Topic Highlight Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Nov 14, 2013; 19(42): 7222-7230
Published online Nov 14, 2013. doi: 10.3748/wjg.v19.i42.7222
Animal models of pancreatitis: Can it be translated to human pain study?
Jing-Bo Zhao, Dong-Hua Liao, Mech-Sense, Department of Gastroenterology and Surgery, Aalborg University Hospital, DK 9000 Aalborg, Denmark
Jing-Bo Zhao, Dong-Hua Liao, Institute of Clinical Medicine, Aarhus University, DK 8200 Aarhus N, Denmark
Jing-Bo Zhao, Dong-Hua Liao, The College of Bioengineering, Chongqing University, Chongqing 400016, China
Thomas Dahl Nissen, Biomedical Research Laboratory, Aalborg University Hospital, DK 9000 Aalborg, Denmark
Author contributions: Zhao JB, Liao DH and Nissen TD designed, drafted, revised and finally approved this paper.
Correspondence to: Jing-Bo Zhao, MD, PhD, Mech-Sense, Department of Gastroenterology and Surgery, Aalborg University Hospital, Sdr. Skovvej 15, DK 9000 Aalborg, Denmark. jz@rn.dk
Telephone: +45-9-9326907 Fax: +45-9-9326801
Received: June 13, 2013
Revised: August 12, 2013
Accepted: August 20, 2013
Published online: November 14, 2013

Abstract

Chronic pancreatitis affects many individuals around the world, and the study of the underlying mechanisms leading to better treatment possibilities are important tasks. Therefore, animal models are needed to illustrate the basic study of pancreatitis. Recently, animal models of acute and chronic pancreatitis have been thoroughly reviewed, but few reviews address the important aspect on the translation of animal studies to human studies. It is well known that pancreatitis is associated with epigastric pain, but the understanding regarding to mechanisms and appropriate treatment of this pain is still unclear. Using animal models to study pancreatitis associated visceral pain is difficult, however, these types of models are a unique way to reveal the mechanisms behind pancreatitis associated visceral pain. In this review, the animal models of acute, chronic and un-common pancreatitis are briefly outlined and animal models related to pancreatitis associated visceral pain are also addressed.

Key Words: Animal model, Pancreatitis, Visceral pain, Mechanism

Core tip: Choosing the right model of pancreatitis is difficult and the scientific rationale needs to be carefully considered. Furthermore, no model of pancreatitis parallels all classical symptoms and the question under investigation is of importance when choosing a model. One of the main symptoms of chronic pancreatitis is visceral pain and in order to improve the pain treatment and obtain more knowledge about the physiology behind the pancreatitis associated visceral pain, animal models of pancreatitis associated visceral pain are needed.



INTRODUCTION

Pancreatitis represents a common disorder of the gastrointestinal tract. Acute pancreatitis (AP) has an incidence ranged from 4.9 to 35 per 100000 populations[1], whereas chronic pancreatitis (CP) has an incidence from 2.4 to 4.4 per 100000 populations[2]. The etiology of this disease is complex and so far a variety of environmental factors including alcohol abuse, nicotine habits, hereditary factors, efferent duct obstructions, immunological factors and rare metabolic factors have all been described. However, the pathophysiology of AP and CP remains poorly defined[3]. As a result appropriate therapies are still limited, and prognosis has not improved to date, which is mainly due to the lack of a satisfactory animal model of pancreatitis[4,5].

It is well known that pancreatitis is associated with visceral pain, however, the understanding of pain signaling related to pancreatitis is poor[6]. In order to facilitate the development of new pharmaceutical treatments for AP and CP, characterization of the mediators and receptors or ion channels on the sensory nerve terminals and the pathways of the pain signaling are needed. Therefore, in this aspect, the animal models of pancreatitis are needed in parallel in order to explore the mechanism behind pancreatitis associated visceral pain, as this is difficult to study in humans.

