Review of experimental animal models of biliary acute pancreatitis and recent advances in basic research

Review of experimental animal models of biliary acute pancreatitis and recent advances in basic research

HPB DOI:10.1111/j.1477-2574.2011.00408.x REVIEW ARTICLE Review of experimental animal models of biliary acute pancreatitis and recent advances in b...

115KB Sizes 0 Downloads 37 Views

HPB

DOI:10.1111/j.1477-2574.2011.00408.x

REVIEW ARTICLE

Review of experimental animal models of biliary acute pancreatitis and recent advances in basic research Mei H. Wan1*, Wei Huang1,2*, Diane Latawiec2, Kun Jiang1, David M. Booth2, Victoria Elliott2, Rajarshi Mukherjee2 & Qing Xia1 1

Pancreatic Diseases Research Group, Department of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, China and 2Liverpool National Institute of Health Research (NIHR) Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, University of Liverpool, Liverpool, UK

Abstract Acute pancreatitis (AP) is a formidable disease, which, in severe forms, causes significant mortality. Biliary AP, or gallstone obstruction-associated AP, accounts for 30–50% of all clinical cases of AP. In biliary AP, pancreatic acinar cell (PAC) death (the initiating event in the disease) is believed to occur as acinar cells make contact with bile salts when bile refluxes into the pancreatic duct. Recent advances have unveiled an important receptor responsible for the major function of bile acids on acinar cells, namely, the cell surface G-protein-coupled bile acid receptor-1 (Gpbar1), located in the apical pole of the PAC. High concentrations of bile acids induce cytosolic Ca2+ overload and inhibit mitochondrial adenosine triphosphate (ATP) production, resulting in cell injury to both PACs and pancreatic ductal epithelial cells. Various bile salts are employed to induce experimental AP, most commonly sodium taurocholate. Recent characterization of taurolithocholic acid 3-sulphate on PACs has led researchers to focus on this bile salt because of its potency in causing acinar cell injury at relatively low, sub-detergent concentrations, which strongly implicates action via the receptor Gpbar1. Improved surgical techniques have enabled the infusion of bile salts into the pancreatic duct to induce experimental biliary AP in mice, which allows the use of these transgenic animals as powerful tools. This review summarizes recent findings using transgenic mice in experimental biliary AP.

Keywords biliary acute pancreatitis, bile acids, pancreatic acinar cells, pancreatic ductal cells, Gpbar1, animal model Received 28 June 2011; accepted 10 October 2011

Correspondence Qing Xia, Department of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China. Tel: + 86 28 8542 3373. Fax: + 86 28 8542 3028. E-mail: [email protected]

Introduction Biliary acute pancreatitis (BAP) refers to acute pancreatitis (AP) caused by biliary calculous diseases. It is the most common cause of AP and is associated with significant morbidity and mortality.1 Obstruction of the common biliopancreatic duct (CBPD) by gallstones blocks the efflux of pancreatic zymogens, creates elevated pressure in the pancreas and leads to bile reflux into the pancreatic duct. A number of experimental models have been designed to recreate this condition. The purely surgical models of BAP, such as *These authors contributed equally to this work.

HPB 2012, 14, 73–81

closed duodenal loop-induced pancreatitis and CBPD or pancreatic duct ligation-induced pancreatitis have been reviewed in detail previously2,3 and thus are beyond the scope of the current review. Cannulation of the pancreatic duct has enabled researchers to apply the bile components into the pancreas of experimental animals in a more controlled way. One of the most significant recent advances involves the adaptation of this technique to a mouse model, which opens the door to transgenic studies of BAP. Another significant development refers to observations of a variety of bile salts that induce pathological responses in single acinar cells in vitro, elucidating some molecular mechanisms of the detrimental action of pancreatic bile. In addition, some

© 2012 International Hepato-Pancreato-Biliary Association

HPB

74

important knowledge on the role of pancreatic duct epithelial cells (PDECs) in the pathogenesis of BAP has recently emerged. Finally, this review summarizes recent advances in basic research and highlights further research prospects in the pursuit of effective clinical interventions in BAP.

Bile acids and their targets Bile acids, in addition to their known functions in dietary fat absorption and cholesterol metabolism through nuclear receptors,4,5 also induce cellular signalling through the recently described cell surface G-protein-coupled bile acid receptor-1 (Gpbar1), found in brown adipose tissue, intestine and gallbladder in mammals.5–7 When Gpbar1 receptor is activated by bile acids, it can modulate energy homeostasis, lipid homeostasis and glucose homeostasis,8,9 and stimulate gallbladder filling.6 In hepatocytes, choleretic and cholestatic bile acids activate and inhibit store-operated Ca2+ channels, respectively, through mechanisms which involve reversible redistribution of stromal interaction molecule 1.10,11 In pancreatic acinar cells (PACs), bile acid transporters, namely, the Na+-dependent Na+ taurocholate co-transporting polypeptide (NTCP), located at the apex of the cell, and HCO3--dependent organic anion transporting polypeptide-1 (OATP1), located on the basolateral portion of the cell membrane, have recently been identified.12 The bile receptor Gpbar1 is also located in the apical region of the cell and is positioned to respond to bile in the lumen of the duct.13 The bile acid transporters and receptor constitute the molecular machinery responsible for the pathological action of refluxed or circulated bile acids on PACs.14,15 Detailed bile acid uptake and targets in PACs have been reviewed recently by Lerch and Aghdassi.14 Effects of bile acids on acinar cells In 2002, Voronina et al.16 demonstrated the effect of taurolithocholic acid 3-sulphate (TLC-S) on isolated murine PACs. TLC-S induced Ca2+ oscillations at concentrations as low as 25 mm and triggered responses in almost all cells at 200 mm. At higher concentrations of 300–500 mm, TLC-S caused longlasting Ca2+ rises comprised of initial release from intracellular Ca2+ stores and followed by the influx of extracellular Ca2+. The study found that other bile salts, taurodeoxycholate (TDC) and taurocholate (TC), also triggered local and global Ca2+ oscillations, although at much higher concentrations (1 mm and 5 mm, respectively).16 These data highlight the role of TLC-S not only as the most potent Ca2+ releaser among bile acids tested on PACs so far, but also as the most effective bile acid in inducing Ca2+-independent current, even at 10 mm. This concentration is close to the concentrations of sulphated lithocholic acid conjugates detected in serum in different pathological conditions.17 Indeed, in patients with severe extrahepatic duct obstruction, concentrations of bile acids of ~200 mm have been detected in peripheral circulation.18 Subsequently, Voronina et al. and other groups revealed that bile acids mediated intracellular Ca2+ release from both endoplas-

HPB 2012, 14, 73–81

mic reticulum and acidic intracellular Ca2+ stores through the activation of inositol trisphosphate receptors (IP3R) and ryanodine receptors,19–21 the inhibition of sarco/endoplasmic reticulum Ca2+-ATPase pumps and the activation of store-operated Ca2+ entry,12 and also reduced mitochondrial membrane potential22 and depleted both cytosolic and mitochondrial adenosine triphosphate (ATP),23 leading to cellular injury.

