You are on page 1of 53

Articles in PresS. Am J Physiol Gastrointest Liver Physiol (November 20, 2014). doi:10.1152/ajpgi.00112.

2013

1 Animal models of ischemia-reperfusion induced intestinal injury: progress and promise

2 for translational research

4 Liara M. Gonzalez,1 Adam J. Moeser,2 Anthony T. Blikslager1

5 Center for Comparative Medicine and Translational Research, 1Department of Clinical Sciences,
2
6 Department of Population Health and Pathobiology, North Carolina State University, Raleigh,

7 NC, USA

9 Correspondence:

10 Anthony T. Blikslager

11 College of Veterinary Medicine

12 1060 William Moore Drive

13 Raleigh, NC 27607

14 Telephone: 919-513-7725

15 Fax: 919-513-6336

16 Email: Anthony_Blikslager@ncsu.edu

17

18 Running Title: Animal Models of Ischemic Injury

19

20 Abstract

21 Research in the field of ischemia-reperfusion injury continues to be plagued by the inability to

22 translate research findings to clinically useful therapies. This may in part relate to the complexity

23 of disease processes that result in intestinal ischemia, but may also result from inappropriate

24 research model selection. Research animal models have been integral to the study of ischemia-

25 reperfusion induced intestinal injury. However, the clinical conditions that compromise intestinal

26 blood flow in clinical patients ranges widely from primary intestinal disease to processes

Copyright © 2014 by the American Physiological Society.


27 secondary to distant organ failure and generalized systemic disease. Thus, models that closely

28 resemble human pathology in clinical conditions as disparate as volvulus, shock, and

29 necrotizing enterocolitis are likely to give the greatest opportunity to understand mechanisms of

30 ischemia that may ultimately translate to patient care. Furthermore, conditions that result in

31 varying levels of ischemia may be further complicated by the reperfusion of blood to tissues

32 that, in some cases, further exacerbates injury. This review assesses animal models of

33 ischemia-reperfusion injury as well as the knowledge that has been derived from each in order

34 to aid selection of appropriate research models. In addition, a discussion of the future of

35 intestinal ischemia-reperfusion research is provided to place some context on the areas likely to

36 provide the greatest benefit from continued research of ischemia-reperfusion injury.

37

38 Introduction

39 The survival rate for human and veterinary patients with ischemic intestinal injury is generally

40 less than 50% (17, 69, 172, 183) because tissue hypoxia, inflammation and cell infiltration result

41 in loss of the mucosal barrier. This barrier is primarily composed of a single layer of columnar

42 epithelial cells bound together by interepithelial junctions, which collectively prevents the

43 translocation of bacteria and associated toxins into the systemic circulation (140). Intestinal

44 ischemia is associated with a broad range of clinical conditions including neonatal necrotizing

45 enterocolitis (125), acute mesenteric ischemia (AMI) (183), volvulus (155), trauma (41),

46 cardiopulmonary disease (151), hemorrhagic shock (41, 53) and intestinal transplant rejection

47 (112) (Figure 1). Ischemic injury may be coupled with subsequent reperfusion of tissue that is

48 known to exacerbate injury in some of these disease processes (112). Since intestinal ischemia

49 is rarely preventable, most research in the field has focused on advancing techniques for early

50 detection of ischemia and the development of novel therapeutic approaches that target the post-

51 ischemic insult (the reperfusion period). Injury attributed to reperfusion is thought to be primarily

52 attributable to reactive oxygen metabolites, associated with activation of oxidant-producing


53 mucosal enzymes, release of lipid chemoattractants from injured cellular membranes, and

54 subsequent infiltration of neutrophils (Figure 2). Therefore, proposed therapeutic interventions to

55 protect against reperfusion injury have targeted inhibition of oxidant injury and of neutrophil

56 activation (144, 157). However, due to the relative failure of clinical therapy directed at

57 reperfusion injury following early studies, the importance of targeting reperfusion injury in cases

58 of intestinal ischemic injury has been brought into question (105). The most recent clinical

59 perspectives on the efficacy of these treatments are derived from studies of tissues more

60 commonly affected by reperfusion, such as the myocardium following infarction (7). These too

61 have failed to demonstrate the clinical utility of these therapies. Encouragingly, recent studies

62 have recognized the modulation of cell death pathways as a novel target for therapeutic

63 intervention (87, 181). Mechanisms of cell death mediated by necrosis and apoptosis have been

64 shown to further exacerbate injury induced by ischemia and reperfusion. In fact, insufficient

65 clearance of these dying cells has been shown to lead to increased inflammation and impaired

66 tissue repair (181). As a result, there has been a shift of emphasis toward modulation of

67 pathways of cell death (87, 181) and regenerative processes (113, 161). However, initial

68 consideration of the animal model to select for study is of paramount importance, given the

69 highly variable clinical presentation of diseases that involve ischemia. Animal models have been

70 indispensable to the study of mechanisms of ischemia and reperfusion injury (22, 39, 134-137),

71 but each model has distinct advantages and disadvantages so that none of these models can

72 perfectly recapitulate the natural onset and progression of human disease (Table 1 and 2). The

73 aim of this review is to provide an in depth assessment of key animal models used for the study

74 of ischemia-reperfusion injury, with the goal of allowing investigators to understand and select

75 from available models for translational research. In addition, the future of intestinal regenerative

76 medicine in light of recent advances will be discussed.

77 Mechanisms of Ischemia
78 Ischemia is the reduction or complete occlusion of blood flow to a target organ that results in a

79 state of tissue oxygen deprivation. In the intestine, overt obstruction of the blood supply can

80 result from mesenteric vascular occlusion derived from thrombus formation or emboli secondary

81 to cardiopulmonary disease (143, 166). Compromise of normal blood flow to the intestine can

82 also be associated with severe intestinal distension and mechanical obstruction as occurs in

83 cases of intestinal strangulation associated with hernia, volvulus and intussusception (1, 183).

84 Additionally, ischemic injury is thought to contribute to the pathogenesis of neonatal necrotizing

85 enterocolitis (NEC), the most common life-threatening gastrointestinal emergency in neonatal

86 patients (69, 125, 127). Other major sources of intestinal ischemic injury result from diseases

87 that reduce systemic blood flow such as cardiopulmonary diseases and shock states (53, 151).

88 Finally, the interruption of blood flow is an important component of the process of intestinal

89 transplantation that inevitably results in a period of ischemia (106, 112) (Figure 1).

90 Decreases in blood flow compromise the oxygen supply required for normal cellular function

91 and results in both cellular damage and death. The intestinal epithelium has a particularly high

92 energy demand, largely as a result of basolateral Na+-K+ATPase used to drive secretion and

93 absorption, and is therefore particularly sensitive to reductions in blood flow. The process of

94 epithelial cell loss that results from ischemic injury is well documented in many research animal

95 species (39, 135, 137, 147). Cells closest to the intestinal lumen on the villus tips in the small

96 intestine and inter-crypt surface epithelium in the colon initially lose their attachment to the

97 basement membrane with progressive loss of cells extending toward the crypt base as the

98 duration of ischemia increases (39, 135) (Figure 3). Interestingly, the cells closest to the luminal

99 surface normally function in a state of ‘physiologic hypoxia’ due to an oxygen gradient that is

100 derived from the mucosal vascular architecture (146, 171). Although there are species

101 differences in the anatomy of the mucosal vasculature (Figure 4), a general construct of counter

102 current exchange exists whereby oxygen diffuses from the arterial blood supply that ascends
103 the mucosa toward the lumen and venous drainage flowing in the opposite direction,

104 progressively decreasing the arterial concentration of oxygen as the vessels near the luminal

105 surface (3, 9). This creates a steep oxygen gradient that makes the villus tips hypoxic (33, 159).

106 A similarly decreasing gradient of oxygen from the crypt base to the luminal surface exists within

107 the colon (171). Therefore, further decreases in oxygen concentrations, as occurs with intestinal

108 ischemia, impact the cells at the villus tips and colonic surface epithelium first. This initially

109 creates a distinct lesion identified histopathologically, called Gruenhagen’s space (16, 39)

110 (Figure 3). Originally, this space was solely attributed to the accumulation of cytoplasmic fluid

111 from ischemically damaged cells (39). More recent studies using in vitro techniques and a novel

112 in vivo human study have shown that contraction of the myofibroblasts within the lamina propria

113 also contribute to the creation of this initial defect (46, 141). In fact, the creation of the space

114 was shown to occur within 30 minutes of complete ischemia and reseal by 60 minutes if the

115 blood supply was subsequently returned (46). This demonstrates the remarkable capacity of the

116 intestine to repair rapidly in cases where vascular compromise can be reversed. However, in

117 most cases the duration of ischemia is likely to be lengthier and the tight junctions that serve to

118 anchor the cells together break down, resulting in cell loss into the intestinal lumen. Epithelial

119 cell separation from the basement membrane and complete cell loss then continue and

120 progress toward the crypt base in a time-dependent manner (39). It has been specifically

121 demonstrated in a porcine model of jejunal mesenteric vascular occlusion that by 60 minutes,

122 epithelium is lost from the upper third of the villus and by 120 minutes there is near complete

123 loss of villus epithelium (22). Any exposure of the basement membrane causes marked

124 compromise of barrier function, allowing luminal bacteria and toxins to gain access to the

125 underlying vasculature within the lamina propria (75, 111, 140). The sequelae are septicemia

126 and multiple organ dysfunction, which are responsible for the high morbidity and mortality rates

127 associated with severe intestinal ischemic injury (86, 143, 166, 183).
128 Models of Ischemia

129 Complete Vascular Occlusion

130 Complete ischemia by temporary vascular occlusion (using atraumatic vascular clamps) or

131 permanent vascular occlusion (using ligation) of the superior mesenteric artery (SMA –

132 technically the cranial mesenteric artery in animals) in rodent models is currently the most

133 commonly used method of inducing intestinal ischemic injury. The SMA is approached through

134 a midline incision and occluded for varying lengths of time (87). In most of these studies,

135 heparin is injected intravenously to prevent thrombus formation within the SMA which enables

136 the circulation to be re-established once the atraumatic vascular clamps are removed for

137 measures of reperfusion injury. However, an early study by Megison et. al. noted a highly

138 variable degree of injury and often high mortality of rats in studies in which occlusion of the SMA

139 was performed (116). That study noted that SMA occlusion alone was unreliable in creating

140 consistent and reproducible injury, whereas SMA occlusion combined with ligation of collateral

141 arcades created a greater degree of injury and a higher but consistent level of mortality in the

142 animals. These findings lent support to a similar study performed in cats in which SMA

143 occlusion reduced blood flow to the mucosa an average of 35% in the proximal duodenum, 61%

144 in the distal duodenum, 71% in the jejunum and ileum, and 63% in the proximal colon. No

145 reduction of blood flow was identified in the mid and distal colon (142). Thus, the effect of SMA

146 occlusion is highly dependent on the intestinal segment being evaluated. These findings may be

147 complicated further by the fact that different segments of the intestine may be more or less

148 sensitive to ischemic injury. For example, differences in resistance to ischemic injury have been

149 identified between the jejunum, ileum and large colon with studies indicating a progressive

150 increase in resistance to ischemic injury in aboral segments (37, 107, 147). Furthermore, a

151 porcine study of small bowel transplantation demonstrated microscopic injury in the ileum only

152 after 5 hours of ischemic injury compared to 1.5 hours in the jejunum (35). Little reperfusion
153 injury was noted in either segment. It should be noted, however, that this study only evaluated

154 injury histologically.

