Banerjee
A: Gut ischemia mediates lung injury by a xanthine oxidasedependent neutrophil mechanism. J Surg Res 54(5):469–473, 1993
2. Collard CD, Gelman S: Pathophysiology, clinical manifestations,
and prevention of ischemia-reperfusion injury. Anesthesiology
94(6):1133–1138, 2001
3. Moore EE, Moore FA, Franciose RJ, Kim FJ, Biffl WL, Banerjee A:
The postischemic gut serves as a priming bed for circulating neutrophils that provoke multiple organ failure. J Trauma 37(6):881–
887, 1994
4. Swank GM, Deitch EA: Role of the gut in multiple organ failure:
Bacterial translocation and permeability changes. World J Surg
20(4):411–417, 1996
5. Stallion A, Kou TD, Miller KA, Dahms BB, Dudgeon DL, Levine
AD: IL-10 is not protective in intestinal ischemia reperfusion injury. J Surg Res 105(2):145–152, 2002
6. Granger DN, Hollwarth ME, Parks DA: Ischemia-reperfusion injury: Role of oxygen-derived free radicals. Acta Physiol Scand
Suppl 548:47–63, 1986
7. Yamamoto S, Tanabe M, Wakabayashi G, Shimazu M, Matsumoto
K, Kitajima M: The role of tumor necrosis factor-alpha and
interleukin-1 beta in ischemia-reperfusion injury of the rat small
intestine. J Surg Res 99(1):134–141, 2001
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
ISCHEMIA–REPERFUSION INJURY OF THE INTESTINE
8. Kong SE, Blennerhassett LR, Heel KA, McCauley RD, Hall JC:
Ischaemia-reperfusion injury to the intestine. Aust NZ J Surg
68(8):554–561, 1998
9. Takeyoshi I, Zhang S, Nakamura K, Ikoma A, Zhu Y, Starzl TE,
Todo S: Effect of ischemia on the canine large bowel: A comparison
with the small intestine. J Surg Res 62(1):41–48, 1996
10. Hinnebusch BF, Ma Q, Henderson JW, Siddique A, Archer SY,
Hodin RA: Enterocyte response to ischemia is dependent on differentiation state. J Gastrointest Surg 6(3):403–409, 2002
11. Molmenti EP, Ziambaras T, Perlmutter DH: Evidence for an acute
phase response in human intestinal epithelial cells. J Biol Chem
268(19):14116–14124, 1993
12. Wang Q, Meyer TA, Boyce ST, Wang JJ, Sun X, Tiao G, Fischer JE, Hasselgren PO: Endotoxemia in mice stimulates production of complement C3 and serum amyloid A in mucosa
of small intestine. Am J Physiol 275(5, Pt 2):R1584–R1592,
1998
13. Zamir O, Hasselgren PO, Higashiguchi T, Frederick JA, Fischer
JE: Effect of sepsis or cytokine administration on release of gut
peptides. Am J Surg 163(1):181–185, 1992
14. Mester MD, Tompkins MD, Gelfand MD, Dinarello MD: Intestinal production of interleukin-1[alpha] during endotoxemia in the
mouse. J Surg Res 54(6):584–591, 1993
15. Meyer TA, Wang J, Tiao GM, Ogle CK, Fischer JE, Hasselgren
PO: Sepsis and endotoxemia stimulate intestinal interleukin-6 production. Surgery 118(2):336–342, 1995
16. Jassem W, Fuggle SV, Rela M, Koo DD, Heaton ND: The role
of mitochondria in ischemia/reperfusion injury. Transplantation
73(4):493–499, 2002
17. Carden DL and Granger DN: Pathophysiology of ischaemiareperfusion injury. J Pathol 190(3):255–266, 2000
18. Ogle CK, Mao JX, Wu JZ, Ogle JD, Alexander JW: The 1994 Lindberg Award. The production of tumor necrosis factor, interleukin1, interleukin-6, and prostaglandin E2 by isolated enterocytes and
gut macrophages: Effect of lipopolysaccharide and thermal injury.
J Burn Care Rehabil 15(6):470–477, 1994
19. Kuenzler KA, Pearson PY, Schwartz MZ: Interleukin-11 enhances
intestinal absorptive function after ischemia-reperfusion injury. J
Pediatr Surg 37(3):457–459, 2002
20. Prasad R, Alavi K, Schwartz MZ: GLP-2alpha accelerates
recovery of mucosal absorptive function after intestinal ischemia/reperfusion. J Pediatr Surg 36(4):570–572, 2001
21. Rajeevprasad R, Alavi K, Schwartz MZ: Glucagonlike peptide-2
analogue enhances intestinal mucosal mass and absorptive function after ischemia-reperfusion injury. J Pediatr Surg 35(11):1537–
1539, 2000
22. Scolapio JS, Fleming CR: Short bowel syndrome. Gastroenterol
Clin North Am 27(2):467–479, viii, 1998
23. Sileri P, Morini S, Schena S, Rastellini C, Abcarian H, Benedetti E,
Cicalese L: Intestinal ischemia/reperfusion injury produces chronic
abnormalities of absorptive function. Transplant Proc 34(3):984,
2002
24. Van Leeuwen PA, Boermeester MA, Houdijk AP, Ferwerda CC,
Cuesta MA, Meyer S, Wesdorp RI: Clinical significance of translocation. Gut 35(Suppl 1 ):S28–S34, 1994
25. Cicalese L, Sileri P, Green M, Abu-Elmagd K, Kocoshis S, Reyes J:
Bacterial translocation in clinical intestinal transplantation. Transplantation 71(10):1414–1417, 2001
26. Aksoyek S, Cinel I, Avlan D, Cinel L, Ozturk C, Gurbuz P, Nayci
A, Oral U: Intestinal ischemic preconditioning protects the intestine and reduces bacterial translocation. Shock 18(5):476–480,
2002
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
27. Iijima S, Shou J, Naama H, Calvano SE, Daly JM: Beneficial effect of enteral glycine in intestinal ischemia/reperfusion injury. J
Gastrointest Surg 1(1):53–60, 1997
28. Xia G, Martin AE, Michalsky MP, Besner GE: Heparin-binding
EGF-like growth factor preserves crypt cell proliferation and decreases bacterial translocation after intestinal ischemia/reperfusion
injury. J Pediatr Surg 37(7):1081 [Discussion], 2002
29. Berg RD: Bacterial translocation from the gastrointestinal tract.
Adv Exp Med Biol 473:11–30, 1999
30. Ceppa EP, Fuh KC, Bulkley GB: Mesenteric hemodynamic response to circulatory shock. Curr Opin Crit Care 9(2):127–132,
2003
31. Koksoy C, Kuzu MA, Kuzu I, Ergun H, Gurhan I: Role of tumour necrosis factor in lung injury caused by intestinal ischaemiareperfusion. Br J Surg 88(3):464–468, 2001
32. Xiao F, Eppihimer MJ, Young JA, Nguyen K, Carden
DL: Lung neutrophil retention and injury after intestinal ischemia/reperfusion. Microcirculation 4(3):359–367, 1997
33. Marshall JC: The gut as a potential trigger of exercise-induced
inflammatory responses. Can J Physiol Pharmacol 76(5):479–484,
1998
34. Parks DA, Williams TK, Beckman JS: Conversion of xanthine dehydrogenase to oxidase in ischemic rat intestine: A reevaluation.