Effects of bile acids on pancreatic duct cells Although PACs have been extensively characterized, it is surprising that few studies have addressed issues of pancreatic duct cells under stressful conditions such as bile acid stimulation. Encouragingly, the work carried out by Venglovecz et al.24 investigated the effects of bile acids on cells of the pancreatic duct. This study has shed some light on the role of pathological agents in the function of the ductal system. Pancreatic duct epithelial cells and PACs have mutual communication and share similar responses to bile acids.25 Pancreatic duct epithelial cells play a fundamental role in secreting fluid rich in HCO3- to wash out harmful digestive enzymes secreted by PACs and in neutralizing acid chyme in the duodenum. Therefore, PDECs represent the first line of defence against bile acid reflux.25 Venglovecz et al. found that a low concentration (100 mm) of unconjugated bile acid chenodeoxycholate (CDC) stimulated HCO3- secretion via phospholipase C- and IP3-mediated Ca2+ signalling in PDECs.24 By contrast, high-concentration (1 mm) CDC inhibited HCO3- secretion, suppressed the glycolytic metabolism of PDECs and depleted mitochondrial ATP, causing mitochondrial damage.26 However, conjugated bile salt glycochenodeoxycholate (GCDC)-elevated intracellular Ca2+ signals failed to stimulate or inhibit HCO3- secretion at various concentrations and caused no morphological change in mitochondria.26 Further work in guinea pigs identified Ca2+-activated large conductance K+ channels expressed at the apical membrane of PDECs, which play a crucial role in regulating bile acid-stimulated or -inhibited HCO3- secretion.27

Induction of AP by bile salts The first experimental BAP model was established in 1856 by Bernard,28 who developed a method of retrograde injection of bile and olive oil into a canine pancreas through the ampulla of Vater. Since then, various bile salts such as sodium CDC (Na-CDC), sodium TC (Na-TC), sodium glycodeoxycholic acid (Na-GDC), TDC (Na-TDC) and TLC-S have been reported to induce AP in different species.

Model of pancreatitis induced by Na-CDC Very few studies have employed the non-conjugated bile salt, Na-CDC, to study the pathogenesis of AP or the effects of treatment regimens. This procedure sometimes requires simultaneous

© 2012 International Hepato-Pancreato-Biliary Association

HPB

ligation of the pancreatic duct and was found to cause necrotizing pancreatitis and associated lung injury in rats (5%, 2 ml/kg)29,30 and rabbits.31,32 Model of pancreatitis induced by Na-GDC An early study demonstrated that ductal infusion of 100 ml of the glycine-conjugated bile salt, Na-GDC, at concentrations of 8.5 mm, 17 mm and 34 mm, caused progressive, severe but nonlethal AP in rats.33 Na-GDC infusions of 17 mm and 34 mm caused oedematous and necrotizing pancreatitis, respectively.34–36 When 200 ng enterokinase was administrated with 34 mm Na-GDC infusate, necrotic pancreatitis with systemic disturbance and rapid lethality was produced.33 In 1992, Schmidt et al.37 established a new model in rats using the combined actions of very low concentrations of ductal infusion of Na-GDC (5 mm or 10 mm) and i.v. caerulein injection (5 mg/kg/h for 6 h). This model features a moderate onset of homogeneous moderate pancreatic injury that lasts ⱖ24 h and provides the potential for modulating severity, as well as clinical relevance. Therefore, this model is particularly suitable for use in the evaluation of new therapeutic modalities and has been used frequently in subsequent studies.38–41 Model of pancreatitis induced by Na-TDC The Na-TDC model was initially used in the 1980s,42,43 both with and without ductal infusion of various concentrations of trypsin. Thereafter, the model was employed frequently to explore the pathogenesis of AP with a commonly adopted regimen of 200 ml of 5% Na-TDC in the rat. Common findings included the perturbation of energy metabolism in the intestinal wall,44 impaired intestinal permeability,45 bacterial translocation,46,47 formation of platelet-activating factor,48 pulmonary endothelial barrier dysfunction,49 microvascular endothelial barrier dysfunction,50 activation of mast cells,51–53 induction of nitric oxide synthase,54 over-induced polyamine catabolism,55–57 increased high-mobility group box,58 increased catalytic activity of phospholipase A,59 upgraded cytokine levels60,61 and increased myeloperoxidase (MPO) activity.61 Models of pancreatitis induced by Na-TC and TLC-S Among these bile salts, the taurine-conjugated bile salt Na-TC is the most widely used and best characterized to date in inducing AP. The Na-TC ductal infusion model has been shown to induce pancreatic oedema, haemorrhage and necrosis in large animals, such as rabbits,62 swans,63 dogs64 and pigs.65,66 However, the pancreatic injury in these large animals is not sufficient to produce multiple organ dysfunction syndromes (MODS). Instead, the rat model of Na-TC infusion serves as a well-defined tool to research MODS in severe AP, as evidenced by lung,67,68 liver,69 gastric,70 intestinal,71,72 kidney72,73 and brain72,74 impairments, which mirror events in the human condition. Once infusion pressure and speed have been fixed by a pump, the severity of pancreatitis is determined by the concentration and volume of Na-TC infused. The

HPB 2012, 14, 73–81

75

regimen of Na-TC infusion was later standardized by Aho et al.75–77 in a series of studies in rats. The volume of 0.2 ml/kg at concentrations of 3.0%, 4.5% or 5.0% induced acute haemorrhage pancreatitis with 72-h mortality rates of 24%, 71% and 100%, respectively. Like the caerulein/lipopolysaccharide (LPS) model, this model is suitable for the study of bacterial translocation when combined with LPS,78 which has a dose-dependent effect on lethality in rats,79 and results in more severe MODS when trypsin is superimposed on the Na-TC/LPS model.80 In a recent study, Zhou et al.81 reported a model of infected severe AP induced by ductal infusion of Na-TC and Escherichia coli in rats. Experimental pancreatitis induced by Na-TC in the rat may have represented the reference standard of BAP for many years. However, the technique has recently been extended to the mouse model,82 thereby enabling the increased utility of genetic approaches.83 TLC-S, which has now been extensively characterized, is favoured by researchers over Na-TC in the induction of AP. Table 1 summarizes the characteristics of pancreatitis induced by Na-TC in different species.