155 Unfortunately, many studies of ischemic injury either assess injury in a single segment of

156 intestine, many times the ileum, or do not specify which segment is being evaluated (Table 3).

157 Additionally, few studies have focused on the colon despite the fact that colonic ischemia is a

158 more common clinical entity in people than small intestinal ischemia (1, 51, 73, 122). The colon

159 does, in fact, respond differently to insult. For example, a study compared regional differences

160 in susceptibility to injury between the small intestine and colon and demonstrated that the colon

161 was more resistant to ischemia in rats (107).

162 A more recent technique used to achieve complete irreversible ischemia was established in a

163 porcine model by embolization of the SMA. The SMA was accessed either percutaneously (96)

164 or via endovascular catheterization (2, 25, 148) and a solution of either buthyl-2-cyanoacrylate

165 (96) or polyvinyl alcohol particles and gel foam (2, 25, 148) were injected intravascularly. These

166 models are particularly useful for imaging studies aimed at improving diagnosis in patients

167 suspected of having AMI. Using this approach, the abdominal contents are not exposed or

168 manipulated in creating the vascular obstruction. Since bowel manipulation alone creates

169 inflammation that otherwise would not be present in clinical cases of AMI, minimally invasive

170 approaches to the SMA may be optimal for studies modeling AMI.

171 The critical assessment of the type of ischemia model with regard to studies of AMI cannot be

172 overstated. Acute mesenteric ischemia is a complex of diseases that can be classified based on

173 whether the mechanism of obstruction is occlusive or nonocclusive. Occlusive intestinal

174 ischemia is further subdivided into acute or chronic and whether arteries or veins are affected.

175 Nonocclusive mesenteric ischemia is a disease process that is more poorly understood but

176 occurs with patent mesenteric arteries and is associated with disease processes such as
177 congestive heart failure, aortic insufficiency, renal or hepatic disease and patients following

178 cardiac surgery (172). Therefore, other approaches to create ischemic injury such as the low-

179 flow model likely better recapitulate this form of injury.

180 Low-Flow Ischemia

181 Most of the original low-flow ischemia studies were performed in cats (Table 3).This model likely

182 best recreates the type of intestinal injury that would be expected following hemorrhagic shock

183 or other events that dramatically decreases the volume of blood delivered to an intestinal

184 segment. In the low-flow ischemia model, blood flow is reduced to 20% of baseline levels (~25-

185 35 mmHg). Using this method, the abdomen is approached through a midline incision and a

186 segment of ileum is typically isolated. An arterial circuit is then established between the superior

187 mesenteric and femoral arteries and pressure cannulas are used within the vessels in order to

188 measure vascular pressure. Reduced blood pressure within the superior mesenteric artery (25-

189 30 mm Hg) is achieved using an adjustable vascular clamp. The majority of injury associated

190 with this model has been attributed to reperfusion as a result of increases in xanthine oxidase

191 and neutrophil activation, making the ischemic event more of a priming mechanism for

192 subsequent injury. This may or may not adequately represent clinical scenarios, in which injury

193 caused by ischemia may be so severe that reperfusion injury is of limited significance. Again,

194 careful selection of models depending upon the human condition being studied is critical.

195 Segmental Mesenteric Vascular Occlusion

196 An additional model of ischemia is segmental mesenteric vascular occlusion, in which the blood

197 flow within a discrete loop of intestine is interrupted by clamping the local mesenteric vascular

198 supply and cross-clamping the bowel. Within a single animal, multiple loops can be subjected to

199 varying lengths of ischemia with or without reperfusion, thereby titrating the degree of injury

200 within each loop. This model can be readily performed in large animals, such as the pig, and
201 rodents (73, 74), and has the advantage of providing multiple treatment groups and controls

202 within a single animal. However, this model of strangulating obstruction relies on complete

203 occlusion of the arterial and venous blood supply, whereas clinically the venous circulation is

204 commonly compromised prior to the arterial blood supply due to differences in vessel wall

205 thickness and compliance (135). This results in continued arterial supply of oxygenated blood

206 for a variable period of time, until the tissue becomes so congested in the absence of venous

207 drainage that the arterial vascular supply ultimately collapses. This complex form of ischemic

208 injury includes elements of ischemia as well as excessive interstitial pressure, which together

209 likely affect the level of epithelial sloughing.

210 Perhaps the most interesting and significant application of the mesenteric vascular occlusion

211 model of intestinal ischemia has been its application in human subjects (46, 47). In these

212 studies, a normal segment of jejunum from patients undergoing Roux-en-Y or similar intestinal

213 reconstruction were isolated and subjected to 30-45 minutes of mesenteric vascular occlusion,

214 with varying lengths of reperfusion, prior to removal from the patient (46, 47, 75, 115). The

215 limitation of these studies is that severe ischemic damage as applies to most patients suffering

216 from ischemia-reperfusion injury cannot be induced in these clinical studies.

217 With regard to each of these models of ischemia, the research animal species used may

218 contribute significantly to the outcome. As previously noted, variations of the mucosal vascular

219 supply, particularly in the small intestine, exist between species (Figure 4). The pig and human

220 villus microvascular architecture are very similar, arborizing at the tip of the villus into a fountain-

221 like pattern and converging into one or two venules located beside the centrally located arteriole

222 (9, 33). In contrast, in the rat, the flattened leaf-shaped villi are supplied by a single arteriole that

223 passes unbranched from the base to the tip of the villus where it bifurcates into a capillary

224 network, a so-called “netted bag” pattern (38). The arterioles begin to converge and form two

225 venules as the vasculature approaches the base of the villus (33, 38). In the mouse, two
226 arterioles supply oxygenated blood to the villus, and then divide into a capillary network that

227 rejoins to form a single venule at the tip of the villus. The venule then travels down the center

228 toward the base of the villus (34). These differences may impact the physiological function of

229 the intestinal mucosa between species and potentially their susceptibility or response to injury.

230 Multiple in vivo and in vitro intestinal permeability studies have demonstrated significant

231 differences between man and rodents and greater correlation between humans and pigs (15,

232 45, 126). In particular, Bijlsma et. al. hypothesize that the differences in permeability between

233 rodents and humans is due to the interspecies variation in villus blood vessel architecture

234 resulting in varying countercurrent exchange efficiency that directly impacts villus epithelial cell

235 function (15).

236 Mechanisms of Reperfusion Injury

237

238 The concept of progressive injury that occurs following the restoration of normal vascular

239 circulation to an ischemically compromised region seems counter-intuitive. After all, re-

240 establishing blood flow is ultimately critical to rescue and maintain cell function. However,

241 ischemia creates an environment primed for the production of injurious metabolites, the influx of

242 inflammatory cells and epithelial cell apoptosis and necrosis. Early studies of reperfusion in cats

243 and rodents attributed the exacerbation of injury during this period to the production of reactive

244 oxygen metabolites that directly contribute to injury as well generation of neutrophils

245 chemoattractants (26, 48, 65, 66, 68, 70, 77, 78, 82, 98, 99, 101, 128-130, 136-138, 153, 164).

246 During ischemic conditions, as tissues continue to utilize adenosine triphosphate as a source of

247 energy, the metabolic byproduct, hypoxanthine, accumulates. In addition, in this oxygen

248 deprived environment, xanthine dehydrogenase which would normally function to metabolize

249 hypoxanthine is converted to xanthine oxidase by locally produced tissue proteases (130).

250 When tissue perfusion is re-established, xanthine oxidase utilizes the now freely available and
251 abundant oxygen as an electron acceptor as it metabolizes hypoxanthine, producing superoxide

252 (66-68, 70) (Figure 2). The direct injury induced by oxygen free radicals combined with their role

253 in the activation and chemoattraction of neutrophils creates the severe tissue injury attributed to

254 reperfusion.

255 Neutrophils inflict tissue injury by a multitude of proposed mechanisms that include occlusion

256 and increased permeability of the microvasculature (44, 83), release of reactive oxygen

257 metabolites (48, 130, 165), cytotoxic enzyme release (186) and mechanical injury induced by

258 migration (58). The vast majority of the studies used to determine these findings used a feline

259 low-flow ischemia model. Upon activation, neutrophils release the contents of their granules that

260 include potent proteases such as elastase, myeloperoxidase, protease-3 and

261 metalloproteinases as well as reactive oxygen species including H2O2 and hypochlorous acid

262 (108, 153, 186). These compounds are bactericidal and aid in neutrophil migration as

263 connective tissues are lysed, but concomitantly cause extensive collateral damage to

264 surrounding matrix proteins and nearby cells. Additional injury is created by the mechanical

265 damage induced by their migration across the epithelial monolayer which has been shown to

266 increase paracellular permeability (58). The detrimental role of neutrophil accumulation on

267 epithelial barrier function post ischemia was supported by findings in a porcine model of

268 complete mesenteric vascular occlusion (58) in which pre-treatment of animals with anti

269 CD11/CD18 monoclonal antibody, targeting neutrophil adhesion, significantly reduced

270 neutrophil infiltration and improved measures of epithelial barrier function (58).

271 Model Dependent Reperfusion Injury

272 The significance of reperfusion to intestinal injury following an ischemic event has been and

273 continues to be controversial. This is partly due to the fact that the degree of reperfusion injury

274 appears to depend on the type of ischemia (complete versus incomplete vascular occlusion)
275 and duration of ischemic injury as well as the research animal species and segment of affected

276 intestine.

277 Complete Vascular Occlusion

278 Currently, the complete vascular occlusion method for induction of ischemic injury in rodents is

279 the most commonly used approach for the study of ischemia-reperfusion injury (Table 5 and 6).