Am J Physiol 254(5, Pt 1):G768–G774, 1988
35. Meneshian A and Bulkley GB: The physiology of endothelial xanthine oxidase: From urate catabolism to reperfusion injury to inflammatory signal transduction. Microcirculation 9(3):161–175,
2002
36. Younes M, Schoenberg MH, Jung H, Fredholm BB, Haglund U,
Schildberg FW: Oxidative tissue damage following regional intestinal ischemia and reperfusion in the cat. Res Exp Med (Berl)
184(4):259–264, 1984
37. Harrison R: Structure and function of xanthine oxidoreductase:
Where are we now? Free Radic Biol Med 33(6):774–797, 2002
38. Halliwell B, Gutteridge J: Free Radicals in Biology and Medicine,
3rd ed. New York: Oxford University Press, 1999
39. Ozel SK, Yuksel M, Haklar G, Durakbasa CU, Dagli TE, Aktan
AO: Nitric oxide and endothelin relationship in intestinal ischemia/reperfusion injury (II). Prostaglandins Leukot Essent Fatty
Acids 64(4–5):253–257, 2001
40. Inoue A, Yanagisawa M, Kimura S, Kasuya Y, Miyauchi T, Goto
K, Masaki T: The human endothelin family: three structurally and
pharmacologically distinct isopeptides predicted by three separate
genes. Proc Natl Acad Sci USA 86(8):2863–2867, 1989
41. Yanagisawa M, Kurihara H, Kimura S, Tomobe Y, Kobayashi M,
Mitsui Y, Yazaki Y, Goto K, Masaki T: A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature
332(6163):411–415, 1988
42. Clozel M, Gray GA, Breu V, Loffler BM, Osterwalder R: The
endothelin ETB receptor mediates both vasodilation and vasoconstriction in vivo. Biochem Biophys Res Commun 186(2):867–873,
1992
43. Anadol AZ, Bayram O, Dursun A, Ercan S: Role of endogenous endothelin peptides in intestinal ischemia-reperfusion injury
in rats. Prostaglandins Leukot Essent Fatty Acids 59(4):279–283,
1998
44. Andrasi TB, Kekesi V, Blazovics A, Dobi I, Szabo G, Juhasz-Nagy
A: ET(A) receptor blockade protects the small intestine against ischaemia/reperfusion injury in dogs via an enhancement of antioxidant defences. Clin Sci (Lond) 103 (Suppl 48):59S–63S, 2002
45. Buyukgebiz O, Aktan AO, Yegen C, Yalcin AS, Haklar G, Yalin R,
Ercan ZS: Captopril increases endothelin serum concentrations and
1371
MALLICK ET AL.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
56.
57.
58.
59.
60.
61.
preserves intestinal mucosa after mesenteric ischemia-reperfusion
injury. Res Exp Med (Berl) 194(6):339–348, 1994
Oktar BK, Gulpinar MA, Bozkurt A, Ghandour S, Cetinel S, Moini
H, Yegen BC, Bilsel S, Granger DN, Kurtel H: Endothelin receptor blockers reduce I/R-induced intestinal mucosal injury: Role of
blood flow. Am J Physiol Gastrointest Liver Physiol 282(4):G647–
G655, 2002
Wolfard A, Szalay L, Kaszaki J, Sahin-Toth G, Vangel R, Balogh A,
Boros M: Dynamic in vivo observation of villus microcirculation
during small bowel autotransplantation: Effects of endothelin-A
receptor inhibition. Transplantation 73(9):1511–1513, 2002
Oktar BK, Coskun T, Bozkurt A, Yegen BC, Yuksel M, Haklar
G, Bilsel S, Aksungar FB, Cetinel U, Granger DN, Kurtel H:
Endothelin-1-induced PMN infiltration and mucosal dysfunction
in the rat small intestine. Am J Physiol Gastrointest Liver Physiol
279(3):G483–G491, 2000
Tsuruma T, Yagihashi A, Matsuno T, Zou XM, Asanuma K,
Sasaki K, Hirata K: The heat-shock protein 70 family reduces
ischemia/reperfusion injury in small intestine. Transplant Proc
28(5):2629–2630, 1996
Hotchkiss R, Nunnally I, Lindquist S, Taulien J, Perdrizet G, Karl I:
Hyperthermia protects mice against the lethal effects of endotoxin.
Am J Physiol 265(6, Pt 2):R1447–R1457, 1993
Ryan AJ, Flanagan SW, Moseley PL, Gisolfi CV: Acute heat stress
protects rats against endotoxin shock. J Appl Physiol 73(4):1517–
1522, 1992
Fleming SD, Starnes BW, Kiang JG, Stojadinovic A, Tsokos GC,
Shea-Donohue T: Heat stress protection against mesenteric I/Rinduced alterations in intestinal mucosa in rats. J Appl Physiol
92(6):2600–2607, 2002
Stojadinovic A, Kiang J, Smallridge R, Galloway R, SheaDonohue T: Induction of heat-shock protein 72 protects against
ischemia/reperfusion in rat small intestine. Gastroenterology
109(2):505–515, 1995
Tsuruma T, Yagihashi A, Watanabe N, Yajima T, Kameshima H,
Araya J, Hirata K: Heat-shock protein-73 protects against small
intestinal warm ischemia-reperfusion injury in the rat. Surgery
125(4):385–395, 1999
Hayward R and Lefer AM: Time course of endothelial-neutrophil
interaction in splanchnic artery ischemia-reperfusion. Am J Physiol
275(6, Pt 2):H2080–H2086, 1998
Hierholzer C, Kalff JC, Audolfsson G, Billiar TR, Tweardy DJ,
Bauer AJ: Molecular and functional contractile sequelae of rat intestinal ischemia/reperfusion injury. Transplantation 68(9):1244–
1254, 1999
Cooper D, Russell J, Chitman KD, Williams MC, Wolf RE, Granger
DN: Leukocyte dependence of platelet adhesion in postcapillary
venules. Am J Physiol Heart Circ Physiol 286(5):H1895–H1900,
2004
Mbachu EM, Klein LV, Rubin BB, Lindsay TF: A monoclonal antibody against cytokine-induced neutrophil chemoattractant attenuates injury in the small intestine in a model of ruptured abdominal
aortic aneurysm. J Vasc Surg 39(5):1104–1111, 2004
Massberg S, Gonzalez AP, Leiderer R, Menger MD, Messmer K:
In vivo assessment of the influence of cold preservation time on
microvascular reperfusion injury after experimental small bowel
transplantation. Br J Surg 85(1):127–133, 1998
Sisley AC, Desai T, Harig JM, Gewertz BL: Neutrophil depletion attenuates human intestinal reperfusion injury. J Surg Res
57(1):192–196, 1994
Granger DN, Korthuis RJ: Physiologic mechanisms of postischemic tissue injury. Annu Rev Physiol 57:311–332, 1995
1372
62. Naito Y, Takagi T, Uchiyama K, Handa O, Tomatsuri N, Imamoto
E, Kokura S, Ichikawa H, Yoshida N, Yoshikawa T: Suppression
of intestinal ischemia-reperfusion injury by a specific peroxisome
proliferator-activated receptor-gamma ligand, pioglitazone, in rats.