In this review, we briefly outline the animal models of acute, chronic and un-common pancreatitis as well as animal models related to pancreatitis associated visceral pain.

ANIMAL MODELS OF ACUTE PANCREATITIS

AP is an inflammatory condition of the pancreas characterized clinically by abdominal pain and elevated levels of pancreatic enzymes in the blood[7]. Other characteristics of AP include edema, acinar cell necrosis, hemorrhage, and severe inflammation of the pancreas. Severe AP may lead to systemic inflammatory response syndrome and multi-organ dysfunction syndrome, which account for the high mortality rates of AP[8,9]. As it is difficult to study AP in the clinic, animal studies are important in order to understand the pathogenesis of AP, however an AP model which is strictly comparable to human AP is still needed. The current animal models of AP have contributed to our knowledge of mechanisms involved in early cellular events, pathogenesis and pathophysiology of AP[10,11]. We have illustrated the summary of existing AP animal models in Table 1[12-59]. Details of different AP animal models including advantages, disadvantages and clinical relevance can be found in a recently published review[4]. From a methodological aspect, selecting the appropriate AP animal model depends on the objectives of each study as different animal models are targeted to different AP features. For developing the effective treatment for AP in the clinic, continued investigation of AP animal models are needed.

Table 1 Different animal models of acute pancreatitis.
MethodsModels and examples
Non-invasiveHormone-induced
Acute caerulein pancreatitis of rats[12], mice[13], dogs[14], and syrian hamsters[15]
Trinidadian scorpion toxin induced acute pancreatitis in dogs[16]
Alcohol-induced: rats[17-19], cats[20] and dogs[21]
Immune-mediated
Ovalbumin in rabbit[22]
Foreign serum in mice[23] and rat[24]
Spontaneous model of autoimmune acute pancreatitis mice[25]
Diet-induced: Fed a choline-deficient diet containing ethionine in mice[26]
Gene knockout: Interleukin (IL)-1 and tumour necrosis factor-α[27], IL-6[28], IL-10[29], chemoattractant cytokine receptor-1[30], neurokinin-1 receptor[31], intercellular adhesion molecule 1 (ICAM-1)[32], metallothionein-1[33], cathepsin B[34], mouse a2-macroglobulin and murinoglobulin[35], complement factor C5a[36], granulocyte-macrophage colony-stimulating factor[37] and phospholipase A2[38]
L-arginine-induced: Administration of a large dose of L-arginine in rats[39,40]
InvasiveClosed duodenal loop (CDL): Dog[41] and rat[42,43]
Antegrade pancreatic duct perfusion: Cat[44] and rat[45]
Various compounds infusion into the pancreatic duct: Rat[46] and dog[47]
Combined intraductal glycodeoxycholic acid with intravenous caerulein: Rat[48]
Vascular-induced
Impairment of pancreatic circulation in dogs[49]
To occlude pancreatic arteries in rats[50]
Occlusion of pancreatic veins in dogs[51] and in rats[52]
Complete but reversible ischaemia of the pancreas by occluding different arteries using microvascular clips: Rats[53] and canine[54]
Duct ligation
Ligating the distal bile duct at the level of the duodenum[55]
Combined pancreatic duct ligation with the secretory stimulation, secretin in dogs[56]
Combining duct ligation with both secretory stimulation and minimal arterial blood[57]
Duct-ligated opossums models[58]
Transient obstruction of the sphincter of Oddi (SO) in Australian brush tailed possums[59]
ANIMAL MODELS OF CHRONIC PANCREATITIS

A recently published review[5] has described the most frequently used and best established models for CP in animals. The majority of the animal models are rodent models, since mice and rats are easy to handle and there is a steadily increasing number of genetic models obtained by gene deletion or transgenic expression of genetic variants. In the same way for animal models of AP, the models of CP can be classified into noninvasive or nonsurgical models and invasive or surgical models. Table 2 summarizes different animal models of CP[60-100].