280 Rodents are advantageous to use because of their relatively low cost, ease of maintenance,

281 and rapid reproduction rate. In recent ischemia-reperfusion research, mouse models have been

282 used to determine genes associated with oxidative stress (12) as well as to provide evidence of

283 increased epithelial proliferation and barrier function in response to endogenous factors such as

284 keratinocyte growth factor administration (30). Furthermore, mice are highly amenable to

285 genetic manipulation. Attenuation of reperfusion injury in mice genetically modified to

286 overexpress superoxide dismutase has provided further proof of the deleterious role of reactive

287 oxygen metabolites in reperfusion (48). Additionally, other transgenic mouse models have been

288 used to demonstrate the protective role of mu opioid receptor signaling (61), and the detrimental

289 effects of cytokines such as IL-17A (104) in ischemia-reperfusion injury. A large number of

290 studies have also utilized rat models. Aside from transgenic murine models, the role of a host of

291 factors has been successfully investigated in rodents, highlighting the beneficial effects of

292 epidermal growth factor (11, 59), heparin-binding epidermal growth factor (36, 50, 114), anti-

293 apoptotic signaling (86) and xanthine oxidase blockade (157) to prevent injury or improve

294 healing. Other studies using rats have shown that atenolol (31), a beta-blocker and L-arginine

295 (62), a substrate of nitric oxide biosynthesis both attenuate enteric nervous system dysfunction

296 that occurs as a result of ischemia-reperfusion injury. However, multiple studies have

297 demonstrated that the degree of injury attributed to reperfusion following mesenteric vascular

298 occlusion is variable. The degree of reperfusion injury appears to depend on the duration of

299 preceding ischemia and the segment of intestine (107, 135). Evidence of reperfusion injury has
300 been shown to occur following intervals between 30 to 60 minutes of ischemia but not when the

301 duration of ischemia is shorter or longer (107, 135). Additionally, the colon appears to be more

302 resistant to reperfusion injury compared to the small intestine and susceptibility to injury along

303 the length of the small intestine is variable with the ileum appearing to be more resistant to

304 reperfusion injury than the jejunum. The reason for these differences remains poorly understood

305 (35, 37, 107), although the lack of oxidant-producing enzymes in the colon may be one reason

306 that injury differs from the small intestine (76, 120, 121).

307 Low-Flow Ischemia

308 As noted above, in models utilizing the low-flow method of ischemia, the majority of tissue

309 damage occurs during reperfusion. In vivo feline studies have shown a direct correlation

310 between elevated xanthine oxidase levels and tissue damage as well as microvascular injury

311 (65, 68, 164, 184). However, important differences exist between species that influence their

312 susceptibility to reperfusion injury. The robust reperfusion injury identified in cats and rodents as

313 a result of mucosal xanthine oxidase-induced oxidant release is not found in other species (22).

314 In fact, humans and pigs lack mucosal xanthine oxidase at birth, and levels remain low even

315 into adulthood making the clinical relevance of the feline and rodent studies questionable (14).

316 Furthermore, no definitive clinical utility has been identified for xanthine oxidase inhibitors in

317 patients with intestinal ischemia-reperfusion injury (105, 132). Perhaps with the development of

318 the novel human model of ischemia-reperfusion injury (47) the controversy regarding the

319 contribution of xanthine oxidase to reperfusion injury will be resolved.

320 Another important species difference is the role of neutrophils in the induction and perpetuation

321 of reperfusion injury. The resident mucosal population of neutrophils, as opposed to neutrophils

322 recruited from circulation during onset of injury, appears to play a significant role in reperfusion

323 injury in rodents and cats (48, 101, 130, 153). In a low-flow cat model, acute versus chronic pre-
324 treatment with a CD18-specific monoclonal antibody enabled the investigators to assess the

325 role of resident mucosal neutrophils, with chronic administration of the antibody depleting

326 mucosal neutrophil reserves. These studies indicated that it was the resident neutrophils, rather

327 than those infiltrating from the microvasculature, that were responsible for the majority of the

328 reperfusion injury documented (101, 130, 153). However, humans have a relatively small

329 population of resident neutrophils (22), a finding that was also found to be the case in horses.

330 Interestingly, equine ischemia studies have shown very limited evidence of reperfusion injury

331 (21, 120). Together, differences in oxidant enzyme expression and neutrophil populations likely

332 affect the reliability of basic studies to translate findings to the treatment of humans with

333 ischemia-reperfusion injury.

334 Segmental Mesenteric Vascular Occlusion

335 Most of the segmental mesenteric vascular occlusion studies have been performed in pigs

336 (Table 7) although other species have been used (Tables 3, 4 and 6). This is likely because the

337 larger size of the pig facilitates temporary occlusion of smaller vessels, such as those found

338 within the mesentery, without compromising the integrity of the vessels, thereby allowing

339 reperfusion. There are other advantages to using the porcine model for the study of reperfusion

340 injury. Pigs share a similar pattern of xanthine oxidase expression as humans (22), whereas in

341 rodents, enzyme expression is 4-5 fold higher (14, 22). Additionally, pigs like humans have a

342 relatively small population of resident neutrophils unlike rodents and cats (22).

343 The larger size of pigs is also advantageous for models requiring surgical manipulation, such as

344 Thiry-Vella loops in which an isolated cannulated segment of intestine can be subject to

345 ischemic injury (19), or where research involves tissue transplantation (182) or testing advanced

346 diagnostic imaging (28). Similar surgical manipulations in rodents have been fraught with

347 complications (6).


348 Although evidence supports the use of pigs to more closely recapitulate human intestine,

349 obvious morphological differences exist which may influence the use of pigs for the study of

350 reperfusion injury (10, 45, 92, 149). In many mammalian species, including humans, the small

351 intestinal epithelium is composed of stem cells that reside within the crypt base, and four post-

352 mitotic cell types: absorptive enterocytes, goblet cells, enteroendocrine cells, and Paneth cells.

353 However, the existence of Paneth cells in pigs remains controversial and there is currently no

354 definitive evidence to prove or disprove the existence of a similarly functioning cell type (63,

355 123, 124). Paneth cells are unique in that they reside adjacent to the intestinal stem cells within

356 the small intestinal crypt base, and are thought to migrate toward the crypt base following

357 complete maturation as compared to other maturing cell lineages that migrate toward the lumen.

358 Paneth cells are thought to play an integral role in modulating stem cell self-renewal or

359 differentiation especially during times of insult (40, 57, 152), making the study of regenerative

360 events following ischemic injury potentially more complex in porcine models. However, there is

361 recent evidence that Paneth cells may actually contribute to the injury induced by ischemia-

362 reperfusion injury (72, 93, 104). Work in rodent models attribute injury within the gut, systemic

363 inflammation and remote organ injury following intestinal ischemia-reperfusion injury to Paneth

364 cell degranulation. In fact, it appears that Paneth cell depletion may attenuate ischemia-

365 reperfusion injury (104).

366 Knowledge regarding the advantages and disadvantages to each model is critical to appropriate

367 experimental design and data interpretation.

368 Future Directions

369 New Insight into Mechanisms of Cell Death

370 Recent insight into the process of cell death by apoptosis and necrosis in both rodent and

371 human models of intestinal ischemia-reperfusion injury have begun to shed light into the role of
372 these pathways in the process of injury and subsequent repair (32, 46, 115, 156, 181). An

373 important component to the combined injury attributed to ischemia and reperfusion is the direct

374 impact on the intestinal epithelial cells. Controlled cell death is critical to the maintenance of

375 normal barrier function and serves to maintain epithelial continuity along the luminal surface

376 while expelling dying cells. Under homeostatic conditions, cells mature as they gradually

377 migrate along the crypt-villus axis. As they near the luminal surface a normal process of

378 desquamation occurs that results in epithelial cell shedding. Briefly, this process is thought to be

379 driven by an interaction between the extracellular matrix and integrins and cadherins, the

380 transmembrane proteins that anchor cells to the basement membrane, and which signal cell

381 survival through pathways including focal adhesion kinase (p125fak), phosphatidylinositol 3’-

382 kinase (PI-K)/Akt and mitogen-activated protein kinases (ERK, JNK, p38 MAPK) (49, 54, 55, 84,

383 174). Changes in these surface proteins and signaling pathways occur as cells approach the

384 villus tip that signal a normal process of cell shedding into the intestinal lumen (29). During

385 ischemia and reperfusion, cell injury results in death by both apoptotic and necrotic processes

386 (75, 87, 109). Sustained tissue injury appears to be associated with inflammation that is

387 perpetuated by the presence of excessive apoptotic and necrotic cells mediated by the

388 complement system (79, 162, 178, 185). However, the exact pathophysiologic mechanisms that

389 occur are complex and incompletely understood (181). Nonetheless, inhibition of apoptosis as

390 well as the facilitated clearance of apoptotic cells appears to reduce bacterial translocation and

391 promote repair my minimizing prolonged inflammation (46, 86, 115, 181). Based on research

392 dedicated to understanding ischemia-reperfusion injury, it seems that the attenuation or

393 modulation of the inflammatory response would be critical to future therapeutic approaches

394 aimed at reducing tissue destruction. This is discussed in a few recent reviews; one that

395 addresses the contribution of oxidative stress to the pathogenesis of gastrointestinal disease

396 (13) and another that summarizes the therapeutic potential for neutrophil elastase inhibitors to

397 decrease inflammatory tissue injury (81). These reviews note that despite the large amount of
398 preclinical research dedicated to these particular approaches to modulation of inflammation, the

399 challenges to translate findings to clinical application remain. However, compared to the field of

400 gastrointestinal research, more current and novel therapeutic strategies appear to be available

401 for ischemic cardiovascular disease (81, 139). Nevertheless, little progress has been made in

402 improving clinical outcome in either field. Therefore a better understanding of reparative

403 processes may provide an alternative means to improve care.

404 Advances in Understanding Epithelial Renewal

405 Recent advances in the field of intestinal stem cell biology and regenerative medicine have

406 improved our understanding of epithelial renewal (145). The recent discovery of biomarkers to

407 clearly distinguish intestinal stem cell populations has contributed to the rapid advancement of

408 the field (8, 64, 167, 175, 176). Interestingly, rodent models of intestinal ischemia have shown

409 architectural preservation of the intestinal epithelial stem cells (56, 88, 134, 173). Additionally, the

410 ability of the intestinal epithelial stem cell compartment to expand after intestinal resection, radiation

411 injury, and doxorubicin treatment has been demonstrated in rodent models (42, 43, 85, 177).

412 Together this makes the intestinal epithelial stem cells a promising therapeutic target to hasten

413 mucosal epithelial regeneration following ischemia-reperfusion injury. It is our hope that this new

414 knowledge and advanced technology will facilitate targeted therapies to improve treatment and

415 outcome of this devastating disease.

416 Summary

417 Ischemia and reperfusion contribute to intestinal injury in a multitude of clinical

418 gastroenterological conditions. Despite this, advances in the field have remained modest. This

419 review aimed to shed light on the most commonly used animal models for the study of ischemia

420 and reperfusion to aid in interpretation of study results and perhaps influence model selection in

421 the future. For example, if investigation of reperfusion injury were of particular interest, a feline
422 model of ischemia would be helpful; whereas strangulating intestinal ischemia would be more

423 appropriately studied using a porcine mesenteric vascular occlusion model. Appropriate

424 modeling of disease states or model selection are arguably components of the failure to

425 translate bench top discoveries to clinical application.

426 Figure Legend

427 Figure 1. Clinical conditions that cause ischemic intestinal injury

428 Figure 2. Mechanism of ischemia-reperfusion injury

429 Figure 3. Epithelial loss associated with ischemic intestinal injury in porcine jejunum. As the

430 duration of ischemia increases there is progressive loss of epithelium that begins at the villus tip

431 and continues toward the crypt base. Asterisk indicates Gruenhagen’s space. Scale Bar 50 µm.