Redox Rep 7(5):294–299, 2002
63. Grisham MB, Granger DN, Lefer DJ: Modulation of leukocyteendothelial interactions by reactive metabolites of oxygen and nitrogen: Relevance to ischemic heart disease. Free Radic Biol Med
25(4–5):404–433, 1998
64. Granger DN: Role of xanthine oxidase and granulocytes in
ischemia-reperfusion injury. Am J Physiol 255(6, Pt 2):H1269–
H1275, 1988
65. Panes J, Perry M, Granger DN: Leukocyte-endothelial cell adhesion: Avenues for therapeutic intervention. Br J Pharmacol
126(3):537–550, 1999
66. Riaz AA, Wan MX, Schaefer T, Schramm R, Ekberg H,
Menger MD, Jeppsson B, Thorlacius H: Fundamental and distinct roles of P-selectin and LFA-1 in ischemia/reperfusion-induced
leukocyte-endothelium interactions in the mouse colon. Ann Surg
236(6):777–784, 2002
67. Carden DL, Smith JK, Korthuis RJ: Neutrophil-mediated microvascular dysfunction in postischemic canine skeletal muscle.
Role of granulocyte adherence. Circ Res 66(5):1436–1444, 1990
68. Weiss SJ: Tissue destruction by neutrophils. N Engl J Med
320(6):365–376, 1989
69. Bagge U, Amundson B, Lauritzen C: White blood cell deformability and plugging of skeletal muscle capillaries in hemorrhagic
shock. Acta Physiol Scand 108(2):159–163, 1980
70. Nalini S, Mathan MM, Balasubramanian KA: Oxygen free radical
induced damage during intestinal ischemia/reperfusion in normal
and xanthine oxidase deficient rats. Mol Cell Biochem 124(1):59–
66, 1993
71. Albrecht EW, Stegeman CA, Heeringa P, Henning RH, van Goor
H: Protective role of endothelial nitric oxide synthase. J Pathol
199(1):8–17, 2003
72. Qu XW, Wang H, De Plaen IG, Rozenfeld RA, Hsueh W: Neuronal
nitric oxide synthase (NOS) regulates the expression of inducible
NOS in rat small intestine via modulation of nuclear factor kappa
B. FASEB J 15(2):439–446, 2001
73. Hasan K, Heesen BJ, Corbett JA, McDaniel ML, Chang K, Allison
W, Wolffenbuttel BH, Williamson JR, Tilton RG: Inhibition of
nitric oxide formation by guanidines. Eur J Pharmacol 249(1):101–
106, 1993
74. Naka M, Nanbu T, Kobayashi K, Kamanaka Y, Komeno M, Yanase
R, Fukutomi T, Fujimura S, Seo HG, Fujiwara N, Ohuchida S,
Suzuki K, Kondo K, Taniguchi N: A potent inhibitor of inducible
nitric oxide synthase, ONO-1714, a cyclic amidine derivative.
Biochem Biophys Res Commun 270(2):663–667, 2000
75. Salzman AL: Nitric oxide in the gut. New Horiz 3(2):352–364,
1995
76. Kubes P and McCafferty DM: Nitric oxide and intestinal inflammation. Am J Med 109(2):150–158, 2000
77. Suzuki Y, Deitch EA, Mishima S, Lu Q, Xu D: Inducible nitric
oxide synthase gene knockout mice have increased resistance to
gut injury and bacterial translocation after an intestinal ischemiareperfusion injury. Crit Care Med 28(11):3692–3696, 2000
78. Cuzzocrea S, Zingarelli B, Caputi AP: Role of constitutive nitric oxide synthase and peroxynitrite production in a rat model
of splanchnic artery occlusion shock. Life Sci 63(9):789–799,
1998
79. Wu B, Iwakiri R, Tsunada S, Utsumi H, Kojima M, Fujise T,
Ootani A, Fujimoto K: iNOS enhances rat intestinal apoptosis
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
ISCHEMIA–REPERFUSION INJURY OF THE INTESTINE
80.
81.
82.
83.
84.
85.
86.
87.
88.
89.
90.
91.
92.
93.
94.
95.
96.
97.
after ischemia-reperfusion. Free Radic Biol Med 33(5):649–658,
2002
Virlos IT, Inglott FS, Williamson RC, Mathie RT: Differential expression of pulmonary nitric oxide synthase isoforms after intestinal ischemia-reperfusion. Hepatogastroenterology 50(49):31–36,
2003
Wink DA, Hanbauer I, Krishna MC, DeGraff W, Gamson J,
Mitchell JB: Nitric oxide protects against cellular damage and cytotoxicity from reactive oxygen species. Proc Natl Acad Sci USA
90(21):9813–9817, 1993
Kosonen O, Kankaanranta H, Malo-Ranta U, Moilanen E: Nitric
oxide-releasing compounds inhibit neutrophil adhesion to endothelial cells. Eur J Pharmacol 382(2):111–117, 1999
Gidday JM, Park TS, Shah AR, Gonzales ER: Modulation of basal
and postischemic leukocyte-endothelial adherence by nitric oxide.
Stroke 29(7):1423–1429, 1998
Guo JP, Murohara T, Buerke M, Scalia R, Lefer AM: Direct measurement of nitric oxide release from vascular endothelial cells. J
Appl Physiol 81(2):774–779, 1996
Lefer AM, Lefer DJ: The role of nitric oxide and cell adhesion
molecules on the microcirculation in ischaemia-reperfusion. Cardiovasc Res 32(4):743–751, 1996
Wink DA, Mitchell JB: Chemical biology of nitric oxide: Insights
into regulatory, cytotoxic, and cytoprotective mechanisms of nitric
oxide. Free Radic Biol Med 25(4–5):434–456, 1998
Gauthier TW, Davenpeck KL, Lefer AM: Nitric oxide attenuates leukocyte-endothelial interaction via P-selectin in splanchnic ischemia-reperfusion. Am J Physiol 267(4, Pt 1):G562–G568,
1994
Ito K, Ozasa H, Kojima N, Miura M, Iwa T, Senoo H, Horikawa
S: Pharmacological preconditioning protects lung injury induced
by intestinal ischemia/reperfusion in rat. Shock 19(5):462–468,
2003
Attuwaybi BO, Kozar RA, Moore-Olufemi SD, Sato N, Hassoun
HT, Weisbrodt NW, Moore FA: Heme oxygenase-1 induction by
hemin protects against gut ischemia/reperfusion injury. J Surg Res
118(1):53–57, 2004
Nakao A, Kimizuka K, Stolz DB, Neto JS, Kaizu T, Choi AM,
Uchiyama T, Zuckerbraun BS, Nalesnik MA, Otterbein LE, Murase
N: Carbon monoxide inhalation protects rat intestinal grafts from
ischemia/reperfusion injury. Am J Pathol 163(4):1587–1598, 2003
Squiers EC, Bruch D, Buelow R, Tice DG: Pretreatment of small
bowel isograft donors with cobalt-protoporphyrin decreases preservation injury. Transplant Proc 31(1–2):585–586, 1999
Choi AM, Alam J: Heme oxygenase-1: Function, regulation, and
implication of a novel stress-inducible protein in oxidant-induced
lung injury. Am J Respir Cell Mol Biol 15(1):9–19, 1996
Katori M, Busuttil RW, Kupiec-Weglinski JW: Heme oxygenase-1
system in organ transplantation. Transplantation 74(7):905–912,
2002
Stocker R, Yamamoto Y, McDonagh AF, Glazer AN, Ames BN:
Bilirubin is an antioxidant of possible physiological importance.