Table 2 Different animal models of chronic pancreatitis.
MethodsModels and examples
Non-invasiveCaerulein-induced
Serial caerulein injections in mice[60] and rats[61]
Combination of repetitive caerulein injections with toxins and other agents such as lipopolysaccharides[62], cyclosporin A[63], dibutyltin dichloride[64] and Alcohol[65-67]
Intraperitoneal caerulein injections are administered in genetically transformed mice such as TRX-1 transgenic mice[68,69]
Arginine-induced
A single L-arginine injection in rat[70]
Serial L-arginine injections[70-72]
Alcohol feeding-induced: Lieber-DeCarli formula[73-76]
Genetic models: Wistar Bonn/Kobori (WBN/Kob) rats[77-79]; R122H transgenic mice[80]; SPINK3-deficient (SPINK3-/-) mice[81]; CFTR-deficient (cftrm1UNC) mice[82] and CFTR(-/-) pigs[83]; Kif3a-deficient mice[84]; PERK-deficient (PERK-/-) mice[85]; Interleukin 1-β transgenic mice[86]
InvasiveSodium taurocholate-induced: Retrograde infusion of sodium taurocholate (NaTc) into the pancreatic duct system of the rat[87]
Oleic acid-induced: Retrograde infusion of oleic acid[72,88-91], viscous solution of zein[92], mixture of zein-oleic acid, or viscous solution consisting of zein-oleic acid-linoleic acid[93,94] into rat pancreatic duct
Congestion of pancreatic fluid flow: Combination of transient stasis of pancreatic juice flow and mild pancreatic duct injury[95]
Duct ligation model
Ligation of the common bile duct close to the duodenum pancreatic tissue in dogs[96], mouse[97] and pigs[98]
Incomplete pancreatic duct ligation in canine[99]
Occlusion with two different tissue glues in the rat[100]

In the non-invasive models, repetitive caerulein injections are amongst the most widely used models. Firstly, caerulein injections are relatively easy to perform and show a high reliability and reproducibility. Secondly, other compounds mediating injury such as lipopolysaccharides or cyclosporin A can easily be added to the design. Thirdly, serial caerulein injections can be performed in transgenic or knockout animals. It is likely that there are dose and frequency dependency for caerulein. The most translational models include repetitive injections of L-arginine, which appears to produce CP similar to that in humans[70-72]. In this model, fibrotic tissues are progressively replaced with adipose tissue. Due to the high impact of alcohol consumption as a risk factor on the pathogenesis in human pancreatic diseases, alcohol has frequently been used to trigger CP in animal models[73,74]. However, it is still being considered whether a model for CP induced by alcohol alone is feasible or satisfactory. The combination of alcohol feeding with caerulein injections exacerbates the course of pancreatitis and consequently increases pancreatic fibrosis and the loss of parenchyma.

Genetic animal models of CP are suitable for different studies. It is well known that activation of trypsinogen is one of the key events in the early phase of pancreatitis, and therefore genetic abnormalities found in the trypsinogen gene and in its inhibitors might be of particular importance of which R122H transgenic mice[80] are a good example. Transgenic expression of the R122H mutation of murine trypsin 4 in the pancreas of mice led to progressive fibrosis and chronic inflammation of the pancreas. Repetitive inductions of experimental pancreatitis with supramaximal doses of cerulein resulted in extensive deposition of collagen in periacinar and perilobular spaces of this transgenic animal. However other genetic models might also help us to understand how CP develops[77-79,81,83-86,101].