432 Figure 4. Species differences in villus microvascular architecture

433

434

435

436

437

438

439

440

441
Table 1. Comparison of Models of Ischemia
Complete Vascular Occlusion Low-Flow Segmental
SMA Ligation SMA Embolization Vascular
Occlusion
Advantages • Models • Models occlusive • Models • Models
occlusive causes of ischemia nonocclusiv occlusive
causes of • Intestinal tract not e causes of causes of
ischemia exposed or ischemia ischemia
• Ease of surgical manipulated (2, 25, • Improved • Creation of
model creation 96, 148) model of multiple loops
• Ideal for diagnostic reperfusion of varying
imaging studies injury duration of
ischemia within
a single animal
• Applied in
human
subjects (46,
47, 73-75, 115)
Disadvantages • Poor model of • Poor model of • Increase • Both arterial
chronic ischemia chronic ischemia difficulty of and venous
• Only arterial • Only arterial supply surgery supply ligated;
supply occluded occluded • Minimal most clinical
• Unreliable to • Requires advanced ischemic cases of
create surgical approach injury strangulating
consistent injury • Likely requires ischemia
(116) large animal venous supply
• High mortality • Ischemia not obstructed first
rate (116) reversible
• No reduction of prohibiting study of
blood flow to reperfusion
distal colon
(142)
• Different
segments of
intestine vary in
resistance to
ischemic injury
(37, 107, 107,
147, 147)
442

443

444

Table 2. Comparison of Large and Small Animal Models of Digestive Disease


Advantages Disadvantages
Large • Select models closely approximate • High cost
Animal human disease • More difficult to manage
(Porcine, • Approximate human size (pig) • Special housing facilities
Dog, • Possess highly developed central and • Few transgenic models available
Cat) peripheral nervous system • Time consuming
• Ease of surgical manipulation • Pathophysiological differences,
specifically with regard to
reperfusion injury (cat)
• Morphological differences in
mucosal epithelial cell populations
compared to humans

Small • Low cost • Pathophysiological differences,


Animal • Ease of maintenance specifically with regard to
(Rodent) • Ease of genetic manipulation reperfusion injury
• Rapid reproduction rate • Absence of clinical signs of human
• Number of available and well disease in some models
established models • Technically difficult to create
surgery models due to size
• Mortality rate
• Genome is ~14% smaller than
humans
445

446

447

448

449

450

451

452

453

454
455 Table 3. Experimental details of in vivo ischemia-reperfusion models in cats

Type of Experimental IR Segment of Intestine Reference


Injury
SMA Complete Occlusion Ileum (95)

SMA Complete Occlusion Jejunum + Ileum (52)

Mesenteric vascular Small intestine segment not (80)

occlusion specific

Low-flow SMA occlusion Ileum (67, 70, 82, 83, 90, 97, 100,

101, 128, 129, 136-138, 153,

164, 165, 180, 186, 187)

Low-flow SMA occlusion Jejunum + Ileum (98)

Low-flow SMA occlusion Jejunum (130)

Low-flow SMA occlusion Small intestine does not (91, 99, 184)

specify segment

456 SMA, Superior Mesenteric Artery

457

458

459

460

461

462

463

464
465 Table 4. Experimental details of in vivo ischemia-reperfusion models in dogs

Type of Experimental IR Segment of Intestine Reference


Injury
SMA occlusion Ileum (5, 94, 163)

Mesenteric vascular Jejunum (27, 71)

occlusion

466

467 SMA, superior mesenteric artery

468

469 Table 5. Experimental details of in vivo ischemia-reperfusion models in mice

Type of Experimental IR Segment of Intestine Reference


Injury
SMA occlusion Jejunum (12, 181)

SMA occlusion Jejunum + Ileum (185)

SMA occlusion Colon (144)

SMA occlusion* Jejunum + Ileum (104)

SMA occlusion* Jejunum (103)

SMA occlusion* Small intestine Segment not (48, 162)

specified

SMA occlusion + collateral Ileum (61)

vessel ligation*

470

471 SMA, superior mesenteric artery; Asterisk indicates transgenic animal use.

472

473

474
475 Table 6. Experimental details of in vivo ischemia-reperfusion models in rats

Type of Experimental IR Segment of Intestine Reference


Injury
Mesenteric Vascular Proximal colon (73, 74)

Occlusion

SMA occlusion Proximal colon (107, 122, 179)

SMA occlusion Jejunum + Ileum (11, 107, 114, 157)

SMA occlusion Ileum (134, 135, 150)

SMA occlusion Jejunum (30, 31, 60, 62, 86, 88, 131)

SMA occlusion Ileum (158, 168, 169)

SMA occlusion Small intestine does not (59, 181)

specify segment

SMA + jejunal and colic Jejunum (87, 116)

artery occlusion

SMA + jejunal and colic Ileum (173)

artery occlusion

476

477 SMA, superior mesenteric artery

478

479

480

481

482

483
484 Table 7. Experimental details of in vivo ischemia-reperfusion models in pigs

Type of Experimental IR Segment of Intestine Reference


Injury
SMV Occlusion Jejunum, ileum, colon (170)

Mesenteric Vascular Ileum (4, 18-20, 22-24, 28, 58, 89,

Occlusion 96, 102, 110, 117-119, 133,

154, 160, 170)

Mesenteric Vascular Jejunum + Ileum (35)

Occlusion

SMA Embolism Jejunum + Ileum (148)

SMA Embolism Small intestine + colon (96)

SMA Embolism Small Intestine segment not (2, 25)

specified

Mesenteric Vascular Jejunum (46, 47, 75, 115)

Occlusion

Mesenteric Vascular Proximal Colon (73, 74)

Occlusion

485

486 SMV, superior mesenteric vein; SMA, superior mesenteric artery

487 Table 8. Experimental details of in vivo ischemia-reperfusion models in humans

Type of Experimental IR Segment of Intestine Reference


Injury
Mesenteric Vascular Jejunum (46, 47, 75, 115)

Occlusion

Mesenteric Vascular Proximal Colon (73, 74)

Occlusion

488
489 References

490 1. American Gastroenterological Association Medical Position Statement: guidelines on

491 intestinal ischemia. Gastroenterology 118: 5: 951-953, 2000.

492 2. Acosta S, Nilsson TK, Malina J and Malina M. L-lactate after embolization of the superior

493 mesenteric artery. J.Surg.Res. 143: 2: 320-328, 2007.

494 3. Aharinejad S, Lametschwandtner A, Franz P and Firbas W. The vascularization of the

495 digestive tract studied by scanning electron microscopy with special emphasis on the teeth,

496 esophagus, stomach, small and large intestine, pancreas, and liver. Scanning Microsc. 5: 3:

497 811-849, 1991.

498 4. Ahdieh N, Blikslager AT, Bhat BG, Coleman RA, Argenzio RA and Rhoads JM. L-glutamine

499 and transforming growth factor-alpha enhance recovery of monoacylglycerol acyltransferase

500 and diacylglycerol acyltransferase activity in porcine postischemic ileum. Pediatr.Res. 43: 2:

501 227-233, 1998.

502 5. Arakawa K, Takeyoshi I, Akao Y, Totsuka O, Matsumoto K and Morishita Y. Bradykinin B2

503 receptor antagonist FR173657 ameliorates small bowel ischemia-reperfusion injury in dogs.

504 Dig.Dis.Sci. 50: 1: 27-36, 2005.

505 6. Avansino JR, Chen DC, Woolman JD, Hoagland VD and Stelzner M. Engraftment of mucosal

506 stem cells into murine jejunum is dependent on optimal dose of cells. J.Surg.Res. 132: 1: 74-79,

507 2006.

508 7. Bainey KR and Armstrong PW. Clinical perspectives on reperfusion injury in acute myocardial

509 infarction. Am.Heart J. 167: 5: 637-645, 2014.


510 8. Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A,

511 Korving J, Begthel H, Peters PJ and Clevers H. Identification of stem cells in small intestine and

512 colon by marker gene Lgr5. Nature 449: 7165: 1003-1007, 2007.

513 9. Bellamy JE, Latshaw WK and Nielsen NO. The vascular architecture of the porcine small

514 intestine. Can.J.Comp.Med. 37: 1: 56-62, 1973.

515 10. Bendixen E, Danielsen M, Larsen K and Bendixen C. Advances in porcine genomics and

516 proteomics--a toolbox for developing the pig as a model organism for molecular biomedical

517 research. Brief Funct.Genomics 9: 3: 208-219, 2010.

518 11. Berlanga J, Prats P, Remirez D, Gonzalez R, Lopez-Saura P, Aguiar J, Ojeda M, Boyle JJ,

519 Fitzgerald AJ and Playford RJ. Prophylactic use of epidermal growth factor reduces

520 ischemia/reperfusion intestinal damage. Am.J.Pathol. 161: 2: 373-379, 2002.

521 12. Bertoletto PR, Ikejiri AT, Somaio Neto F, Chaves JC, Teruya R, Bertoletto ER, Taha MO

522 and Fagundes DJ. Oxidative stress gene expression profile in inbred mouse after

523 ischemia/reperfusion small bowel injury. Acta Cir.Bras. 27: 11: 773-782, 2012.

524 13. Bhattacharyya A, Chattopadhyay R, Mitra S and Crowe SE. Oxidative stress: an essential

525 factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol.Rev. 94: 2: 329-354,

526 2014.

527 14. Bianciardi P, Scorza R, Ghilardi G and Samaja M. Xanthine oxido-reductase activity in

528 ischemic human and rat intestine. Free Radic.Res. 38: 9: 919-925, 2004.

529 15. Bijlsma PB, Peeters RA, Groot JA, Dekker PR, Taminiau JAJM and Van Der Meer R.

530 Differential in vivo and in vitro intestinal permeability to lactulose and mannitol in animals and

531 humans: A hypothesis. Gastroenterology 108: 3: 687-696, 1995.


532 16. Blikslager AT. Life in the gut without oxygen: adaptive mechanisms and inflammatory bowel

533 disease. Gastroenterology 134: 1: 346-348, 2008.

534 17. Blikslager AT. Treatment of gastrointestinal ischemic injury. Vet.Clin.North Am.Equine Pract.

535 19: 3: 715-727, 2003.

536 18. Blikslager AT, Pell SM and Young KM. PGE2 triggers recovery of transmucosal resistance

537 via EP receptor cross talk in porcine ischemia-injured ileum. Am.J.Physiol.Gastrointest.Liver

538 Physiol. 281: 2: G375-81, 2001.

539 19. Blikslager AT, Rhoads JM, Bristol DG, Roberts MC and Argenzio RA. Glutamine and

540 transforming growth factor-alpha stimulate extracellular regulated kinases and enhance

541 recovery of villous surface area in porcine ischemic-injured intestine. Surgery 125: 2: 186-194,

542 1999.