Science 235(4792):1043–1046, 1987
Balla G, Jacob HS, Balla J, Rosenberg M, Nath K, Apple F,
Eaton JW, Vercellotti GM: Ferritin: a cytoprotective antioxidant
strategem of endothelium. J Biol Chem 267(25):18148–18153,
1992
Willis D, Moore AR, Frederick R, Willoughby DA: Heme oxygenase: A novel target for the modulation of the inflammatory
response. Nat Med 2(1):87–90, 1996
Vachharajani TJ, Work J, Issekutz AC, Granger DN: Heme oxygenase modulates selectin expression in different regional vascular
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
98.
99.
100.
101.
102.
103.
104.
105.
106.
107.
108.
109.
110.
111.
112.
113.
114.
beds. Am J Physiol Heart Circ Physiol 278(5):H1613–H1617,
2000
Coito AJ, Buelow R, Shen XD, Amersi F, Moore C, Volk HD,
Busuttil RW, Kupiec-Weglinski JW: Heme oxygenase-1 gene
transfer inhibits inducible nitric oxide synthase expression and protects genetically fat Zucker rat livers from ischemia-reperfusion
injury. Transplantation 74(1):96–102, 2002
Ceran C, Sonmez K, Turkyllmaz Z, Demirogullarl B, Dursun
A, Duzgun E, Basaklar AC, Kale N: Effect of bilirubin in ischemia/reperfusion injury on rat small intestine. J Pediatr Surg
36(12):1764–1767, 2001
Hammerman C, Goldschmidt D, Caplan MS, Kaplan M, Bromiker
R, Eidelman AI, Gartner LM, Hochman A: Protective effect of
bilirubin in ischemia-reperfusion injury in the rat intestine. J Pediatr
Gastroenterol Nutr 35(3):344–349, 2002
Anaya-Prado R, Toledo-Pereyra LH, Lentsch AB, Ward PA:
Ischemia/reperfusion injury. J Surg Res 105(2):248–258, 2002
Zingarelli B, Sheehan M, Wong HR: Nuclear factor-kappaB as
a therapeutic target in critical care medicine. Crit Care Med
31(Suppl 1):S105–S111, 2003
Siebenlist U, Franzoso G, Brown K: Structure, regulation and function of NF-kappa B. Annu Rev Cell Biol 10:405–455, 1994
Collins T, Read MA, Neish AS, Whitley MZ, Thanos D,
Maniatis T: Transcriptional regulation of endothelial cell adhesion
molecules: NF-kappa B and cytokine-inducible enhancers. FASEB
J 9(10):899–909, 1995
Barnes PJ: Nuclear factor-kappa B. Int J Biochem Cell Biol
29(6):867–870, 1997
Blackwell TS, Christman JW: The role of nuclear factor-kappa B
in cytokine gene regulation. Am J Respir Cell Mol Biol 17(1):3–9,
1997
Spiecker M, Darius H, Kaboth K, Hubner F, Liao JK: Differential
regulation of endothelial cell adhesion molecule expression by nitric oxide donors and antioxidants. J Leukoc Biol 63(6):732–739,
1998
Peng HB, Libby P, Liao JK: Induction and stabilization of I kappa
B alpha by nitric oxide mediates inhibition of NF-kappa B. J Biol
Chem 270(23):14214–14219, 1995
Austen Jr WG, Kobzik L, Carroll MC, Hechtman HB, Moore Jr FD:
The role of complement and natural antibody in intestinal ischemiareperfusion injury. Int J Immunopathol Pharmacol 16(1):1–8,
2003
Heller T, Hennecke M, Baumann U, Gessner JE, zu Vilsendorf
AM, Baensch M, Boulay F, Kola A, Klos A, Bautsch W, Kohl
J: Selection of a C5a receptor antagonist from phage libraries
attenuating the inflammatory response in immune complex disease and ischemia/reperfusion injury. J Immunol 163(2):985–994,
1999
Kimura T, Andoh A, Fujiyama Y, Saotome T, Bamba T: A blockade of complement activation prevents rapid intestinal ischaemiareperfusion injury by modulating mucosal mast cell degranulation
in rats. Clin Exp Immunol 111(3):484–490, 1998
Wada K, Montalto MC, Stahl GL: Inhibition of complement
C5 reduces local and remote organ injury after intestinal ischemia/reperfusion in the rat. Gastroenterology 120(1):126–133,
2001
Gibbs SA, Weiser MR, Kobzik L, Valeri CR, Shepro D,
Hechtman HB: P-selectin mediates intestinal ischemic injury
by enhancing complement deposition. Surgery 119(6):652–656,
1996
Montalto MC, Hart ML, Jordan JE, Wada K, Stahl GL: Role for
complement in mediating intestinal nitric-oxide synthase-2 and
1373
MALLICK ET AL.
115.
116.
117.
118.
119.
120.
121.
122.
123.
124.
125.
126.
127.
128.
129.
130.
131.
132.
superoxide dismutase expression. Am J Physiol Gastrointest Liver
Physiol, 285(1):G 197–206, 2003
Hernandez LA, Grisham MB, Granger DN: A role for iron in
oxidant-mediated ischemic injury to intestinal microvasculature.
Am J Physiol 253(1, Pt 1):G49–G53, 1987
Lelli JL Jr, Pradhan S, Cobb LM: Prevention of postischemic injury
in immature intestine by deferoxamine. J Surg Res 54(1):34–38,
1993
Richard V, Kaeffer N, Tron C, Thuillez C: Ischemic preconditioning protects against coronary endothelial dysfunction induced by
ischemia and reperfusion. Circulation 89(3):1254–1261, 1994
Murry CE, Jennings RB, Reimer KA: Preconditioning with ischemia: A delay of lethal cell injury in ischemic myocardium.