Invasive animal models can also be used to induce CP. As an example, retrograde infusion of sodium taurocholate (NaTc) into the pancreatic duct[46] or intraductal infusion of NaTc[72] can generate pancreatitis, however the structure of the pancreatic tissue will return to an almost normal state after 14 d. Retrograde infusion of oleic acid[72,88-91], viscous solution of zein[92], a mixture of zein-oleic acid or a viscous solution consisting of zeinoleic acid-linoleic acid[93,94] into rat pancreatic duct will cause severe pancreatic atrophy with irregular fibrosis and fat replacement over a period of 6 mo. However, these models of pancreatitis appear quite distinct from CP in humans. As one factor alone is inadequate to cause persistent pancreatic injury, a combination of transient stasis of pancreatic juice flow and mild pancreatic duct injury is a well established and reliable method to generate CP in animal models[95]. It is well known that pancreatic ductal hypertension contributes to the pathogenesis of CP; therefore animal models can also be generated by complete obstruction of the pancreatic duct[96-98], incomplete pancreatic duct ligation[99] and occlusion with different tissue glues[100]. Yamamoto et al[102] developed an animal model with pancreatic ductal hypertension and demonstrated that this plays an important role in the onset and development of CP in rats. However, models for CP based on duct obstruction are not common and there is only a minority of studies examining the morphological and biochemical changes of the pancreas after duct ligation[41,103,104].

ANIMAL MODELS OF UN-COMMON PANCREATITIS

Un-common types of pancreatitis can include autoimmune pancreatitis (AIP), hereditary pancreatitis[105], groove pancreatitis[106], tropical pancreatitis, pancreatitis in ectopic or heterotopic pancreatic tissue, ascaris-induced pancreatitis, pancreatitis in cystic fibrosis, pancreas divisum, annular pancreas, pancreatic cancer manifesting as AP, and duodenal villous adenoma with pancreatitis. With exception of AIP and hereditary pancreatitis, no relevant animal models were found for other un-common pancreatitis. Furthermore, hereditary pancreatitis animal models were mentioned in the genetic animal models of CP above. Therefore only animal models of AIP are briefly introduced in this section.

To date, several animal models of AIP have been described. The first model involves the adoptive transfer of amylase-specific (an antigen mainly located in acinar cells) CD4+ T cells and results in pancreatitis in naive syngenic recipient animals[107]. Notably, the histological lesions of this model mimic the lobulocentric inflammatory reaction in type 1 AIP. A model developed by immunization of neonatally thymectomized mice with CA (an antigen mainly located on the pancreatic epithelium) and later transfer of CD4+ lymphocytes resulted in a duct-centric pattern of pancreatitis resembling type 2 AIP[108]. In another model, NTx-NFS/sld mice spontaneously developed sialoadenitis in which a-fodrin was involved as an autoantigen, as reported in some patients with Sjogren syndrome and AIP[109]. Transforming growth factor-β (TGFβ) appears to be an important regulatory factor in maintaining immune homeostasis. Loss of TGFβ signalling contributes to AIP in TGFβ dominant negative mutant mice[110].

Recently two animal models for AIP were proposed. The WBN/Kob rat model, associated with congenital decreased peripheral Tregs spontaneously develops sialoadenitis, thyroiditis, sclerosing cholangitis and tubulointerstitial nephritis[111]. Although the target antigens remain unclear, CD8+ cells may be the effector cell in this rat model[112]. Another recently described animal model of AIP is the Treg-deficient NOD mouse[113]. CD28KO mice spontaneously develop AIP that closely resembles the human disease[113]. More recently, Haruta et al[114] investigated the possible involvement of chronic, persistent exposure to avirulent bacteria in the pathogenesis of AIP using C57BL/6 mice.

Existing animal models for AIP have several limitations. In most models the disease is induced by adoptive transfer of autoreactive cells and/or antibodies rather than spontaneous development of the disease with identical antigen specificity. The distribution of lesions produced in animal models for AIP is also variable. This may be attributed to the diversity of target antigens, different methods of immune staining and different mouse strains. In addition, typical histopathological findings of AIP (e.g., lymphoplasmacytic infiltration with fibrosis, obliterative phlebitis and GELs) are rarely observed in animal models. Thus, there is a need to develop spontaneous animal models with identical autoantigens and typical histopathological findings for AIP.