Recent advances in experimental BAP in mice Recently, Laukkarinen et al.82 established a non-lethal BAP model in mice using a retrograde ductal infusion of 50 ml 2% Na-TC and published a detailed methodology.84 This protocol produced necrotizing pancreatitis in the head of the pancreas, but did not elicit associated lung injury. These authors’ parallel in vitro findings confirmed that Na-TC induced pathological Ca2+ signals and Ca2+-dependent trypsinogen activation and cell death in isolated PACs. This model in mice has been further validated.85,86 Ductal infusions at 2 ml/kg of 4% and 5% Na-TC resulted in necrotizing pancreatitis, lung injury and increased cytokines, causing mortality rates of 10% and 60% at 24 h, respectively.85 These new developments allow for coherent, coordinated studies of pancreatic pathological stimuli on many layers of scale from isolated mitochondria87 to whole animals,88 especially genetically manipulated mice. Gpbar1 The presence of Gpbar1 on the apical pole of PACs was first identified by Perides et al.13 using C57BL/6 mice deleted for Gpbar1. These authors infused 50 ml of 3 mm TLC-S or Na-TC into the pancreatic duct and found that, whereas TLC-S injection caused pancreatitis, Na-TC failed to do so. Gpbar1 knockout mice were completely protected against pancreatitis induced by TLC-S. In their parallel in vitro study, they found that 500 mm TLC-S elicited pathological Ca2+ signals, intrapancreatic trypsinogen activation and cell death in isolated PACs.13 Again, these effects were mostly absent in PACs from Gpbar1-/- mice. Interestingly, 500 mm TLC-S was still able to induce amylase secretion in isolated PACs from Gpbar1-/- mice, an event that was Ca2+-dependent. When the concentration of TLC-S was increased to 1 mm, the induced amylase secretion seemed to be neither Gpbar1- nor

© 2012 International Hepato-Pancreato-Biliary Association

HPB

76

Table 1 Characteristics of models of pancreatitis induced by Na-TC in different species

Dose

Species

Effect

Reference(s)

10%, 1.0 ml/kg

Dog

Significantly increased serum and ascites amylase, lipase levels; pancreatic parenchymal oedema, inflammatory cell infiltration and necrosis; other organ injury: N/A

64

5%, 1.0 ml/kg

Rabbit

Pancreatic oedema, inflammatory cell infiltration, necrosis, haemorrhage and fat necrosis; other organ injury: N/A

62

20%, 1.0 ml/kg

Pig

Significantly increased serum lipase level, WBC counts and neutrophils; significantly decreased mean blood pressure, serum lymphocytes and platelets; pancreatic oedema, haemorrhage, necrosis and accumulation of neutrophils, lymphocytes and macrophages; other organ injury: N/A

66

5%, 0.5 ml/kg plus 5% trypsin

Pig

Significantly increased serum amylase and WBC counts; focal liquefactive necrosis in most of the pancreas, with neutrophil infiltration; other organ injury: N/A

63

3–5%, 1.0 mg/kg

Rat

Significantly increased serum amylase, lipase and proinflammatory cytokine levels; pancreatic oedema, vacuolization, inflammation, haemorrhage, acinar cell and fat necrosis; lung, liver, gastric, kidney and brain injuries

67–72,74,123–125

2%, 50 ml

Mouse

Increased serum amylase and IL-6; transient increase of intrapancreatic trypsin activity; pancreatic oedema, leukocyte infiltration and necrosis; no mortality

82

4%, 5%, 2.0 ml/kg

Mouse

Increased serum amylase, lipase and IL-6; decreased serum IL-12; pancreatic oedema, leukocyte infiltration, necrosis, haemorrhage and fat necrosis; increased pulmonary BAL fluid albumin and MPO activity; 10% and 60% mortality rates at 24 h for 4% and 5% Na-TC, respectively

85

Na-TC, sodium taurocholate; WBC, white blood cell; IL, interleukin; BAL, bronchoalveolar lavage; MPO, myeloperoxidase; N/A, not applicable.

Ca2+-dependent. These findings suggest that: (i) the mechanism by which TLC-S acts on PACs is independent of bile acid co-transporters and exchangers, and occurs specifically through luminal surface Gpbar1; (ii) as amylase secretion seems to be independent of Gpbar1 and, at higher concentrations, even independent of Ca2+, non-receptor-mediated mechanisms may also account for complicated effects of bile acids on PACs, and (iii) unlike caerulein, TLC-S does not block pancreatic secretion, which suggests that the early damage to the pancreas during AP, at least in the case of TLC-S, can occur independently of the inhibition of zymogen secretion and intracellular trypsin activation. Cathepsin L Cathepsins are proteases that break down other proteins. They are found in all animals in many types of cell and are classified according to structure and catalytic type into serine, aspartic and cysteine families, the last of which includes cathepsin B and cathepsin L.89 Cathepsin B inhibitors or gene knockout have been found to block trypsinogen activation in vitro90 and to protect against AP in vivo.91–93 Cathepsin L demonstrates enzymatic properties similar to those of cathepsin B and is widely expressed and exhibits much stronger endoproteolytic activity than cathepsin B.94 The role of cathepsin L in AP was recently investigated by Wartmann et al.95 using Ctsl-/- mice96 in models of pancreatitis induced by Na-TC (2%, 50 ml) and caerulein, respectively. Similarly to cathepsin B,97 cathepsin L was expressed in human and mouse exocrine pancreas. In pancreatitis, it was found to be colocalized with trypsinogen and to be secreted into pancreatic juice. By contrast with Ctsb-/- mice, which showed a decrease in trypsin activity, Ctsl-/- mice demonstrated increased intrapancreatic

HPB 2012, 14, 73–81

trypsin activity upon pancreatitis.95 Interestingly, Ctsl-/- mice showed promoted apoptosis and reduced severity of pancreatitis compared with their littermates. These findings suggest that intrapancreatic trypsin activity may not reflect the severity of AP, and the crosstalk among trypsinogen activation, cathepsin B and cathepsin L needs further investigation. Toll-like receptor 9 and NOD-like receptor 3 inflammasome Zhang et al.98 recently demonstrated that cellular injury can release into the circulation endogenous damage-associated molecular patterns (DAMPs) that activate polymorphonuclear neutrophils through formyl peptide receptor-1 and Toll-like receptor-9 (TLR9), respectively. TLR9 is expressed on the cell surface and endocytic compartment and plays an important role in detecting self molecules and DNA.99 NOD-like receptor-3 (NLR3) is expressed in cytosol and is required by uric acid and ATP for inflammasome activation via plasma membrane puriergic receptor P2X7, which is activated by DAMPs.100,101 Hoque et al.102 studied the roles of TLR9, NLRP3 inflammasome (NLRP3-ASC) and DAMPs in AP using models of AP induced by TCL-S (3 mm, 50 ml) and caerulein, respectively. They found that, in the early stages of AP, circulating genomic DNA was markedly increased, but serum mitochondria appeared to be unaltered. This suggests that mitochondrial DAMPs may be largely restricted to the injury site.103 Deletion of TLR9 or application of its antagonist IRS954, as well as deletion of inflammasome components NLRP, ASC and P2X7 receptor or application of P2X7 receptor antagonist A-439079, were all shown to reduce the severity of AP induced by caerulein.102 Moreover, pancreatic prointerleukin-1b (pro-IL-1b) mRNA was reduced in TLR9-/- or