Circulation 74(5):1124–1136, 1986
Koti RS, Seifalian AM, McBride AG, Yang W, Davidson BR:
The relationship of hepatic tissue oxygenation with nitric oxide
metabolism in ischemic preconditioning of the liver. FASEB J
16(12):1654–1656, 2002
Pang CY, Neligan P, Zhong A, He W, Xu H, Forrest CR: Effector mechanism of adenosine in acute ischemic preconditioning of
skeletal muscle against infarction. Am J Physiol 273(3, Pt 2):R887–
R895, 1997
Glazier SS, O’Rourke DM, Graham DI, Welsh FA: Induction of
ischemic tolerance following brief focal ischemia in rat brain. J
Cereb Blood Flow Metab 14(4):545–553, 1994
Sakurai M, Hayashi T, Abe K, Aoki M, Sadahiro M, Tabayashi
K: Enhancement of heat shock protein expression after transient
ischemia in the preconditioned spinal cord of rabbits. J Vasc Surg
27(4):720–725, 1998
Turman MA, Bates CM: Susceptibility of human proximal tubular
cells to hypoxia: effect of hypoxic preconditioning and comparison
to glomerular cells. Ren Fail 19(1):47–60, 1997
Du ZY, Hicks M, Winlaw D, Spratt P, Macdonald P: Ischemic
preconditioning enhances donor lung preservation in the rat. J Heart
Lung Transplant 15(12):1258–1267, 1996
Li Y, Roth S, Laser M, Ma JX, Crosson CE: Retinal preconditioning
and the induction of heat-shock protein 27. Invest Ophthalmol Vis
Sci 44(3):1299–1304, 2003
Sola A, De Oca J, Gonzalez R, Prats N, Rosello-Catafau J, Gelpi E,
Jaurrieta E, Hotter G: Protective effect of ischemic preconditioning
on cold preservation and reperfusion injury associated with rat
intestinal transplantation. Ann Surg 234(1):98–106, 2001
Davis JM, Gute DC, Jones S, Krsmanovic A, Korthuis RJ: Ischemic
preconditioning prevents postischemic P-selectin expression in the
rat small intestine. Am J Physiol 277(6, Pt 2):H2476–H2481, 1999
Vlasov TD, Smirnov DA, Nutfullina GM: Preconditioning of
the small intestine to ischemia in rats. Neurosci Behav Physiol
32(4):449–453, 2002
Jenkins DP, Pugsley WB, Alkhulaifi AM, Kemp M, Hooper J,
Yellon DM: Ischaemic preconditioning reduces troponin T release in patients undergoing coronary artery bypass surgery. Heart
77(4):314–318, 1997
Clavien PA, Selzner M, Rudiger HA, Graf R, Kadry Z, Rousson
V, Jochum W: A prospective randomized study in 100 consecutive patients undergoing major liver resection with versus without
ischemic preconditioning. Ann Surg 238(6):843–850, 2003
Hotter G, Closa D, Prados M, Fernandez-Cruz L, Prats N, Gelpi
E, Rosello-Catafau J: Intestinal preconditioning is mediated by a
transient increase in nitric oxide. Biochem Biophys Res Commun
222(1):27–32, 1996
Unal S, Demirkan F, Arslan E, Cin I, Cinel L, Eskandari G,
Cinel I: Comparison of ischemic and chemical preconditioning
1374
133.
134.
135.
136.
137.
138.
139.
140.
141.
142.
143.
144.
145.
146.
147.
148.
149.
in jejunal flaps in the rat. Plast Reconstr Surg 112(4):1024–1031,
2003
McCallion K, Wattanasirichaigoon S, Gardiner KR, Fink MP: Ischemic preconditioning ameliorates ischemia- and reperfusioninduced intestinal epithelial hyperpermeability in rats. Shock
14(4):429–434, 2000
Cohen MV, Baines CP, Downey JM: Ischemic preconditioning:
From adenosine receptor of KATP channel. Annu Rev Physiol
62:79–109, 2000
Bolli R: The late phase of preconditioning. Circ Res 87(11):972–
983, 2000
Yellon DM, Dana A: The preconditioning phenomenon: A tool for
the scientist or a clinical reality? Circ Res 87(7):543–550, 2000
Ferencz A, Szanto Z, Kalmar-Nagy K, Horvath OP, Roth E: Mitigation of oxidative injury by classic and delayed ischemic preconditioning prior to small bowel autotransplantation. Transplant Proc
36(2):286–288, 2004
Ferencz A, Szanto Z, Borsiczky B, Kiss K, Kalmar-Nagy K, Szeberenyi J, Horvath PO, Roth E: The effects of preconditioning on
the oxidative stress in small-bowel autotransplantation. Surgery
132(5):877–884, 2002
Tamion F, Richard V, Lacoume Y, Thuillez C: Intestinal preconditioning prevents systemic inflammatory response in hemorrhagic
shock. Role of HO-1. Am J Physiol Gastrointest Liver Physiol
283(2):G408–G414, 2002
Wu B, Ootani A, Iwakiri R, Fujise T, Tsunada S, Toda S, Fujimoto
K: Ischemic preconditioning attenuates ischemia-reperfusioninduced mucosal apoptosis by inhibiting the mitochondriadependent pathway in rat small intestine. Am J Physiol Gastrointest
Liver Physiol 286(4):G580–G587, 2004
Sola A, Hotter G, Prats N, Xaus C, Gelpi E, Rosello-Catafau J:
Modification of oxidative stress in response to intestinal preconditioning. Transplantation 69(5):767–772, 2000
Cinel I, Avlan D, Cinel L, Polat G, Atici S, Mavioglu I, Serinol H,
Aksoyek S, Oral U: Ischemic preconditioning reduces intestinal
epithelial apoptosis in rats. Shock 19(6):588–592, 2003
Ahmadinejad M, Rex M, Sutton RH, Pollitt CC, Cribb B: The
effects of allopurinol on the ultrastructure of ischaemic and
reperfused large intestine of sheep. Aust Vet J 74(2):135–139,
1996
Cuzzocrea S, Mazzon E, Dugo L, Caputi AP, Aston K, Riley DP,
Salvemini D: Protective effects of a new stable, highly active SOD
mimetic, M40401 in splanchnic artery occlusion and reperfusion.
Br J Pharmacol 132(1):19–29, 2001
Balogh N, Krausz F, Levai P, Ribiczeyne PS, Vajdovich P, Gaal T:
Effect of deferoxamine and L-arginine treatment on lipid peroxidation in an intestinal ischaemia-reperfusion model in rats. Acta
Vet Hung 50(3):343–356, 2002
Cuzzocrea S, Mazzon E, Costantino G, Serraino I, De Sarro
A, Caputi AP: Effects of n-acetylcysteine in a rat model of ischemia and reperfusion injury. Cardiovasc Res 47(3):537–548,
2000
Yamaguchi T, Dayton C, Shigematsu T, Carter P, Yoshikawa T,
Gute DC, Korthuis RJ: Preconditioning with ethanol prevents
postischemic leukocyte-endothelial cell adhesive interactions. Am
J Physiol Heart Circ Physiol 283(3):H1019–H1030, 2002
Nakamura M, Ozaki M, Fuchinoue S, Teraoka S, Ota K: Ascorbic
acid prevents ischemia-reperfusion injury in the rat small intestine.
Transpl Int 10(2):89–95, 1997
Gunel E, Caglayan F, Caglayan O, Dilsiz A, Duman S, Aktan
M: Treatment of intestinal reperfusion injury using antioxidative
agents. J Pediatr Surg 33(10):1536–1539, 1998
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
ISCHEMIA–REPERFUSION INJURY OF THE INTESTINE
150. Sener G, Akgun U, Satiroglu H, Topaloglu U, Keyer-Uysal M: The
effect of pentoxifylline on intestinal ischemia/reperfusion injury.