543 20. Blikslager AT, Roberts MC and Argenzio RA. Prostaglandin-induced recovery of barrier

544 function in porcine ileum is triggered by chloride secretion. Am.J.Physiol. 276: 1 Pt 1: G28-36,

545 1999.

546 21. Blikslager AT, Roberts MC, Gerard MP and Argenzio RA. How important is intestinal

547 reperfusion injury in horses? J.Am.Vet.Med.Assoc. 211: 11: 1387-1389, 1997.

548 22. Blikslager AT, Roberts MC, Rhoads JM and Argenzio RA. Is reperfusion injury an important

549 cause of mucosal damage after porcine intestinal ischemia? Surgery 121: 5: 526-534, 1997.

550 23. Blikslager AT, Roberts MC, Rhoads JM and Argenzio RA. Prostaglandins I2 and E2 have a

551 synergistic role in rescuing epithelial barrier function in porcine ileum. J.Clin.Invest. 100: 8:

552 1928-1933, 1997.


553 24. Blikslager AT, Roberts MC, Young KM, Rhoads JM and Argenzio RA. Genistein augments

554 prostaglandin-induced recovery of barrier function in ischemia-injured porcine ileum.

555 Am.J.Physiol.Gastrointest.Liver Physiol. 278: 2: G207-16, 2000.

556 25. Block T, Isaksson HS, Acosta S, Bjorck M, Brodin D and Nilsson TK. Altered mRNA

557 expression due to acute mesenteric ischaemia in a porcine model. Eur.J.Vasc.Endovasc.Surg.

558 41: 2: 281-287, 2011.

559 26. Boros M, Takaichi S and Hatanaka K. Ischemic time-dependent microvascular changes and

560 reperfusion injury in the rat small intestine. J.Surg.Res. 59: 2: 311-320, 1995.

561 27. Brath E, Miko I, Nemeth N, Kovacs J, Peto K and Furka I. Effects of allopurinol and

562 preconditioning on apoptosis due to ischemia-reperfusion on a double jejunum-segment canine

563 model. Acta Cir.Bras. 26: 3: 186-193, 2011.

564 28. Bruhn RS, Distelmaier MS, Hellmann-Sokolis M, Naami A, Kuhl CK and Hohl C. Early

565 detection of acute mesenteric ischemia using diffusion-weighted 3.0-T magnetic resonance

566 imaging in a porcine model. Invest.Radiol. 48: 4: 231-237, 2013.

567 29. Bullen TF, Forrest S, Campbell F, Dodson AR, Hershman MJ, Pritchard DM, Turner JR,

568 Montrose MH and Watson AJ. Characterization of epithelial cell shedding from human small

569 intestine. Lab.Invest. 86: 10: 1052-1063, 2006.

570 30. Cai Y, Wang W, Liang H, Sun L, Teitelbaum DH and Yang H. Keratinocyte growth factor

571 improves epithelial structure and function in a mouse model of intestinal ischemia/reperfusion.

572 PLoS One 7: 9: e44772, 2012.


573 31. Campos VF, Miranda-Ferreira R, Taha NS, Teixeira GD, Souza WT, Carmo CE, Silva-Neto

574 LA, Gomes IT, Monteiro HP, Montero EF, Fagundes DJ, Caricati-Neto A and Taha MO. Atenolol

575 to treat intestinal ischemia and reperfusion in rats. Transplant.Proc. 44: 8: 2313-2316, 2012.

576 32. Carden DL and Granger DN. Pathophysiology of ischaemia-reperfusion injury. J.Pathol.

577 190: 3: 255-266, 2000.

578 33. Casley-Smith JR GB. Physiology of the intestinal circulation. In: , edited by editors

579 APS,D.N.Granger., Granger DN and Shepherd AP(P). New York: Raven Press, 1984, p. 9-31.

580 34. Casley-Smith JR. Endothelial fenestrae in intestinal villi: differences between the arterial and

581 venous ends of the capillaries. Microvasc.Res. 3: 1: 49-68, 1971.

582 35. Chan KL, Chan KW and Tam PK. Segmental small bowel allograft--ischemic injury and

583 regeneration. J.Pediatr.Surg. 33: 11: 1703-1706, 1998.

584 36. Chen CL, Yu X, James IO, Zhang HY, Yang J, Radulescu A, Zhou Y and Besner GE.

585 Heparin-binding EGF-like growth factor protects intestinal stem cells from injury in a rat model of

586 necrotizing enterocolitis. Lab.Invest. 92: 3: 331-344, 2012.

587 37. Chen SH, Tang YB and Chen HC. Survival of transferred ileum after ischemia time longer

588 than 1 hour: a clinical result different from animal studies. J.Am.Coll.Surg. 217: 2: 300-305,

589 2013.

590 38. Chen YM, Zhang JS and Duan XL. Changes of microvascular architecture, ultrastructure

591 and permeability of rat jejunal villi at different ages. World J.Gastroenterol. 9: 4: 795-799, 2003.
592 39. Chiu CJ, McArdle AH, Brown R, Scott HJ and Gurd FN. Intestinal mucosal lesion in low-flow

593 states. I. A morphological, hemodynamic, and metabolic reappraisal. Arch.Surg. 101: 4: 478-

594 483, 1970.

595 40. Clevers HC and Bevins CL. Paneth cells: maestros of the small intestinal crypts.

596 Annu.Rev.Physiol. 75: 289-311, 2013.

597 41. Corcos O and Nuzzo A. Gastro-intestinal vascular emergencies. Best

598 Pract.Res.Clin.Gastroenterol. 27: 5: 709-725, 2013.

599 42. Dekaney CM, Fong JJ, Rigby RJ, Lund PK, Henning SJ and Helmrath MA. Expansion of

600 intestinal stem cells associated with long-term adaptation following ileocecal resection in mice.

601 Am.J.Physiol.Gastrointest.Liver Physiol. 293: 5: G1013-22, 2007.

602 43. Dekaney CM, Gulati AS, Garrison AP, Helmrath MA and Henning SJ. Regeneration of

603 intestinal stem/progenitor cells following doxorubicin treatment of mice.

604 Am.J.Physiol.Gastrointest.Liver Physiol. 297: 3: G461-70, 2009.

605 44. del Zoppo GJ, Schmid-Schonbein GW, Mori E, Copeland BR and Chang CM.

606 Polymorphonuclear leukocytes occlude capillaries following middle cerebral artery occlusion

607 and reperfusion in baboons. Stroke 22: 10: 1276-1283, 1991.

608 45. Delahunty T and Hollander D. A comparison of intestinal permeability between humans and

609 three common laboratory animals. Comp.Biochem.Physiol.A.Comp.Physiol. 86: 3: 565-567,

610 1987.

611 46. Derikx JP, Matthijsen RA, de Bruine AP, van Bijnen AA, Heineman E, van Dam RM, Dejong

612 CH and Buurman WA. Rapid reversal of human intestinal ischemia-reperfusion induced damage

613 by shedding of injured enterocytes and reepithelialisation. PLoS One 3: 10: e3428, 2008.
614 47. Derikx JP, Matthijsen RA, de Bruine AP, van Dam RM, Buurman WA and Dejong CH. A new

615 model to study intestinal ischemia-reperfusion damage in man. J.Surg.Res. 166: 2: 222-226,

616 2011.

617 48. Deshmukh DR, Mirochnitchenko O, Ghole VS, Agnese D, Shah PC, Reddell M, Brolin RE

618 and Inouye M. Intestinal ischemia and reperfusion injury in transgenic mice overexpressing

619 copper-zinc superoxide dismutase. Am.J.Physiol. 273: 4 Pt 1: C1130-5, 1997.

620 49. Dufour G, Demers MJ, Gagne D, Dydensborg AB, Teller IC, Bouchard V, Degongre I,

621 Beaulieu JF, Cheng JQ, Fujita N, Tsuruo T, Vallee K and Vachon PH. Human intestinal

622 epithelial cell survival and anoikis. Differentiation state-distinct regulation and roles of protein

623 kinase B/Akt isoforms. J.Biol.Chem. 279: 42: 44113-44122, 2004.

624 50. Feng J and Besner GE. Heparin-binding epidermal growth factor-like growth factor promotes

625 enterocyte migration and proliferation in neonatal rats with necrotizing enterocolitis.

626 J.Pediatr.Surg. 42: 1: 214-220, 2007.

627 51. Feuerstadt P and Brandt LJ. Colon ischemia: recent insights and advances.

628 Curr.Gastroenterol.Rep. 12: 5: 383-390, 2010.

629 52. Filez L, Stalmans W, Penninckx F and Kerremans R. Influences of ischemia and reperfusion

630 on the feline small-intestinal mucosa. J.Surg.Res. 49: 2: 157-163, 1990.

631 53. Fishman JE, Sheth SU, Levy G, Alli V, Lu Q, Xu D, Qin Y, Qin X and Deitch EA. Intraluminal

632 Nonbacterial Intestinal Components Control Gut and Lung Injury After Trauma Hemorrhagic

633 Shock. Ann.Surg. 2014.


634 54. Fouquet S, Lugo-Martinez VH, Faussat AM, Renaud F, Cardot P, Chambaz J, Pincon-

635 Raymond M and Thenet S. Early loss of E-cadherin from cell-cell contacts is involved in the

636 onset of Anoikis in enterocytes. J.Biol.Chem. 279: 41: 43061-43069, 2004.

637 55. Frisch SM and Francis H. Disruption of epithelial cell-matrix interactions induces apoptosis.

638 J.Cell Biol. 124: 4: 619-626, 1994.

639 56. Fuller MK, Faulk DM, Sundaram N, Mahe MM, Stout KM, von Furstenberg RJ, Smith BJ,

640 McNaughton KK, Shroyer NF, Helmrath MA and Henning SJ. Intestinal stem cells remain viable

641 after prolonged tissue storage. Cell Tissue Res. 2013.

642 57. Garabedian EM, Roberts LJ, McNevin MS and Gordon JI. Examining the role of Paneth

643 cells in the small intestine by lineage ablation in transgenic mice. J.Biol.Chem. 272: 38: 23729-

644 23740, 1997.

645 58. Gayle J, Jones SL, Argenzio RA and Blikslager AT. Neutrophils increase paracellular

646 permeability of restituted ischemic-injured porcine ileum. Surgery 132: 3: 461-470, 2002.

647 59. Geng Y, Li J, Wang F, Li Q, Wang X, Sun L and Li W. Epidermal growth factor promotes

648 proliferation and improves restoration after intestinal ischemia-reperfusion injury in rats.

649 Inflammation 36: 3: 670-679, 2013.

650 60. Ghadie MM, Miranda-Ferreira R, Taha NS, Maroso AS, Moreti RJ, Andraus MP, Zempulski

651 P, Monteiro HP, Simoes MJ, Fagundes DJ, Caricati-Neto A and Taha MO. Study of heparin in

652 intestinal ischemia and reperfusion in rats: morphologic and functional evaluation.