© 2012 International Hepato-Pancreato-Biliary Association

HPB

TLR9 antagonist-treated mice. In pancreatitis induced by TLC-S, pretreatment with TLR9 antagonist IRS954 significantly reduced serum amylase, pancreatic oedema, inflammation, necrosis and lung histology.102 This study highlighted new therapeutic potentials of DAMPs-receptor pathway antagonism, such as TLR9 and P2X7 receptor antagonists. Tumour necrosis factor-a and Ly-6Chi monocytes Recently, genetically altered mouse strains have been employed to show either the complete deletion of tumour necrosis factor-a (TNF-a) or the expression of human diphtheria toxin receptor (DTR) coupled to the CD11b promoter (CD11b-DTR).104 In CD11b-DTR mice, the severity of pancreatitis was associated with pancreatic Ly-6Chi monocytes/macrophages,105 which are released from bone marrow to injured tissue in response to distant organ injuries.106 Prior administration of diphtheria toxin (DT) abolished Ly-6Chi monocytes and prevented pancreatic oedema and necrosis in pancreatitis induced by either caerulein or Na-TC (37 mm, 50 ml). This protective effect by DT was reversed by adoptive transfer of purified Ly-6Chi monocytes harvested from CD11b-DTR mice or TNF-a+/+ donors but not from TNF-a-/donor mice.105 These findings indicate that monocytes and macrophages regulate pancreatic oedema and necrosis through the TNF-a pathway and therefore that targeting either Ly-6Chi monocytes or TNF-a may represent a promising strategy in the treatment of AP. Indeed, TNF-a antagonism has been evaluated as a pharmacological strategy for treating pancreatitis in response to promising results in numerous studies of various experimental AP models.107,108 However, other reports have demonstrated that TNF-a blockage was ineffective in the prevention of postendoscopic retrograde cholangiopancreatography pancreatitis in canines109 and even augmented the severity of caerulein-induced pancreatitis in rats.110 Moreover, neutralization of TNF-a has failed in sepsis clinical trials.111–113 These findings indicate that the extrapolation of laboratory results into clinical trials is complex. Rho-kinase, lymphocyte function antigen-1 and CD40 ligand Awla et al.114,115 and Abdulla et al.116 investigated the roles of Rhokinase, lymphocyte function antigen-1 (LFA-1) and CD40 ligand (CD40L) in experimental AP induced by Na-TC (5%, 10 ml) and/or caerulein. Rho-kinase inhibitor Y-27632 (5 mg/kg), given prior to the induction of pancreatitis, was found to dramatically reduce the severity of AP and associated lung injury.114 Moreover, pancreatitis-induced trypsinogen activation peptide levels were reduced in vivo and in vitro (caerulein) by Y-27632 treatment. However, the administration of Y-27632 after induction of pancreatitis had no significant effect on pancreatic or systemic injuries. These authors therefore postulated that Rho-kinase inhibitor may regulate trypsinogen activation in pancreatitis.114 In another study, they showed that both LFA gene deletion and anti-LFA-1 antibody were protective against AP in genetically targeted LFA-1 mice.115 The inhibition of LFA-1 function not only greatly reduced

HPB 2012, 14, 73–81

77

Na-TC-induced leukocyte adhesion, neutrophil accumulation in the pancreas and acinar cell necrosis, but also attenuated pulmonary neutrophil infiltration. However, trypsinogen activation induced by Na-TC was not affected.115 The authors concluded that LFA-1 may act on a downstream pathway of trypsinogen activation and that blocking LFA-1 may provide therapeutic potential.115 CD40L is a transmembrane glycoprotein that belongs to the TNF family of cell surface interaction molecules.117 Induced to express mainly on a CD4+ T cell subset, it has been found to activate platelets and to exert proinflammatory117–120 and procoagulant effects.121,122 Abdulla et al. examined responses in CD40L-/- mice to AP induced by either Na-TC or caerulein, but observed no beneficial effects in CD40L-/- mice compared with their littermates.116

Conclusions The extensive research conducted over the last decade has advanced our understanding of the deleterious effects of bile acids/salts on PACs and PDECs in the pathogenesis of BAP. As BAP models mirror the aetiology of gallstone-induced pancreatitis in the clinical setting, the application of these models in mice would facilitate studies on genetic aspects of this disease, as well as allowing the scrutiny of candidate therapeutic drugs. Recent advances in basic research using this BAP model have revealed the importance of Gpbar1 and cathepsin L, and have provided potential therapeutic targets such as the DAMPs-receptor pathway, TNF-a and Ly-6Chi monocytes. Further investigation might eventually lead to the discovery of effective treatments for AP. Acknowledgements We thank Professor Robert Sutton, Liverpool NIHR Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, University of Liverpool, for his continuous support as a visiting professor at Sichuan University. We also thank Dr Michael Chvanov, Department of Cellular and Molecular Physiology, University of Liverpool, for his support of this work. This project was supported by the National Natural Science Foundation of China (grant nos. 81072910 and 30801457), the National Institute for Health Research, UK, and the UK/China Postgraduate Research Scholarships for Excellence. Conflicts of interest None declared. References 1. Pandol SJ, Saluja AK, Imrie CW, Banks PA. (2007) Acute pancreatitis: bench to the bedside. Gastroenterology 132:1127–1151. 2. Su KH, Cuthbertson C, Christophi C. (2006) Review of experimental animal models of acute pancreatitis. HPB (Oxford) 8:264–286. 3. Chan YC, Leung PS. (2007) Acute pancreatitis: animal models and recent advances in basic research. Pancreas 34:1–14. 4. Pineda Torra I, Claudel T, Duval C, Kosykh V, Fruchart JC, Staels B. (2003) Bile acids induce the expression of the human peroxisome proliferator-activated receptor alpha gene via activation of the farnesoid X receptor. Mol Endocrinol 17:259–272. 5. Maruyama T, Miyamoto Y, Nakamura T, Tamai Y, Okada H, Sugiyama E et al. (2002) Identification of membrane-type receptor for bile acids (M-BAR). Biochem Biophys Res Commun 298:714–719.

© 2012 International Hepato-Pancreato-Biliary Association

HPB

78

6. Li T, Holmstrom SR, Kir S, Umetani M, Schmidt DR, Kliewer SA et al.

22. Voronina SG, Barrow SL, Gerasimenko OV, Petersen OH, Tepikin AV.

(2011) The G protein-coupled bile acid receptor, TGR5, stimulates gall-

(2004) Effects of secretagogues and bile acids on mitochondrial mem-

bladder filling. Mol Endocrinol 25:1066–1071.

brane potential of pancreatic acinar cells: comparison of different

7. Kawamata Y, Fujii R, Hosoya M, Harada M, Yoshida H, Miwa M et al. (2003) A G protein-coupled receptor responsive to bile acids. J Biol Chem 278:9435–9440. 8. Thomas C, Auwerx J, Schoonjans K. (2008) Bile acids and the membrane bile acid receptor TGR5 – connecting nutrition and metabolism. Thyroid 18:167–174. 9. Pols TW, Noriega LG, Nomura M, Auwerx J, Schoonjans K. (2011) The bile acid membrane receptor TGR5 as an emerging target in metabolism and inflammation. J Hepatol 54:1263–1272.

modes of evaluating DeltaPsim. J Biol Chem 279:27327–27338. 23. Voronina SG, Barrow SL, Simpson AW, Gerasimenko OV, da Silva Xavier G, Rutter GA et al. (2010) Dynamic changes in cytosolic and mitochondrial ATP levels in pancreatic acinar cells. Gastroenterology 138:1976–1987. 24. Venglovecz V, Rakonczay Z, Jr., Ozsvari B, Takacs T, Lonovics J, Varro A et al. (2008) Effects of bile acids on pancreatic ductal bicarbonate secretion in guinea pig. Gut 57:1102–1112. 25. Hegyi P, Pandol S, Venglovecz V, Rakonczay Z, Jr. (2011) The acinar–

10. Aromataris EC, Castro J, Rychkov GY, Barritt GJ. (2008) Store-operated

ductal tango in the pathogenesis of acute pancreatitis. Gut 60:544–552.