Fundam Clin Pharmacol 15(1):19–22, 2001
151. Hammerman C, Goldschmidt D, Caplan MS, Kaplan M, Schimmel
MS, Eidelman AI, Branski D, Hochman A: Amelioration of
ischemia-reperfusion injury in rat intestine by pentoxifyllinemediated inhibition of xanthine oxidase. J Pediatr Gastroenterol
Nutr 29(1):69–74, 1999
152. Mocan H, Gedik Y, Erduran E, Mocan ZM, Okten A, Gacar N,
Pul N: The role of calcium channel entry blocker in experimental
ischemia-reperfusion-induced intestinal injury. Pol J Pharmacol
47(2):179–183, 1995
153. Simpson R, Alon R, Kobzik L, Valeri CR, Shepro D, Hechtman
HB: Neutrophil and nonneutrophil-mediated injury in intestinal
ischemia-reperfusion. Ann Surg 218(4):444–453, 1993
154. Kojima M, Iwakiri R, Wu B, Fujise T, Watanabe K, Lin T,
Amemori S, Sakata H, Shimoda R, Oguzu T, Ootani A, Tsunada
S, Fujimoto K: Effects of antioxidative agents on apoptosis induced by ischaemia-reperfusion in rat intestinal mucosa. Aliment
Pharmacol Ther 18 (Suppl 1):139–145, 2003
155. Tomatsuri N, Yoshida N, Takagi T, Katada K, Isozaki Y, Imamoto
E, Uchiyama K, Kokura S, Ichikawa H, Naito Y, Okanoue T,
Yoshikawa T: Edaravone, a newly developed radical scavenger,
protects against ischemia-reperfusion injury of the small intestine
in rats. Int J Mol Med 13(1):105–109, 2004
156. Kawata K, Takeyoshi I, Iwanami K, Sunose Y, Aiba M, Ohwada
S, Matsumoto K, Morishita Y: A spontaneous nitric oxide donor
ameliorates small bowel ischemia-reperfusion injury in dogs. Dig
Dis Sci 46(8):1748–1756, 2001
157. Aoki N, Johnson G, III, Lefer AM: Beneficial effects of two forms
of NO administration in feline splanchnic artery occlusion shock.
Am J Physiol 258(2, Pt 1):G275–G281, 1990
158. Kalia N, Brown NJ, Hopkinson K, Stephenson TJ, Wood RF,
Pockley AG: FK409 inhibits both local and remote organ damage after intestinal ischaemia. J Pathol 197(5):595–602, 2002
159. Fox-Robichaud A, Payne D, Hasan SU, Ostrovsky L, Fairhead T,
Reinhardt P, Kubes P: Inhaled NO as a viable antiadhesive therapy
for ischemia/reperfusion injury of distal microvascular beds. J Clin
Invest 101(11):2497–2505, 1998
160. Luo CC, Chen HM, Chiu CH, Lin JN, Chen JC: Effect of N(G)nitro-L-arginine methyl ester on intestinal permeability following
intestinal ischemia-reperfusion injury in a rat model. Biol Neonate
80(1):60–63, 2001
161. Naito Y, Takagi T, Ichikawa H, Tomatsuri N, Kuroda M, Isozaki
Y, Katada K, Uchiyama K, Kokura S, Yoshida N, Okanoue T,
Yoshikawa T: A novel potent inhibitor of inducible nitric oxide inhibitor, ONO-1714, reduces intestinal ischemia-reperfusion injury
in rats. Nitric Oxide 10(3):170–177, 2004
162. Khanna A, Rossman J, Caty MG, Fung HL: Beneficial effects of
intraluminal nitroglycerin in intestinal ischemia-reperfusion injury
in rats. J Surg Res 114(1):15–24, 2003
163. Arumugam TV, Shiels IA, Woodruff TM, Reid RC, Fairlie DP,
Taylor SM: Protective effect of a new C5a receptor antagonist
against ischemia-reperfusion injury in the rat small intestine. J Surg
Res 103(2):260–267, 2002
164. Zhao H, Montalto MC, Pfeiffer KJ, Hao L, Stahl GL: Murine
model of gastrointestinal ischemia associated with complementdependent injury. J Appl Physiol 93(1):338–345, 2002
165. Williams JP, Pechet TT, Weiser MR, Reid R, Kobzik L, Moore
FD Jr, Carroll MC, Hechtman HB: Intestinal reperfusion injury is
mediated by IgM and complement. J Appl Physiol 86(3):938–942,
1999
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
166. Karpel-Massler G, Fleming SD, Kirschfink M, Tsokos GC:
Human C1 esterase inhibitor attenuates murine mesenteric ischemia/reperfusion induced local organ injury. J Surg Res
115(2):247–256, 2003
167. Souza DG, Coutinho SF, Silveira MR, Cara DC, Teixeira MM: Effects of a BLT receptor antagonist on local and remote reperfusion
injuries after transient ischemia of the superior mesenteric artery
in rats. Eur J Pharmacol 403(1–2):121–128, 2000
168. Souza DG, Cara DC, Cassali GD, Coutinho SF, Silveira MR,
Andrade SP, Poole SP, Teixeira MM: Effects of the PAF receptor antagonist UK74505 on local and remote reperfusion injuries
following ischaemia of the superior mesenteric artery in the rat. Br
J Pharmacol 131(8):1800–1808, 2000
169. Sun Z, Olanders K, Lasson A, Dib M, Annborn M, Andersson
K, Wang X, Andersson R: Effective treatment of gut barrier dysfunction using an antioxidant, a PAF inhibitor, and monoclonal
antibodies against the adhesion molecule PECAM-1. J Surg Res
105(2):220–233, 2002
170. Souza DG, Cassali GD, Poole S, Teixeira MM: Effects of inhibition
of PDE4 and TNF-alpha on local and remote injuries following
ischaemia and reperfusion injury. Br J Pharmacol 134(5):985–994,
2001
171. Zou L, Attuwaybi B, Kone BC: Effects of NF-kappa B inhibition on
mesenteric ischemia-reperfusion injury. Am J Physiol Gastrointest
Liver Physiol 284(4):G713–G721, 2003
172. Hassoun HT, Zou L, Moore FA, Kozar RA, Weisbrodt NW,
Kone BC: Alpha-melanocyte-stimulating hormone protects against
mesenteric ischemia-reperfusion injury. Am J Physiol Gastrointest
Liver Physiol 282(6):G1059–G1068, 2002
173. Riaz AA, Wan MX, Schafer T, Dawson P, Menger MD, Jeppsson
B, Thorlacius H: Allopurinol and superoxide dismutase protect
against leucocyte-endothelium interactions in a novel model of
colonic ischaemia-reperfusion. Br J Surg 89(12):1572–1580, 2002
174. Harris NR, Granger DN: Organ procurements and preservation
for transplantation. In: Ischemia/Reperfusion injury. New York:
Springer Verlag, 1997, pp. 67–81
175. Carmody IC, Meng L, Shen XD, Anselmo D, Gao F, Ke B, Ma
JP, Kupiec-Weglinski JW, McDiarmid SV, Busuttil RW, Shaw G,
Farmer DG: P-Selectin knockout mice have improved outcomes
with both warm ischemia and small bowel transplantation. Transplant Proc 36(2):263–264, 2004
176. Bowles MJ, Pockley AG, Wood RF: Effect of anti-LFA-1 monoclonal antibody on rat small bowel allograft survival and circulating
leukocyte populations. Transpl Immunol 8(1):75–80, 2000
177. Floyd TF, Boroughs A, Garvey C, Dasher J, Ikeda CB, Sloviter HA,
Ziegler MM: Intestinal ischemia: treatment by peritoneal lavage
with oxygenated perfluorochemical. J Pediatr Surg 22(12):1191–
1197, 1987
178. Oldham KT, Guice KS, Gore D, Gourley WK, Lobe TE: Treatment
of intestinal ischemia with oxygenated intraluminal perfluorocarbons. Am J Surg 153(3):291–294, 1987
179. Ricci JL, Sloviter HA, Ziegler MM: Intestinal ischemia: Reduction
of mortality utilizing intraluminal perfluorochemical. Am J Surg
149(1):84–90, 1985
180. O’Donnell KA, Caty MG, Zheng S, Rossman JE, Azizkhan RG:
Oxygenated intraluminal perfluorocarbon protects intestinal muscosa from ischemia/reperfusion injury. J Pediatr Surg 32(2):361–
365, 1997
181. Ohara M, Unno N, Mitsuoka H, Kaneko H, Nakamura S: Peritoneal
lavage with oxygenated perfluorochemical preserves intestinal mucosal barrier function after ischemia-reperfusion and ameliorates
lung injury. Crit Care Med 29(4):782–788, 2001
1375
MALLICK ET AL.