653 Transplant.Proc. 44: 8: 2300-2303, 2012.


654 61. Goldsmith JR, Perez-Chanona E, Yadav PN, Whistler J, Roth B and Jobin C. Intestinal

655 epithelial cell-derived mu-opioid signaling protects against ischemia reperfusion injury through

656 PI3K signaling. Am.J.Pathol. 182: 3: 776-785, 2013.

657 62. Gomez JS, Miranda-Ferreira R, Taha NS, Rodrigues LW, Bento DR, Fernandes DT,

658 Chihara RT, Simoes MJ, Oliveira-Junior IS, Monteiro HP, Fagundes DJ, Caricati-Neto A and

659 Taha MO. Study of L-arginine in intestinal lesions caused by ischemia-reperfusion in rats.

660 Transplant.Proc. 44: 8: 2309-2312, 2012.

661 63. Gonzalez LM, Williamson I, Piedrahita JA, Blikslager AT and Magness ST. Cell lineage

662 identification and stem cell culture in a porcine model for the study of intestinal epithelial

663 regeneration. PLoS One 8: 6: e66465, 2013.

664 64. Gracz AD, Fuller MK, Wang F, Li L, Stelzner M, Dunn JC, Martin MG and Magness ST.

665 CD24 and CD44 Mark Human Intestinal Epithelial Cell Populations with Characteristics of Active

666 and Facultative Stem Cells. Stem Cells 2013.

667 65. Granger DN, Benoit JN, Suzuki M and Grisham MB. Leukocyte adherence to venular

668 endothelium during ischemia-reperfusion. Am.J.Physiol. 257: 5 Pt 1: G683-8, 1989.

669 66. Granger DN, Hollwarth ME and Parks DA. Ischemia-reperfusion injury: role of oxygen-

670 derived free radicals. Acta Physiol.Scand.Suppl. 548: 47-63, 1986.

671 67. Granger DN, McCord JM, Parks DA and Hollwarth ME. Xanthine oxidase inhibitors

672 attenuate ischemia-induced vascular permeability changes in the cat intestine. Gastroenterology

673 90: 1: 80-84, 1986.

674 68. Granger DN, Rutili G and McCord JM. Superoxide radicals in feline intestinal ischemia.

675 Gastroenterology 81: 1: 22-29, 1981.


676 69. Gregory KE, Deforge CE, Natale KM, Phillips M and Van Marter LJ. Necrotizing enterocolitis

677 in the premature infant: neonatal nursing assessment, disease pathogenesis, and clinical

678 presentation. Adv.Neonatal Care. 11: 3: 155-64; quiz 165-6, 2011.

679 70. Grisham MB, Hernandez LA and Granger DN. Xanthine oxidase and neutrophil infiltration in

680 intestinal ischemia. Am.J.Physiol. 251: 4 Pt 1: G567-74, 1986.

681 71. Grogaard B, Parks DA, Granger DN, McCord JM and Forsberg JO. Effects of ischemia and

682 oxygen radicals on mucosal albumin clearance in intestine. Am.J.Physiol. 242: 5: G448-54,

683 1982.

684 72. Grootjans J, Hodin CM, de Haan JJ, Derikx JP, Rouschop KM, Verheyen FK, van Dam RM,

685 Dejong CH, Buurman WA and Lenaerts K. Level of activation of the unfolded protein response

686 correlates with Paneth cell apoptosis in human small intestine exposed to ischemia/reperfusion.

687 Gastroenterology 140: 2: 529-539.e3, 2011.

688 73. Grootjans J, Hundscheid IH and Buurman WA. Goblet cell compound exocytosis in the

689 defense against bacterial invasion in the colon exposed to ischemia-reperfusion. Gut Microbes

690 4: 3: 232-235, 2013.

691 74. Grootjans J, Hundscheid IH, Lenaerts K, Boonen B, Renes IB, Verheyen FK, Dejong CH,

692 von Meyenfeldt MF, Beets GL and Buurman WA. Ischaemia-induced mucus barrier loss and

693 bacterial penetration are rapidly counteracted by increased goblet cell secretory activity in

694 human and rat colon. Gut 62: 2: 250-258, 2013.

695 75. Grootjans J, Lenaerts K, Derikx JP, Matthijsen RA, de Bruine AP, van Bijnen AA, van Dam

696 RM, Dejong CH and Buurman WA. Human intestinal ischemia-reperfusion-induced inflammation
697 characterized: experiences from a new translational model. Am.J.Pathol. 176: 5: 2283-2291,

698 2010.

699 76. Grosche A, Morton AJ, Graham AS, Sanchez LC, Blikslager AT, Polyak MM and Freeman

700 DE. Ultrastructural changes in the equine colonic mucosa after ischaemia and reperfusion.

701 Equine Vet.J.Suppl. (39):8-15. doi: 39: 8-15, 2011.

702 77. Grum CM, Gross TJ, Mody CH and Sitrin RG. Expression of xanthine oxidase activity by

703 murine leukocytes. J.Lab.Clin.Med. 116: 2: 211-218, 1990.

704 78. Haglind E, Haglund U, Lundgren O and Stenberg B. Mucosal lesions of the small intestine

705 after intestinal vascular obstruction in the rat. Acta Chir.Scand. 151: 2: 147-150, 1985.

706 79. Hart ML, Ceonzo KA, Shaffer LA, Takahashi K, Rother RP, Reenstra WR, Buras JA and

707 Stahl GL. Gastrointestinal ischemia-reperfusion injury is lectin complement pathway dependent

708 without involving C1q. J.Immunol. 174: 10: 6373-6380, 2005.

709 80. Henninger DD, Granger DN and Aw TY. Enterocyte respiration rates in feline small intestine

710 exposed to graded ischemia. Am.J.Physiol. 268: 1 Pt 1: G116-20, 1995.

711 81. Hernandez LA, Grisham MB and Granger DN. A role for iron in oxidant-mediated ischemic

712 injury to intestinal microvasculature. Am.J.Physiol. 253: 1 Pt 1: G49-53, 1987.

713 82. Hernandez LA, Grisham MB, Twohig B, Arfors KE, Harlan JM and Granger DN. Role of

714 neutrophils in ischemia-reperfusion-induced microvascular injury. Am.J.Physiol. 253: 3 Pt 2:

715 H699-703, 1987.


716 83. Hofmann C, Obermeier F, Artinger M, Hausmann M, Falk W, Schoelmerich J, Rogler G and

717 Grossmann J. Cell-cell contacts prevent anoikis in primary human colonic epithelial cells.

718 Gastroenterology 132: 2: 587-600, 2007.

719 84. Hua G, Thin TH, Feldman R, Haimovitz-Friedman A, Clevers H, Fuks Z and Kolesnick R.

720 Crypt base columnar stem cells in small intestines of mice are radioresistant. Gastroenterology

721 143: 5: 1266-1276, 2012.

722 85. Huang CY, Hsiao JK, Lu YZ, Lee TC and Yu LC. Anti-apoptotic PI3K/Akt signaling by

723 sodium/glucose transporter 1 reduces epithelial barrier damage and bacterial translocation in

724 intestinal ischemia. Lab.Invest. 91: 2: 294-309, 2011.

725 86. Ikeda H, Suzuki Y, Suzuki M, Koike M, Tamura J, Tong J, Nomura M and Itoh G. Apoptosis

726 is a major mode of cell death caused by ischaemia and ischaemia/reperfusion injury to the rat

727 intestinal epithelium. Gut 42: 4: 530-537, 1998.

728 87. Itoh H, Yagi M, Hasebe K, Fushida S, Tani T, Hashimoto T, Shimizu K and Miwa K.

729 Regeneration of small intestinal mucosa after acute ischemia-reperfusion injury. Dig.Dis.Sci. 47:

730 12: 2704-2710, 2002.

731 88. Jacobi SK, Moeser AJ, Corl BA, Harrell RJ, Blikslager AT and Odle J. Dietary long-chain

732 PUFA enhance acute repair of ischemia-injured intestine of suckling pigs. J.Nutr. 142: 7: 1266-

733 1271, 2012.

734 89. Kanwar S and Kubes P. Ischemia/reperfusion-induced granulocyte influx is a multistep

735 process mediated by mast cells. Microcirculation 1: 3: 175-182, 1994.


736 90. Kanwar S, Tepperman BL, Payne D, Sutherland LR and Kubes P. Time course of nitric

737 oxide production and epithelial dysfunction during ischemia/reperfusion of the feline small

738 intestine. Circ.Shock 42: 3: 135-140, 1994.

739 91. Kararli TT. Comparison of the gastrointestinal anatomy, physiology, and biochemistry of

740 humans and commonly used laboratory animals. Biopharm.Drug Dispos. 16: 5: 351-380, 1995.

741 92. Kaser A, Tomczak M and Blumberg RS. "ER stress(ed out)!": Paneth cells and ischemia-

742 reperfusion injury of the small intestine. Gastroenterology 140: 2: 393-396, 2011.

743 93. Kawata K, Takeyoshi I, Iwanami K, Sunose Y, Tsutsumi H, Ohwada S, Matsumoto K and

744 Morishita Y. The effects of a selective cyclooxygenase-2 inhibitor on small bowel ischemia-

745 reperfusion injury. Hepatogastroenterology 50: 54: 1970-1974, 2003.

746 94. Kim MY, Suh CH, Kim ST, Lee JH, Kong K, Lim TH and Suh JS. Magnetic resonance

747 imaging of bowel ischemia induced by ligation of superior mesenteric artery and vein in a cat

748 model. J.Comput.Assist.Tomogr. 28: 2: 187-192, 2004.

749 95. Klein HM, Klosterhalfen B, Kinzel S, Jansen A, Seggewiss C, Weghaus P, Kamp M, Tons C

750 and Gunther RW. CT and MRI of experimentally induced mesenteric ischemia in a porcine

751 model. J.Comput.Assist.Tomogr. 20: 2: 254-261, 1996.

752 96. Kubes P. Ischemia-reperfusion in feline small intestine: a role for nitric oxide. Am.J.Physiol.

753 264: 1 Pt 1: G143-9, 1993.

754 97. Kubes P, Hunter J and Granger DN. Ischemia/reperfusion-induced feline intestinal

755 dysfunction: importance of granulocyte recruitment. Gastroenterology 103: 3: 807-812, 1992.


756 98. Kubes P, Hunter J and Granger DN. Effects of cyclosporin A and FK506 on

757 ischemia/reperfusion-induced neutrophil infiltration in the cat. Dig.Dis.Sci. 36: 10: 1469-1472,

758 1991.

759 99. Kubes P, Ibbotson G, Russell J, Wallace JL and Granger DN. Role of platelet-activating

760 factor in ischemia/reperfusion-induced leukocyte adherence. Am.J.Physiol. 259: 2 Pt 1: G300-5,

761 1990.

762 100. Kurtel H, Tso P and Granger DN. Granulocyte accumulation in postischemic intestine: role

763 of leukocyte adhesion glycoprotein CD11/CD18. Am.J.Physiol. 262: 5 Pt 1: G878-82, 1992.