Ca(2+) channels and stromal interaction molecule 1 (STIM1) are targets

26. Maleth J, Venglovecz V, Razga Z, Tiszlavicz L, Rakonczay Z, Jr.,

for the actions of bile acids on liver cells. Biochim Biophys Acta

Hegyi P. (2011) Non-conjugated chenodeoxycholate induces severe

1783:874–885.

mitochondrial damage and inhibits bicarbonate transport in pancreatic

11. Castro J, Aromataris EC, Rychkov GY, Barritt GJ. (2009) A small com-

duct cells. Gut 60:136–138.

ponent of the endoplasmic reticulum is required for store-operated

27. Venglovecz V, Hegyi P, Rakonczay Z, Jr., Tiszlavicz L, Nardi A,

Ca2+ channel activation in liver cells: evidence from studies using

Grunnet M et al. (2011) Pathophysiological relevance of apical large-

TRPV1 and taurodeoxycholic acid. Biochem J 418:553–566.

conductance Ca(2)+-activated potassium channels in pancreatic duct

12. Kim JY, Kim KH, Lee JA, Namkung W, Sun AQ, Ananthanarayanan M et al. (2002) Transporter-mediated bile acid uptake causes Ca2+dependent cell death in rat pancreatic acinar cells. Gastroenterology 122:1941–1953. 13. Perides G, Laukkarinen JM, Vassileva G, Steer ML. (2010) Biliary acute pancreatitis in mice is mediated by the G-protein-coupled cell surface bile acid receptor Gpbar1. Gastroenterology 138:715– 725. 14. Lerch MM, Aghdassi AA. (2010) The role of bile acids in gallstoneinduced pancreatitis. Gastroenterology 138:429–433. 15. Senninger N, Moody FG, Coelho JC, Van Buren DH. (1986) The role of biliary obstruction in the pathogenesis of acute pancreatitis in the opossum. Surgery 99:688–693.

epithelial cells. Gut 60:361–369. 28. Bernard C. (1856) Lecons de Physiologie Experimentale. 2. Paris: Bailliere, p. 278. 29. Sun W, Watanabe Y, Toki A, Wang ZQ. (2007) Beneficial effects of hydrocortisone in induced acute pancreatitis of rats. Chin Med J 120:1757–1761. 30. Sun W, Watanabe Y, Wang ZQ. (2006) Expression and significance of ICAM-1 and its counter receptors LFA-1 and Mac-1 in experimental acute pancreatitis of rats. World J Gastroenterol 12:5005–5009. 31. Ottesen LH, Bladbjerg EM, Osman M, Lausten SB, Jacobsen NO, Gram J et al. (1999) Protein C activation during the initial phase of experimental acute pancreatitis in the rabbit. Dig Surg 16:486–495. 32. Osman MO, Kristensen JU, Jacobsen NO, Lausten SB, Deleuran B,

16. Voronina S, Longbottom R, Sutton R, Petersen OH, Tepikin A. (2002)

Deleuran M et al. (1998) A monoclonal anti-interleukin 8 antibody

Bile acids induce calcium signals in mouse pancreatic acinar cells:

(WS-4) inhibits cytokine response and acute lung injury in experimental

implications for bile-induced pancreatic pathology. J Physiol 540 (Pt 1):49–55. 17. Kuipers F, Bijleveld CM, Kneepkens CM, van Zanten A, Fernandes J, Vonk RJ. (1985) Sulphated lithocholic acid conjugates in serum from children with hepatic and intestinal diseases. Scand J Gastroenterol 20:1255–1261. 18. Ross PE, Pennington CR, Bouchier IA. (1977) Gas-liquid chromatographic assay of serum bile acids. Anal Biochem 80:458–465.

severe acute necrotizing pancreatitis in rabbits. Gut 43:232–239. 33. Terry TR, Grant DA, Hermon-Taylor J. (1987) Intraduct enterokinase is lethal in rats with experimental bile-salt pancreatitis. Br J Surg 74:40– 43. 34. Rosen HR, Tuchler H. (1992) Pulmonary injury in acute experimental pancreatitis correlates with elevated levels of free fatty acids in rats. HPB Surg 6:79–90. 35. Rattner DW, Napolitano LM, Corsetti J, Compton C, Stanford GG,

19. Gerasimenko JV, Flowerdew SE, Voronina SG, Sukhomlin TK,

Warshaw AL et al. (1990) Hypocalcaemia in experimental pancreatitis

Tepikin AV, Petersen OH et al. (2006) Bile acids induce Ca2+ release

occurs independently of changes in serum non-esterified fatty acid

from both the endoplasmic reticulum and acidic intracellular calcium stores through activation of inositol trisphosphate receptors and ryanodine receptors. J Biol Chem 281:40154–40163. 20. Fischer L, Gukovskaya AS, Penninger JM, Mareninova OA, Friess H,

levels. Int J Pancreatol 6:249–262. 36. Schmidt J, Lewandrowsi K, Warshaw AL, Compton CC, Rattner DW. (1992) Morphometric characteristics and homogeneity of a new model of acute pancreatitis in the rat. Int J Pancreatol 12:41–51.

Gukovsky I et al. (2007) Phosphatidylinositol 3-kinase facilitates bile

37. Schmidt J, Rattner DW, Lewandrowski K, Compton CC, Mandavilli U,

acid-induced Ca(2+) responses in pancreatic acinar cells. Am J Physiol

Knoefel WT et al. (1992) A better model of acute pancreatitis for evalu-

Gastrointest Liver Physiol 292:875–886.

ating therapy. Ann Surg 215:44–56.

21. Malo A, Kruger B, Seyhun E, Schafer C, Hoffmann RT, Goke B et al.

38. Bloechle C, Kusterer K, Kuehn RM, Schneider C, Knoefel WT, Izbicki

(2010) Tauroursodeoxycholic acid reduces endoplasmic reticulum

JR. (1998) Inhibition of bradykinin B2 receptor preserves microcircula-

stress, trypsin activation, and acinar cell apoptosis while increasing

tion in experimental pancreatitis in rats. Am J Physiol 274 (Pt 1):42–51.

secretion in rat pancreatic acini. Am J Physiol Gastrointest Liver Physiol

39. Rivera JA, Werner J, Warshaw AL, Lewandrowski KB, Rattner DW,

299:877–886.

HPB 2012, 14, 73–81

Fernandez del Castillo C. (1998) Lexipafant fails to improve survival in

© 2012 International Hepato-Pancreato-Biliary Association

HPB

severe necrotizing pancreatitis in rats. Int J Pancreatol 23:101– 106.

79

pancreatitis induced by intraductal infusion of taurodeoxycholate. Surgery 144:49–56.