182. Vejchapipat P, Proctor E, Ramsay A, Petros A, Gadian DG, Spitz L,
Pierro A: Intestinal energy metabolism after ischemia-reperfusion:
Effects of moderate hypothermia and perfluorocarbons. J Pediatr
Surg 37(5):786–790, 2002
183. Fujino Y, Suzuki Y, Kakinoki K, Tanioka Y, Ku Y, Kuroda Y: Protection against experimental small intestinal ischaemia-reperfusion
injury with oxygenated perfluorochemical. Br J Surg 90(8):1015–
1020, 2003
184. Shashikant MP, Badellino MM, Cooper B, Shaffer TH, Myers
SI, Wolfson MR: Physicochemical properties of perfluorochemical
liquids influence ventilatory requirements, pulmonary mechanics,
and microvascular permeability during partial liquid ventilation
following intestinal ischemia/reperfusion injury. Crit Care Med
30(10):2300–2305, 2002
185. Fukatsu K, Zarzaur BL, Johnson CD, Lundberg AH, Wilcox HG,
Kudsk KA: Enteral nutrition prevents remote organ injury and
death after a gut ischemic insult. Ann Surg 233(5):660–668, 2001
186. Fukatsu K, Lundberg AH, Hanna MK, Wu Y, Wilcox HG, Granger
DN, Gaber AO, Kudsk KA: Increased expression of intestinal Pselectin and pulmonary E-selectin during intravenous total parenteral nutrition. Arch Surg 135(10):1177–1182, 2000
187. Wu Y, Kudsk KA, DeWitt RC, Tolley EA, Li J: Route and type of
nutrition influence IgA-mediating intestinal cytokines. Ann Surg
229(5):662–667, 1999
188. Lacey JM and Wilmore DW: Is glutamine a conditionally essential
amino acid? Nutr Rev 48(8):297–309, 1990
189. Askanazi J, Carpentier YA, Michelsen CB, Elwyn DH, Furst P,
Kantrowitz LR, Gump FE, Kinney JM: Muscle and plasma amino
acids following injury. Influence of intercurrent infection. Ann Surg
192(1):78–85, 1980
190. Wilmore DW: Growth factors and nutrients in the short bowel
syndrome. JPEN J Parenter Enteral Nutr 23(Suppl 5):S117–S120,
1999
191. Ziegler TR, Estivariz CF, Jonas CR, Gu LH, Jones DP, Leader
LM: Interactions between nutrients and peptide growth factors in
intestinal growth, repair, and function. JPEN J Parenter Enteral
Nutr 23(Suppl 6):S174–S183, 1999
192. Li YS, Li JS, Jiang JW, Liu FN, Li N, Qin WS, Zhu H: Glycylglutamine-enriched long-term total parenteral nutrition attenuates
bacterial translocation following small bowel transplantation in the
pig. J Surg Res 82(1):106–111, 1999
193. Sakawaki T, Sasaki K, Hirata K: Saline with glutamine improves cold preserved small bowel graft mortality. Transplant Proc
30(7):3471–3474, 1998
194. Tamaki T, Konoeda Y, Yasuhara M, Tanaka M, Yokota N, Hayashi
T, Katori M, Uchida Y, Kawamura A: Glutamine-induced heme
oxygenase-1 protects intestines and hearts from warm ischemic
injury. Transplant Proc 31(1–2):1018–1019, 1999
195. Zhang W, Bain A, Rombeau JL: Insulin-like growth factor-I (IGF-I)
and glutamine improve structure and function in the small bowel
allograft. J Surg Res 59(1):6–12, 1995
196. Ikeda S, Zarzaur BL, Johnson CD, Fukatsu K, Kudsk KA: Total
parenteral nutrition supplementation with glutamine improves survival after gut ischemia/reperfusion. JPEN J Parenter Enteral Nutr
26(3):169–173, 2002
197. Wischmeyer PE: Glutamine and heat shock protein expression.
Nutrition 18(3):225–228, 2002
198. Kelly D, Wischmeyer PE: Role of L-glutamine in critical illness:
new insights. Curr Opin Clin Nutr Metab Care 6(2):217–222, 2003
199. Harward TR, Coe D, Souba WW, Klingman N, Seeger JM:
Glutamine preserves gut glutathione levels during intestinal ischemia/reperfusion. J Surg Res 56(4):351–355, 1994
1376
200. Lee MA, McCauley RD, Kong SE, Hall JC: Pretreatment
with glycine reduces the severity of warm intestinal ischemicreperfusion injury in the rat. Ann Plast Surg 46(3):320–326, 2001
201. Lee MA, McCauley RD, Kong SE, Hall JC: Influence of glycine
on intestinal ischemia-reperfusion injury. JPEN J Parenter Enteral
Nutr 26(2):130–135, 2002
202. Grotz MR, Pape HC, van Griensven M, Stalp M, Rohde F, Bock D,
Krettek C: Glycine reduces the inflammatory response and organ
damage in a two-hit sepsis model in rats. Shock 16(2):116–121,
2001
203. Kelly D, Zhong Z, Wheeler MD, Li X, Froh M, Schemmer P,
Yin M, Bunzendaul H, Bradford B, Lemasters JJ: L-Glycine:
A novel antiinflammatory, immunomodulatory, and cytoprotective agent. Curr Opin Clin Nutr Metab Care 6(2):229–240,
2003
204. Jacob T, Ascher E, Hingorani A, Kallakuri S: Glycine prevents the induction of apoptosis attributed to mesenteric ischemia/reperfusion injury in a rat model. Surgery 134(3):457–466,
2003
205. Ferrer JV, Ariceta J, Guerrero D, Gomis T, Larrea MM, Balen
E, Lera JM: Allopurinol and N-acetylcysteine avoid 60% of intestinal necrosis in an ischemia-reperfusion experimental model.