764 101. Lauronen J, Pakarinen MP, Pirinen P, Kuusanmaki P, Raivio P, Savilahti E, Paavonen T

765 and Halttunen J. Effects of extrinsic denervation with or without ischemia-reperfusion injury on

766 constitutional mucosal characteristics in porcine jejunoileum. Dig.Dis.Sci. 46: 3: 476-485, 2001.

767 102. Lee H, Ko EH, Lai M, Wei N, Balroop J, Kashem Z and Zhang M. Delineating the

768 relationships among the formation of reactive oxygen species, cell membrane instability and

769 innate autoimmunity in intestinal reperfusion injury. Mol.Immunol. 58: 2: 151-159, 2014.

770 103. Lee HT, Kim M, Kim JY, Brown KM, Ham A, D'Agati VD and Mori-Akiyama Y. Critical role

771 of interleukin-17A in murine intestinal ischemia-reperfusion injury.

772 Am.J.Physiol.Gastrointest.Liver Physiol. 304: 1: G12-25, 2013.

773 104. Lehr HA, Menger MD and Granger DN. Ischemia-reperfusion injury: enthusiasm in

774 laboratory research but dilemma in clinical trials? Circulation 90: 3: 1580, 1994.

775 105. Lenaerts K, Ceulemans LJ, Hundscheid IH, Grootjans J, Dejong CH and Olde Damink SW.

776 New insights in intestinal ischemia-reperfusion injury: implications for intestinal transplantation.

777 Curr.Opin.Organ.Transplant. 2013.


778 106. Leung FW, Su KC, Passaro E,Jr and Guth PH. Regional differences in gut blood flow and

779 mucosal damage in response to ischemia and reperfusion. Am.J.Physiol. 263: 3 Pt 1: G301-5,

780 1992.

781 107. Linfert D, Chowdhry T and Rabb H. Lymphocytes and ischemia-reperfusion injury.

782 Transplant.Rev.(Orlando) 23: 1: 1-10, 2009.

783 108. Linkermann A, Hackl MJ, Kunzendorf U, Walczak H, Krautwald S and Jevnikar AM.

784 Necroptosis in immunity and ischemia-reperfusion injury. Am.J.Transplant. 13: 11: 2797-2804,

785 2013.

786 109. Little D, Dean RA, Young KM, McKane SA, Martin LD, Jones SL and Blikslager AT. PI3K

787 signaling is required for prostaglandin-induced mucosal recovery in ischemia-injured porcine

788 ileum. Am.J.Physiol.Gastrointest.Liver Physiol. 284: 1: G46-56, 2003.

789 110. Louis K, Netea MG, Carrer DP, Kotsaki A, Mylona V, Pistiki A, Savva A, Roditis K, Alexis

790 A, Van der Meer JW and Giamarellos-Bourboulis EJ. Bacterial translocation in an experimental

791 model of multiple organ dysfunctions. J.Surg.Res. 183: 2: 686-694, 2013.

792 111. Mallick IH, Yang W, Winslet MC and Seifalian AM. Ischemia-reperfusion injury of the

793 intestine and protective strategies against injury. Dig.Dis.Sci. 49: 9: 1359-1377, 2004.

794 112. Markel TA, Crisostomo PR, Lahm T, Novotny NM, Rescorla FJ, Tector J and Meldrum DR.

795 Stem cells as a potential future treatment of pediatric intestinal disorders. J.Pediatr.Surg. 43: 11:

796 1953-1963, 2008.

797 113. Martin AE, Luquette MH and Besner GE. Timing, route, and dose of administration of

798 heparin-binding epidermal growth factor-like growth factor in protection against intestinal

799 ischemia-reperfusion injury. J.Pediatr.Surg. 40: 11: 1741-1747, 2005.


800 114. Matthijsen RA, Derikx JP, Kuipers D, van Dam RM, Dejong CH and Buurman WA.

801 Enterocyte shedding and epithelial lining repair following ischemia of the human small intestine

802 attenuate inflammation. PLoS One 4: 9: e7045, 2009.

803 115. Megison SM, Horton JW, Chao H and Walker PB. A new model for intestinal ischemia in

804 the rat. J.Surg.Res. 49: 2: 168-173, 1990.

805 116. Moeser AJ, Haskell MM, Shifflett DE, Little D, Schultz BD and Blikslager AT. ClC-2 chloride

806 secretion mediates prostaglandin-induced recovery of barrier function in ischemia-injured

807 porcine ileum. Gastroenterology 127: 3: 802-815, 2004.

808 117. Moeser AJ, Nighot PK, Engelke KJ, Ueno R and Blikslager AT. Recovery of mucosal

809 barrier function in ischemic porcine ileum and colon is stimulated by a novel agonist of the ClC-2

810 chloride channel, lubiprostone. Am.J.Physiol.Gastrointest.Liver Physiol. 292: 2: G647-56, 2007.

811 118. Moeser AJ, Nighot PK, Ryan KA, Wooten JG and Blikslager AT. Prostaglandin-mediated

812 inhibition of Na+/H+ exchanger isoform 2 stimulates recovery of barrier function in ischemia-

813 injured intestine. Am.J.Physiol.Gastrointest.Liver Physiol. 291: 5: G885-94, 2006.

814 119. Moore RM, Bertone AL, Muir WW, Stromberg PC and Beard WL. Histopathologic evidence

815 of reperfusion injury in the large colon of horses after low-flow ischemia. Am.J.Vet.Res. 55: 10:

816 1434-1443, 1994.

817 120. Moore RM, Muir WW and Granger DN. Mechanisms of gastrointestinal ischemia-

818 reperfusion injury and potential therapeutic interventions: a review and its implications in the

819 horse. J.Vet.Intern.Med. 9: 3: 115-132, 1995.

820 121. Murthy S, Hui-Qi Q, Sakai T, Depace DE and Fondacaro JD. Ischemia/reperfusion injury in

821 the rat colon. Inflammation 21: 2: 173-190, 1997.


822 122. Myer MS. Paneth cells in the pig-a controversial issue. J.S.Afr.Vet.Assoc. 53: 1: 69, 1982.

823 123. Myer MS. The presence of Paneth cells confirmed in the pig. Onderstepoort J.Vet.Res. 49:

824 2: 131-132, 1982.

825 124. Nankervis CA, Giannone PJ and Reber KM. The neonatal intestinal vasculature:

826 contributing factors to necrotizing enterocolitis. Semin.Perinatol. 32: 2: 83-91, 2008.

827 125. Nejdfors P, Ekelund M, Jeppsson B and Westrom BR. Mucosal in vitro permeability in the

828 intestinal tract of the pig, the rat, and man: species- and region-related differences.

829 Scand.J.Gastroenterol. 35: 5: 501-507, 2000.

830 126. Neu J and Walker WA. Necrotizing enterocolitis. N.Engl.J.Med. 364: 3: 255-264, 2011.

831 127. Nilsson UA, Aberg J, Aneman A and Lundgren O. Feline intestinal ischemia and

832 reperfusion: relation between radical formation and tissue damage. Eur.Surg.Res. 25: 1: 20-29,

833 1993.

834 128. Nilsson UA, Lundgren O, Haglind E and Bylund-Fellenius AC. Radical production during in

835 vivo intestinal ischemia and reperfusion in the cat. Am.J.Physiol. 257: 3 Pt 1: G409-14, 1989.

836 129. Nilsson UA, Schoenberg MH, Aneman A, Poch B, Magadum S, Beger HG and Lundgren

837 O. Free radicals and pathogenesis during ischemia and reperfusion of the cat small intestine.

838 Gastroenterology 106: 3: 629-636, 1994.

839 130. Osborne DL, Aw TY, Cepinskas G and Kvietys PR. Development of ischemia/reperfusion

840 tolerance in the rat small intestine. An epithelium-independent event. J.Clin.Invest. 94: 5: 1910-

841 1918, 1994.


842 131. Pacher P, Nivorozhkin A and Szabo C. Therapeutic effects of xanthine oxidase inhibitors:

843 renaissance half a century after the discovery of allopurinol. Pharmacol.Rev. 58: 1: 87-114,

844 2006.

845 132. Pakarinen MP, Pirinen P, Lauronen J, Raivio P, Kuusanmaki P and Halttunen J. Effects of

846 transection and extrinsic denervation and a model of autotransplantation of the porcine

847 jejunoileum on cholesterol biodynamics. J.Pediatr.Surg. 38: 11: 1585-1590, 2003.

848 133. Park PO and Haglund U. Regeneration of small bowel mucosa after intestinal ischemia.

849 Crit.Care Med. 20: 1: 135-139, 1992.

850 134. Park PO, Haglund U, Bulkley GB and Falt K. The sequence of development of intestinal

851 tissue injury after strangulation ischemia and reperfusion. Surgery 107: 5: 574-580, 1990.

852 135. Parks DA, Bulkley GB, Granger DN, Hamilton SR and McCord JM. Ischemic injury in the

853 cat small intestine: role of superoxide radicals. Gastroenterology 82: 1: 9-15, 1982.

854 136. Parks DA and Granger DN. Contributions of ischemia and reperfusion to mucosal lesion

855 formation. Am.J.Physiol. 250: 6 Pt 1: G749-53, 1986.

856 137. Parks DA and Granger DN. Ischemia-induced vascular changes: role of xanthine oxidase

857 and hydroxyl radicals. Am.J.Physiol. 245: 2: G285-9, 1983.

858 138. Perricone AJ and Vander Heide RS. Novel therapeutic strategies for ischemic heart

859 disease. Pharmacol.Res. 89C: 36-45, 2014.

860 139. Podolsky DK. Mucosal immunity and inflammation. V. Innate mechanisms of mucosal

861 defense and repair: the best offense is a good defense. Am.J.Physiol. 277: 3 Pt 1: G495-9,

862 1999.
863 140. Powell DW, Mifflin RC, Valentich JD, Crowe SE, Saada JI and West AB. Myofibroblasts. II.

864 Intestinal subepithelial myofibroblasts. Am.J.Physiol. 277: 2 Pt 1: C183-201, 1999.

865 141. Premen AJ, Banchs V, Womack WA, Kvietys PR and Granger DN. Importance of collateral

866 circulation in the vascularly occluded feline intestine. Gastroenterology 92: 5 Pt 1: 1215-1219,

867 1987.

868 142. Puleo F, Arvanitakis M, Van Gossum A and Preiser JC. Gut failure in the ICU.

869 Semin.Respir.Crit.Care.Med. 32: 5: 626-638, 2011.

870 143. Riaz AA, Wan MX, Schafer T, Dawson P, Menger MD, Jeppsson B and Thorlacius H.

871 Allopurinol and superoxide dismutase protect against leucocyte-endothelium interactions in a

872 novel model of colonic ischaemia-reperfusion. Br.J.Surg. 89: 12: 1572-1580, 2002.

873 144. Rizk P and Barker N. Gut stem cells in tissue renewal and disease: methods, markers, and

874 myths. Wiley Interdiscip.Rev.Syst.Biol.Med. 4: 5: 475-496, 2012.