40. Chen CC, Wang SS, Lee FY, Tsay SH, Wu SL, Lu RH et al. (1999)

57. Jin HT, Lamsa T, Merentie M, Hyvonen MT, Sand J, Raty S et al. (2008)

Prophylactic octreotide reduces the severity of histopathologic changes

Polyamine levels in the pancreas and the blood change according to the

and haemodynamic shock in early taurodeoxycholate-induced experimental pancreatitis. Proc Natl Sci Counc Repub China B 23:1–6. 41. Kilian M, Heukamp I, Gregor JI, Bretthauer C, Walz MK, Jacobi CA et al. (2006) n-3, n-6, and n-9 polyunsaturated fatty acids – which composition in parenteral nutrition decreases severity of acute haemorrhagic necrotizing pancreatitis in rats? Int J Colorectal Dis 21:57–63. 42. Evander A, Ihse I, Lundquist I. (1981) Influence of hormonal stimulation by caerulein on acute experimental pancreatitis in the rat. Eur Surg Res 13:257–268. 43. Seung WP, Feldman BF. (1985) Early phase components of the kallikrein kinin system in haemorrhagic ascitic fluid and plasma in the rat with induced acute pancreatitis. Am J Vet Res 46:1961–1966. 44. Ederoth P, Sun Z, Nordstrom CH, Andersson R. (2002) Experimental pancreatitis causes acute perturbation of energy metabolism in the intestinal wall. Pancreas 25:270–276. 45. Wang XD, Wang Q, Andersson R, Ihse I. (1996) Alterations in intestinal function in acute pancreatitis in an experimental model. Br J Surg 83:1537–1543. 46. Wang X, Andersson R, Soltesz V, Leveau P, Ihse I. (1996) Gut origin sepsis, macrophage function, and oxygen extraction associated with acute pancreatitis in the rat. World J Surg 20:299–307; discussion 307–308. 47. Leveau P, Wang X, Soltesz V, Ihse I, Andersson R. (1996) Alterations in intestinal motility and microflora in experimental acute pancreatitis. Int J Pancreatol 20:119–125. 48. Kald B, Kald A, Ihse I, Tagesson C. (1993) Release of platelet-activating factor in acute experimental pancreatitis. Pancreas 8:440–442.

severity of pancreatitis. Pancreatology 8:15–24. 58. Cheng BQ, Liu CT, Li WJ, Fan W, Zhong N, Zhang Y et al. (2007) Ethyl pyruvate improves survival and ameliorates distant organ injury in rats with severe acute pancreatitis. Pancreas 35:256–261. 59. Shi C, Zhao X, Wang X, Zhao L, Andersson R et al. (2007) Potential effects of PKC or protease inhibitors on acute pancreatitis-induced tissue injury in rats. Vascul Pharmacol 46:406–411. 60. Chen CC, Wang SS, Tsay SH, Lee FY, Lu RH, Chang FY et al. (2006) Effects of gabexate mesilate on serum inflammatory cytokines in rats with acute necrotizing pancreatitis. Cytokine 33:95–99. 61. Shi C, Zhao X, Wang X, Andersson R. (2005) Role of nuclear factorkappaB, reactive oxygen species and cellular signalling in the early phase of acute pancreatitis. Scand J Gastroenterol 40:103–108. 62. Apodaca-Torrez FR, Goldenberg A, Lobo EJ, Farah JF, Trivino T, Montero EF et al. (2007) Evaluation of the effects of non-iodinized and iodinized ionic contrast media and gadoteric acid in acute necrotizing pancreatitis: experimental study in rabbits. Pancreas 35:41–44. 63. Wang J, Shao C, Zuo C, Zheng J, Hao J, Zhang F et al. (2011) Establishment of a secondary infection model of severe acute pancreatitis in swine. Pancreas 40:114–119. 64. Satoh H, Harada M, Tashiro S, Shiroya T, Imawaka H, Machii K. (2004) The effect of continuous arterial infusion of gabexate mesilate (FOY-007) on experimental acute pancreatitis. J Med Invest 51:186– 193. 65. Kinnala PJ, Kuttila KT, Gronroos JM, Havia TV, Nevalainen TJ, Niinikoski J. (1999) Central haemodynamics in experimental acute pancreatitis. Eur J Surg 165:598–603.

49. Wang XD, Borjesson A, Sun ZW, Wallen R, Deng XM, Zhang HY et al.

66. Merilainen S, Makela J, Anttila V, Koivukangas V, Kaakinen H,

(1998) The association of type II pneumocytes and endothelial perme-

Niemela E et al. (2008) Acute oedematous and necrotic pancreatitis in a

ability with the pulmonary custocyte system in experimental acute pancreatitis. Eur J Clin Invest 28:778–785. 50. Wang X, Sun Z, Borjesson A, Haraldsen P, Aldman M, Deng X et al.

porcine model. Scand J Gastroenterol 43:1259–1268. 67. de Campos T, Deree J, Martins JO, Loomis WH, Shenvi E, Putnam JG et al. (2008) Pentoxifylline attenuates pulmonary inflammation and neu-

(1999) Treatment with lexipafant ameliorates the severity of pancreatic

trophil activation in experimental acute pancreatitis. Pancreas 37:42–49.

microvascular endothelial barrier dysfunction in rats with acute haem-

68. Chen X, Li SL, Wu T, Liu JD. (2008) Proteasome inhibitor ameliorates

orrhagic pancreatitis. Int J Pancreatol 25:45–52. 51. Dib M, Zhao X, Wang XD, Andersson R. (2002) Role of mast cells in the development of pancreatitis-induced multiple organ dysfunction. Br J Surg 89:172–178. 52. Dib M, Zhao X, Wang X, Andersson R. (2002) Mast cells contribute to early pancreatitis-induced systemic endothelial barrier dysfunction. Pancreatology 2:396–401. 53. Zhao X, Dib M, Wang X, Widegren B, Andersson R. (2005) Influence of mast cells on the expression of adhesion molecules on circulating and migrating leukocytes in acute pancreatitis-associated lung injury. Lung 183:253–264. 54. Chen CC, Wang SS, Tsay SH, Lee FY, Lu RH, Chang FY et al. (2004) Effects of nitric oxide synthase inhibitors on retrograde bile salt-induced pancreatitis in rats. J Chin Med Assoc 67:9–14. 55. Jin HT, Lamsa T, Nordback PH, Hyvonen MT, Raty S, Nordback I et al. (2011) Polyamine catabolism in relation to trypsin activation and apoptosis in experimental acute pancreatitis. Pancreatology 11:83–91. 56. Jin HT, Lamsa T, Hyvonen MT, Sand J, Raty S, Grigorenko N et al. (2008) A polyamine analogue bismethylspermine ameliorates severe