Transplant Proc 30(6):2672, 1998
206. Ozden A, Tetik C, Bilgihan A, Calli N, Bostanci B, Yis O, Duzcan
E: Antithrombin III prevents 60 min warm intestinal ischemia
reperfusion injury in rats. Res Exp Med (Berl) 198(5):237–246,
1999
207. Puglisi RN, Strande L, Santos M, Doolin EJ, Hewitt CW, Whalen
TV: The effect of cyclosporine in gut ischemic injury: a computerized morphometric and enzymatic analysis. J Pediatr Surg
31(2):319–322, 1996
208. Berlanga J, Prats P, Remirez D, Gonzalez R, Lopez-Saura P, Aguiar
J, Ojeda M, Boyle JJ, Fitzgerald AJ, Playford RJ: Prophylactic use
of epidermal growth factor reduces ischemia/reperfusion intestinal
damage. Am J Pathol 161(2):373–379, 2002
209. Ustundag B, Kazez A, Demirbag M, Canatan H, Halifeoglu I,
Ozercan IH: Protective effect of melatonin on antioxidative system in experimental ischemia-reperfusion of rat small intestine.
Cell Physiol Biochem 10(4):229–236, 2000
210. Kazez A, Demirbag M, Ustundag B, Ozercan IH, Saglam M: The
role of melatonin in prevention of intestinal ischemia-reperfusion
injury in rats. J Pediatr Surg 35(10):1444–1448, 2000
211. Dun Y, Hao YB, Wu YX, Zhang Y, Zhao RR: Protective effects
of nitroglycerin-induced preconditioning mediated by calcitonin
gene-related peptide in rat small intestine. Eur J Pharmacol 430(2–
3):317–324, 2001
212. Cicalese L, Lee K, Schraut W, Watkins S, Borle A, Stanko R:
Pyruvate prevents ischemia-reperfusion mucosal injury of rat small
intestine. Am J Surg 171(1):97–100, 1996
213. Tetik C, Ozden A, Calli N, Bilgihan A, Bostanci B, Yis O,
Bayramoglu H: Cytoprotective effect of trimetazidine on 60 minutes of intestinal ischemia-reperfusion injury in rats. Transpl Int
12(2):108–112, 1999
214. Tsuruma T, Yagihashi A, Hirata K, Matsuno T, Zou XM, Sasaki K,
Asanuma K, Endo T: Evaluation of plasma IL-8 (CINC) concentration during ischemia and after reperfusion in the small intestine.
Transplant Proc 28(3):1917–1918, 1996
215. Droy-Lefaix MT, Drouet Y, Geraud G, Hosford D, Braquet
P: Superoxide dismutase (SOD) and the PAF-antagonist (BN
52021) reduce small intestinal damage induced by ischemiareperfusion. Free Radic Res Commun 12–13 (Pt 2):725–735,
1991
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
ISCHEMIA–REPERFUSION INJURY OF THE INTESTINE
216. Souza DG, Mendonca VA, de ACM, Poole S, Teixeira MM: Role of
tachykinin NK receptors on the local and remote injuries following
ischaemia and reperfusion of the superior mesenteric artery in the
rat. Br J Pharmacol 135(2):303–312, 2002
217. Yagihashi A, Tsuruma T, Tarumi K, Kameshima T, Yajima T,
Yanai Y, Watanabe N, Hirata K: Prevention of small intestinal
ischemia-reperfusion injury in rat by anti-cytokine-induced neutrophil chemoattractant monoclonal antibody. J Surg Res 78(2):92–
96, 1998
218. Chen Y, Hung WT, Chen SM, Tseng SH: Suppression of elevated
plasma interleukin-8 levels due to total ischemia and reperfusion
of the small intestine by luminal perfusion with fetal bovine serum.
Pediatr Surg Int 18(2–3):107–109, 2002
219. Mazzon E, Dugo L, De SA, Li JH, Caputi AP, Zhang J, Cuzzocrea
S: Beneficial effects of GPI 6150, an inhibitor of poly(ADP-ribose)
polymerase in a rat model of splanchnic artery occlusion and reperfusion. Shock 17(3):222–227, 2002
220. Xia G, Martin AE, Besner GE: Heparin-binding EGFlike growth factor downregulates expression of adhesion
molecules and infiltration of inflammatory cells after intestinal ischemia/reperfusion injury. J Pediatr Surg 38(3):434–439,
2003
221. Kuenzler KA, Pearson PY, Schwartz MZ: Hepatoctye growth factor
pretreatment reduces apoptosis and mucosal damage after intestinal
ischemia-reperfusion. J Pediatr Surg 37(7):1093 [discussion], 2002
222. Yang ZJ, Bosco G, Montante A, Ou XI, Camporesi EM: Hyperbaric O2 reduces intestinal ischemia-reperfusion-induced TNFalpha production and lung neutrophil sequestration. Eur J Appl
Physiol 85(1–2):96–103, 2001
223. Lane JS, Todd KE, Lewis MP, Gloor B, Ashley SW, Reber
HA, McFadden DW, Chandler CF: Interleukin-10 reduces the
Digestive Diseases and Sciences, Vol. 49, No. 9 (September 2004)
224.
225.
226.
227.
228.
229.
230.
systemic inflammatory response in a murine model of intestinal
ischemia/reperfusion. Surgery 122(2):288–294, 1997
Dowdall JF, Winter DC, Bouchier-Hayes DJ: Inosine modulates gut
barrier dysfunction and end organ damage in a model of ischemiareperfusion injury. J Surg Res 108(1):61–68, 2002
Stroh R, Christopher TA, Lopez BL, Guo YP, Amico-Roxas M,
Ma XL: L-Propionyl carnitine, an endogenous ester in fatty acid
metabolism, exerts anti-shock and endothelial protective effects in
rat splanchnic ischemia-reperfusion injury. Shock 9(3):216–222,
1998
Cuzzocrea S, Tailor A, Zingarelli B, Salzman AL, Flower RJ, Szabo
C, Perretti M: Lipocortin 1 protects against splanchnic artery occlusion and reperfusion injury by affecting neutrophil migration. J
Immunol 159(10):5089–5097, 1997
Warnecke HB, Schirmeier A, Nussler AK, Platz KP, Stange B,
Nussler NC, Radke C, Neuhaus P, Mueller AR: The combined treatment with L-arginine and methylprednisolone improves graft morphology and mucosal barrier function. Transplant Proc 34(3):996–
998, 2002
Loong CC, Chiu JH, Tiao RC, Chiu YY, Wu CW, Lui WY: Pretreatment with magnolol attenuates ischemia-reperfusion injury in
rat small intestine. Transplant Proc 33(7–8):3737–3738, 2001
Farmer DG, Amersi F, Shen XD, Gao F, Anselmo D, Ma J, Dry
S, McDiarmid SV, Shaw G, Busuttil RW, Kupiec-Weglinski J: Improved survival through the reduction of ischemia-reperfusion injury after rat intestinal transplantation using selective P-selectin
blockade with P-selectin glycoprotein ligand-Ig. Transplant Proc
34(3):985, 2002
Arumugam TV, Shiels IA, Margolin SB, Taylor SM, Brown L:
Pirfenidone attenuates ischaemia-reperfusion injury in the rat small
intestine. Clin Exp Pharmacol Physiol 29(11):996–1000, 2002
1377