875 145. Robinson A, Keely S, Karhausen J, Gerich ME, Furuta GT and Colgan SP. Mucosal

876 protection by hypoxia-inducible factor prolyl hydroxylase inhibition. Gastroenterology 134: 1:

877 145-155, 2008.

878 146. Robinson JW, Mirkovitch V, Winistorfer B and Saegesser F. Response of the intestinal

879 mucosa to ischaemia. Gut 22: 6: 512-527, 1981.

880 147. Rosow DE, Sahani D, Strobel O, Kalva S, Mino-Kenudson M, Holalkere NS, Alsfasser G,

881 Saini S, Lee SI, Mueller PR, Fernandez-del Castillo C, Warshaw AL and Thayer SP. Imaging of

882 acute mesenteric ischemia using multidetector CT and CT angiography in a porcine model.

883 J.Gastrointest.Surg. 9: 9: 1262-74; discussion 1274-5, 2005.


884 148. Rothkotter HJ. Anatomical particularities of the porcine immune system--a physician's view.

885 Dev.Comp.Immunol. 33: 3: 267-272, 2009.

886 149. San Norberto Garcia EM, Taylor JH, Cenizo N and Vaquero C. Beneficial effects of intra-

887 arterial and intravenous prostaglandin E1 in intestinal ischaemia-reperfusion injury.

888 Interact.Cardiovasc.Thorac.Surg. 18: 4: 466-474, 2014.

889 150. Sastry P, Hardman G, Page A, Parker R, Goddard M, Large S and Jenkins DP. Mesenteric

890 ischaemia following cardiac surgery: the influence of intraoperative perfusion parameters.

891 Interact.Cardiovasc.Thorac.Surg. 2014.

892 151. Sato T, van Es JH, Snippert HJ, Stange DE, Vries RG, van den Born M, Barker N, Shroyer

893 NF, van de Wetering M and Clevers H. Paneth cells constitute the niche for Lgr5 stem cells in

894 intestinal crypts. Nature 469: 7330: 415-418, 2011.

895 152. Schoenberg MH, Poch B, Younes M, Schwarz A, Baczako K, Lundberg C, Haglund U and

896 Beger HG. Involvement of neutrophils in postischaemic damage to the small intestine. Gut 32:

897 8: 905-912, 1991.

898 153. Schwartz CA, Haage P and Hohl C. Experimental early detection of acute mesenteric

899 ischemia with functional MRI (DWI) and parallel imaging. Rofo 184: 6: 520-526, 2012.

900 154. Schwartz MZ. Novel therapies for the management of short bowel syndrome in children.

901 Pediatr.Surg.Int. 29: 10: 967-974, 2013.

902 155. Semenza GL. Series introduction: tissue ischemia: pathophysiology and therapeutics.

903 J.Clin.Invest. 106: 5: 613-614, 2000.


904 156. Shafik AN. Febuxostat improves the local and remote organ changes induced by intestinal

905 ischemia/reperfusion in rats. Dig.Dis.Sci. 58: 3: 650-659, 2013.

906 157. Shen ZY, Zhang J, Song HL and Zheng WP. Bone-marrow mesenchymal stem cells

907 reduce rat intestinal ischemia-reperfusion injury, ZO-1 downregulation and tight junction

908 disruption via a TNF-alpha-regulated mechanism. World J.Gastroenterol. 19: 23: 3583-3595,

909 2013.

910 158. Shepherd AP. Local control of intestinal oxygenation and blood flow. Annu.Rev.Physiol. 44:

911 13-27, 1982.

912 159. Shifflett DE, Bottone FG,Jr, Young KM, Moeser AJ, Jones SL and Blikslager AT.

913 Neutrophils augment recovery of porcine ischemia-injured ileal mucosa by an IL-1beta- and

914 COX-2-dependent mechanism. Am.J.Physiol.Gastrointest.Liver Physiol. 287: 1: G50-7, 2004.

915 160. Spurrier RG and Grikscheit TC. Tissue engineering the small intestine.

916 Clin.Gastroenterol.Hepatol. 11: 4: 354-358, 2013.

917 161. Stahl GL, Xu Y, Hao L, Miller M, Buras JA, Fung M and Zhao H. Role for the alternative

918 complement pathway in ischemia/reperfusion injury. Am.J.Pathol. 162: 2: 449-455, 2003.

919 162. Suto Y, Oshima K, Arakawa K, Sato H, Yamazaki H, Matsumoto K and Takeyoshi I. The

920 effect of nicorandil on small intestinal ischemia-reperfusion injury in a canine model. Dig.Dis.Sci.

921 56: 8: 2276-2282, 2011.

922 163. Suzuki M, Grisham MB and Granger DN. Leukocyte-endothelial cell adhesive interactions:

923 role of xanthine oxidase-derived oxidants. J.Leukoc.Biol. 50: 5: 488-494, 1991.


924 164. Suzuki M, Inauen W, Kvietys PR, Grisham MB, Meininger C, Schelling ME, Granger HJ

925 and Granger DN. Superoxide mediates reperfusion-induced leukocyte-endothelial cell

926 interactions. Am.J.Physiol. 257: 5 Pt 2: H1740-5, 1989.

927 165. Swank GM and Deitch EA. Role of the gut in multiple organ failure: bacterial translocation

928 and permeability changes. World J.Surg. 20: 4: 411-417, 1996.

929 166. Takeda N, Jain R, LeBoeuf MR, Wang Q, Lu MM and Epstein JA. Interconversion between

930 intestinal stem cell populations in distinct niches. Science 334: 6061: 1420-1424, 2011.

931 167. Takizawa Y, Kishimoto H, Kitazato T, Tomita M and Hayashi M. Changes in protein and

932 mRNA expression levels of claudin family after mucosal lesion by intestinal

933 ischemia/reperfusion. Int.J.Pharm. 426: 1-2: 82-89, 2012.

934 168. Takizawa Y, Kishimoto H, Tomita M and Hayashi M. Changes in the expression levels of

935 tight junction components during reconstruction of tight junction from mucosal lesion by

936 intestinal ischemia/reperfusion. Eur.J.Drug Metab.Pharmacokinet. 2013.

937 169. Tang ZH, Qiang JW, Feng XY, Li RK, Sun RX and Ye XG. Acute mesenteric ischemia

938 induced by ligation of porcine superior mesenteric vein: multidetector CT evaluations.

939 Acad.Radiol. 17: 9: 1146-1152, 2010.

940 170. Taylor CT and Colgan SP. Hypoxia and gastrointestinal disease. J.Mol.Med.(Berl) 85: 12:

941 1295-1300, 2007.

942 171. Trompeter M, Brazda T, Remy CT, Vestring T and Reimer P. Non-occlusive mesenteric

943 ischemia: etiology, diagnosis, and interventional therapy. Eur.Radiol. 12: 5: 1179-1187, 2002.
944 172. Udassin R, Vromen A and Haskel Y. The time sequence of injury and recovery following

945 transient reversible intestinal ischemia. J.Surg.Res. 56: 3: 221-225, 1994.

946 173. Vachon PH, Harnois C, Grenier A, Dufour G, Bouchard V, Han J, Landry J, Beaulieu JF,

947 Vezina A, Dydensborg AB, Gauthier R, Cote A, Drolet JF and Lareau F. Differentiation state-

948 selective roles of p38 isoforms in human intestinal epithelial cell anoikis. Gastroenterology 123:

949 6: 1980-1991, 2002.

950 174. van der Flier LG, Haegebarth A, Stange DE, van de Wetering M and Clevers H. OLFM4 is

951 a robust marker for stem cells in human intestine and marks a subset of colorectal cancer cells.

952 Gastroenterology 137: 1: 15-17, 2009.

953 175. van der Flier LG, van Gijn ME, Hatzis P, Kujala P, Haegebarth A, Stange DE, Begthel H,

954 van den Born M, Guryev V, Oving I, van Es JH, Barker N, Peters PJ, van de Wetering M and

955 Clevers H. Transcription factor achaete scute-like 2 controls intestinal stem cell fate. Cell 136: 5:

956 903-912, 2009.

957 176. Van Landeghem L, Santoro MA, Krebs AE, Mah AT, Dehmer JJ, Gracz AD, Scull BP,

958 McNaughton K, Magness ST and Lund PK. Activation of two distinct Sox9-EGFP-expressing

959 intestinal stem cell populations during crypt regeneration after irradiation.

960 Am.J.Physiol.Gastrointest.Liver Physiol. 302: 10: G1111-32, 2012.

961 177. Wada K, Montalto MC and Stahl GL. Inhibition of complement C5 reduces local and

962 remote organ injury after intestinal ischemia/reperfusion in the rat. Gastroenterology 120: 1:

963 126-133, 2001.

964 178. Wang F, Li Q, Wang C, Tang C and Li J. Dynamic alteration of the colonic microbiota in

965 intestinal ischemia-reperfusion injury. PLoS One 7: 7: e42027, 2012.


966 179. Weixiong H, Aneman A, Nilsson U and Lundgren O. Quantification of tissue damage in the

967 feline small intestine during ischaemia-reperfusion: the importance of free radicals. Acta

968 Physiol.Scand. 150: 3: 241-250, 1994.

969 180. Wu R, Dong W, Wang Z, Jacob A, Cui T and Wang P. Enhancing apoptotic cell clearance

970 mitigates bacterial translocation and promotes tissue repair after gut ischemia-reperfusion

971 injury. Int.J.Mol.Med. 30: 3: 593-598, 2012.

972 181. Yandza T, Tauc M, Saint-Paul MC, Ouaissi M, Gugenheim J and Hebuterne X. The pig as

973 a preclinical model for intestinal ischemia-reperfusion and transplantation studies. J.Surg.Res.

974 178: 2: 807-819, 2012.

975 182. Yasuhara H. Acute mesenteric ischemia: the challenge of gastroenterology. Surg.Today

976 35: 3: 185-195, 2005.

977 183. Younes M, Schoenberg MH, Jung H, Fredholm BB, Haglund U and Schildberg FW.

978 Oxidative tissue damage following regional intestinal ischemia and reperfusion in the cat.

979 Res.Exp.Med.(Berl) 184: 4: 259-264, 1984.

980 184. Zhao H, Montalto MC, Pfeiffer KJ, Hao L and Stahl GL. Murine model of gastrointestinal

981 ischemia associated with complement-dependent injury. J.Appl.Physiol.(1985) 93: 1: 338-345,

982 2002.

983 185. Zimmerman BJ and Granger DN. Reperfusion-induced leukocyte infiltration: role of

984 elastase. Am.J.Physiol. 259: 2 Pt 2: H390-4, 1990.

985 186. Zimmerman BJ, Grisham MB and Granger DN. Role of oxidants in ischemia/reperfusion-

986 induced granulocyte infiltration. Am.J.Physiol. 258: 2 Pt 1: G185-90, 1990.


987

988

You might also like