HPB 2012, 14, 73–81

severe acute pancreatitis and associated lung injury of rats. World J Gastroenterol 14:3249–3253. 69. Paszt A, Takacs T, Rakonczay Z, Kaszaki J, Wolfard A, Tiszlavicz L et al. (2004) The role of the glucocorticoid-dependent mechanism in the progression of sodium taurocholate-induced acute pancreatitis in the rat. Pancreas 29:75–82. 70. Dang SC, Zhang JX, Qu JG, Wang XQ, Fan X. (2007) Ligustrazine alleviates gastric mucosal injury in a rat model of acute necrotizing pancreatitis. Hepatobiliary Pancreat Dis Int 6:213–218. 71. Leveau P, Wang X, Sun Z, Borjesson A, Andersson E, Andersson R. (2005) Severity of pancreatitis-associated gut barrier dysfunction is reduced following treatment with the PAF inhibitor lexipafant. Biochem Pharmacol 69:1325–1331. 72. Zhang XP, Ye Q, Jiang XG, Ma ML, Zhu FB, Zhang RP et al. (2007) Preparation method of an ideal model of multiple organ injury of rat with severe acute pancreatitis. World J Gastroenterol 13:4566–4573. 73. Yamano M, Miyata K, Yamada T. (1998) Protective effect of a pancreatic elastase inhibitor against a variety of acute pancreatitis in rats. Jpn J Pharmacol 77:193–203.

© 2012 International Hepato-Pancreato-Biliary Association

HPB

80

74. Yang YL, Li JP, Li KZ, Dou KF. (2004) Tumour necrosis factor-alpha antibody prevents brain damage of rats with acute necrotizing pancreatitis. World J Gastroenterol 10:2898–2900.

91. Korsten MA, Dlugosz JW. (1993) Cathepsin B inhibition in two models of acute pancreatitis. Int J Pancreatol 14:149–155. 92. Van Acker GJ, Saluja AK, Bhagat L, Singh VP, Song AM, Steer ML.

75. Aho HJ, Koskensalo SM, Nevalainen TJ. (1980) Experimental pancre-

(2002) Cathepsin B inhibition prevents trypsinogen activation and

atitis in the rat. Sodium taurocholate-induced acute haemorrhagic pan-

reduces pancreatitis severity. Am J Physiol Gastrointest Liver Physiol

creatitis. Scand J Gastroenterol 15:411–416.

283:794–800.

76. Aho HJ, Nevalainen TJ, Lindberg RL, Aho AJ. (1980) Experimental

93. Halangk W, Lerch MM, Brandt-Nedelev B, Roth W, Ruthenbuerger M,

pancreatitis in the rat. The role of phospholipase A in sodium

Reinheckel T et al. (2000) Role of cathepsin B in intracellular trypsino-

taurocholate-induced acute haemorrhagic pancreatitis. Scand J

gen activation and the onset of acute pancreatitis. J Clin Invest

Gastroenterol 15:1027–1031. 77. Aho HJ, Nevalainen TJ, Aho AJ. (1983) Experimental pancreatitis in the rat. Development of pancreatic necrosis, ischaemia and oedema after intraductal sodium taurocholate injection. Eur Surg Res 15:28–36.

106:773–781. 94. Kirschke H, Barrett AJ. (1985) Cathepsin L – a lysosomal cysteine proteinase. Prog Clin Biol Res 180:61–69. 95. Wartmann T, Mayerle J, Kahne T, Sahin-Toth M, Ruthenburger M,

78. Yamanel L, Mas MR, Comert B, Isik AT, Aydin S, Mas N et al. (2005) The

Matthias R et al. (2010) Cathepsin L inactivates human trypsinogen,

effect of activated protein C on experimental acute necrotizing pancre-

whereas cathepsin L-deletion reduces the severity of pancreatitis in

atitis. Crit Care 9:184–190.

mice. Gastroenterology 138:726–737.

79. Segersvard R, Sylvan M, Lempinen M, Larsson J, Permert J. (2004)

96. Roth W, Deussing J, Botchkarev VA, Pauly-Evers M, Saftig P, Hafner A

Impact of chronic and acute high-fat feeding on acute experimental

et al. (2000) Cathepsin L deficiency as molecular defect of furless:

pancreatitis complicated by endotoxinaemia. Scand J Gastroenterol

hyperproliferation of keratinocytes and perturbation of hair follicle

39:74–80.

cycling. FASEB J 14:2075–2086.

80. Yamano M, Umeda M, Miyata K, Yamada T. (1998) Protective effect of the

97. Kukor Z, Mayerle J, Kruger B, Toth M, Steed PM, Halangk W et al.

combined treatment of pancreatic and neutrophil elastase inhibitors on

(2002) Presence of cathepsin B in the human pancreatic secretory

acute pancreatitis elicited by lipopolysaccharide in rats given intraductal

pathway and its role in trypsinogen activation during hereditary pancre-

injection of taurocholate plus trypsin. Naunyn Schmiedebergs Arch Pharmacol 357:558–564. 81. Zhou M, Zhang Q, Zeng Q, Qiu Y, Liu N, Zhu Y et al. (2008) Establishment of an infected necrotizing pancreatitis model by retrograde pancreatic duct injection of sodium taurocholate and E. coli in rats. J Huazhong Univ Sci Technolog Med Sci 28:73–76. 82. Laukkarinen JM, Van Acker GJ, Weiss ER, Steer ML, Perides G. (2007) A mouse model of acute biliary pancreatitis induced by retrograde pancreatic duct infusion of Na-taurocholate. Gut 56:1590–1598. 83. Schafer C, Tietz AB, Goke B. (2005) Pathophysiology of acute experimental pancreatitis: lessons from genetically engineered animal models and new molecular approaches. Digestion 71:162–172. 84. Perides G, van Acker GJ, Laukkarinen JM, Steer ML. (2010) Experimental acute biliary pancreatitis induced by retrograde infusion of bile acids into the mouse pancreatic duct. Nat Protoc 5:335–341. 85. Wittel UA, Wiech T, Chakraborty S, Boss B, Lauch R, Batra SK et al. (2008) Taurocholate-induced pancreatitis: a model of severe necrotizing pancreatitis in mice. Pancreas 36:9–21. 86. Ziegler KM, Wade TE, Wang S, Swartz-Basile DA, Pitt HA, Zyromski NJ. (2011) Validation of a novel, physiologic model of experimental acute pancreatitis in the mouse. Am J Transl Res 3:159–165. 87. Odinokova IV, Sung KF, Mareninova OA, Hermann K, Evtodienko Y, Andreyev A et al. (2009) Mechanisms regulating cytochrome c release in pancreatic mitochondria. Gut 58:431–442.

atitis. J Biol Chem 277:21389–21396. 98. Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W et al. (2010) Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 464:104–107. 99. Barrat FJ, Meeker T, Gregorio J, Chan JH, Uematsu S, Akira S et al. (2005) Nucleic acids of mammalian origin can act as endogenous ligands for Toll-like receptors and may promote systemic lupus erythematosus. J Exp Med 202:1131–1139. 100. Martinon F, Mayor A, Tschopp J. (2009) The inflammasomes: guardians of the body. Annu Rev Immunol 27:229–265. 101. Kono H, Chen CJ, Ontiveros F, Rock KL. (2010) Uric acid promotes an acute inflammatory response to sterile cell death in mice. J Clin Invest 120:1939–1949. 102. Hoque R, Sohail M, Malik A, Sarwar S, Luo Y, Shah A et al. (2011) TLR9 and the NLRP3 inflammasome link acinar cell death with inflammation in acute pancreatitis. Gastroenterology 141:358–369. 103. McDonald B, Pittman K, Menezes GB, Hirota SA, Slaba I, Waterhouse CC et al. (2010) Intravascular danger signals guide neutrophils to sites of sterile inflammation. Science 330:362–366. 104. Shifrin AL, Chirmule N, Zhang Y, Raper SE. (2005) Macrophage ablation attenuates adenoviral vector-induced pancreatitis. Surgery 137:545– 551. 105. Perides G, Weiss ER, Michael ES, Laukkarinen JM, Duffield JS, Steer ML. (2011) TNF-a-dependent regulation of acute pancreatitis

88. Mareninova OA, Hermann K, French SW, O'Konski MS, Pandol SJ,

severity by Ly-6Chi monocytes in mice. J Biol Chem 286:13327–13335.

Webster P et al. (2009) Impaired autophagic flux mediates acinar cell

106. Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, Kayal S et al. (2007)

vacuole formation and trypsinogen activation in rodent models of acute

Monitoring of blood vessels and tissues by a population of monocytes

pancreatitis. J Clin Invest 119:3340–3355. 89. Reiser J, Adair B, Reinheckel T. (2010) Specialized roles for cysteine cathepsins in health and disease. J Clin Invest 120:3421–3431. 90. Saluja AK, Donovan EA, Yamanaka K, Yamaguchi Y, Hofbauer B,

with patrolling behaviour. Science 317:666–670. 107. Malleo G, Mazzon E, Siriwardena AK, Cuzzocrea S. (2007) Role of tumour necrosis factor-alpha in acute pancreatitis: from biological basis to clinical evidence. Shock 28:130–140.

Steer ML. (1997) Cerulein-induced in vitro activation of trypsinogen in

108. Malleo G, Mazzon E, Siriwardena AK, Cuzzocrea S. (2007) TNF-alpha as

rat pancreatic acini is mediated by cathepsin B. Gastroenterology

a therapeutic target in acute pancreatitis – lessons from experimental

113:304–310.

models. ScientificWorldJournal 7:431–448.

HPB 2012, 14, 73–81

© 2012 International Hepato-Pancreato-Biliary Association

HPB

81

109. Buscaglia JM, Simons BW, Prosser BJ, Ruben DS, Giday SA, Magno P

117. Zhang S, Rahman M, Qi Z, Thorlacius H. (2011) Simvastatin antago-

et al. (2008) Etanercept, a TNF-alpha binding agent, is ineffective in the

nizes CD40L secretion, CXC chemokine formation, and pulmonary infil-

prevention of post-ERCP pancreatitis in canines. JOP 9:456–467. 110. Guice KS, Oldham KT, Remick DG, Kunkel SL, Ward PA. (1991) Antitumour necrosis factor antibody augments oedema formation in caerulein-induced acute pancreatitis. J Surg Res 51:495–499.

tration of neutrophils in abdominal sepsis. J Leukoc Biol 89:735–742. 118. Nurden AT. (2011) Platelets, inflammation and tissue regeneration. Thromb Haemost 105 (Suppl. 1):S13–S33. 119. Apelseth TO, Hervig T, Wentzel-Larsen T, Petersen K, Reikvam H,

111. Abraham E, Wunderink R, Silverman H, Perl TM, Nasraway S, Levy H

Bruserud O. (2011) A prospective observational study of the

et al. (1995) Efficacy and safety of monoclonal antibody to human

effect of platelet transfusions on levels of platelet-derived cytokines,

tumour necrosis factor alpha in patients with sepsis syndrome. A ran-

chemokines and interleukins in acute leukaemia patients with

domized, controlled, double-blind, multicentre clinical trial. TNF-alpha

severe chemotherapy-induced cytopenia. Eur Cytokine Netw 22:52–

MAb Sepsis Study Group. JAMA 273:934–941.

62.

112. Dhainaut JF, Vincent JL, Richard C, Lejeune P, Martin C, Fierobe L et al.

120. Chew M, Rahman M, Ihrman L, Erson A, Zhang S, Thorlacius H. (2010)

(1995) CDP571, a humanized antibody to human tumour necrosis

Soluble CD40L (CD154) is increased in patients with shock. Inflamm

factor-alpha: safety, pharmacokinetics, immune response, and influence of the antibody on cytokine concentrations in patients with septic shock. CPD571 Sepsis Study Group. Crit Care Med 23:1461–1469. 113. Reinhart K, Wiegand-Lohnert C, Grimminger F, Kaul M, Withington S, Treacher D et al. (1996) Assessment of the safety and efficacy of

Res 59:979–982. 121. Kaya Z, Ozdemir K, Kayrak M, Gul EE, Altunbas G, Duman C et al. (2011) Soluble CD40 ligand levels in acute pulmonary embolism: a prospective, randomized, controlled study. Heart Vessels DOI:10.1007/ s00380-011-0142-4 [Epub ahead of print].

the monoclonal anti-tumour necrosis factor antibody-fragment, MAK

122. Giannini S, Falcinelli E, Bury L, Guglielmini G, Rossi R, Momi S et al.

195F, in patients with sepsis and septic shock: a multicentre, random-

(2011) Interaction with damaged vessel wall in vivo in humans induces

ized, placebo-controlled, dose-ranging study. Crit Care Med 24:733–

platelets to express CD40L resulting in endothelial activation. No effect

742.

of aspirin intake. Am J Physiol Heart Circ Physiol 300:H2072–H2079.

114. Awla D, Hartman H, Abdulla A, Zhang S, Rahman M, Regner S et al.

123. Jung KH, Song SU, Yi T, Jeon MS, Hong SW, Zheng HM et al. (2011)

(2011) Rho-kinase signalling regulates trypsinogen activation and tissue

Human bone marrow-derived clonal mesenchymal stem cells inhibit

damage in severe acute pancreatitis. Br J Pharmacol 162:648–658.

inflammation and reduce acute pancreatitis in rats. Gastroenterology

115. Awla D, Abdulla A, Zhang S, Roller J, Menger MD, Regner S et al. (2011) Lymphocyte function antigen-1 regulates neutrophil recruitment and tissue damage in acute pancreatitis. Br J Pharmacol 163:413–423. 116. Abdulla A, Awla D, Jeppsson B, Regner S, Thorlacius H. (2011) CD40L is not involved in acute experimental pancreatitis. Eur J Pharmacol DOI:10.1016/j.ejphar.2011.03.008 [Epub ahead of print].

HPB 2012, 14, 73–81

140:998–1008. 124. Xia XM, Wang FY, Wang ZK, Wan HJ, Xu WA, Lu H. (2010) Emodin enhances alveolar epithelial barrier function in rats with experimental acute pancreatitis. World J Gastroenterol 16:2994–3001. 125. Qian M, Fang L, Cui Y. (2010) Expression of NOD2 in a rat model of acute pancreatitis. Pancreas 39:1034–1040.

© 2012 International Hepato-Pancreato-Biliary Association