VISCERAL PAIN IN ANIMAL MODELS OF CHRONIC PANCREATITIS

One of the main clinical symptoms of CP in humans is pain, occurring either in episodes or as a constant disabling pain[115,116]. Hence, an important goal of treatment for CP is to relieve the pain. The analgesic treatment is often inadequate as the pathophysiology behind CP as well as the mechanisms behind the accompanying pain is not yet fully understood[117]. As described in the previous sections, no single animal model displays all aspects of CP and each of the different models display histological similarities to the human condition to various degrees. In order to improve the pain treatment and obtain more knowledge about the physiology behind CP associated pain, animal models of CP associated pain are needed.

Rat models of CP where pancreatic nociception was investigated, have been established through invasive, noninvasive and spontaneous models[118,119]. In these models pancreatic pain has been shown through both mechanical and thermal stimulation of the abdomen (referred pain[120]) as well as direct electrical stimulation of the pancreas[118,121]. These models had histopathological similarities to the human disease and had progressive fibrosis and inflammation. Furthermore, the models showed correlation between nociceptive behaviour and increased expression of nerve growth factor (NGF) in the pancreas and calcitonin gene-related peptide (CGRP), substance P (SP), proteinase-activated receptor 2 (PAR2), and brain-derived neurotrophic factor (BDNF) in thoracic dorsal root ganglion and spinal cord segments[118,122-124]. Increased expression of NGF, CGRP, SP and BDNF has also been shown in human patients with CP[125-127].

Several animal models have investigated the mechanisms involved in pain accompanying CP. Takamido et al[119] reported morphological changes of the nervous system being involved in development of CP pain. This study suggested that elongation of dorsal root ganglia axons and enlargement of intrapancreatic nerve bundles as being a possible mechanism of pain generation in CP. On a supraspinal level, findings have suggested that descending facilitation from the rostral ventromedial medulla plays an important role in persistent pain associated with CP[128]. Furthermore, recent rat experiments have suggested that spinal microglia becomes activated during CP and has an important role in initiating and maintaining chronic pain[129].

TRANSLATION OF PANCREATITIS-ASSOCIATED VISCERAL PAIN STUDY FROM ANIMAL TO HUMAN

It may be difficult to use animal models to study pancreatitis associated visceral pain as pain is a subjective experience. However animal models are needed to explore the molecular mechanisms behind pancreatitis associated visceral pain as this is difficult to study in humans. The molecular mechanisms behind the chronic pain associated with CP are poorly understood, but within recent years, animal experiments have suggested some mechanisms that might be involved. The transient receptor potential vanilloid 1 (TRPV1) and transient receptor potential ankyrin 1 (TRPA1) have been shown to be contributing factors to pain in CP[122,130,131]. It has been shown that CP is accompanied by an increased level of NGF which caused an up-regulation of TRPV1 expression and sensitivity, resulting in hyperalgesia and allodynia[122,130]. TRPA1 is important in both inflammation and pain in CP and can be sensitized through activation of PAR2[131].

The mechanisms mentioned above could be used as targets for the development of novel therapeutics, aiming at treating the chronic pain accompanying CP. Neutralizing antibodies against neurotransmitters such as BDNF and NGF[124,130] or receptor specific antagonists[122] has proven to reverse the characteristic nociceptive behavioral changes induced by CP in several of the experimental models. Furthermore, inhibition of trypsin or inhibition of microglia activation has also abrogated the pain related behavior seen in response to CP[123,129]. All these different mechanisms of pain treatment in CP models could have a potential as targets for novel pharmacological treatment of the chronic pain associated with CP in human patients. Also established analgesic drugs such as gabapentin, buprenorphine, and morphine have been tested in animal models of CP[118,121,132], and shown to have analgesic effect. However, many of these therapeutic approaches need to be tested in humans, before their true potential analgesic treatment of CP pain in humans can be established. It is known that some of these analgesic mechanisms are species specific and specific to the different models of induced CP.

CONCLUSION

Choosing the right model of pancreatitis is difficult and the scientific rationale needs to be carefully considered. Furthermore, no model of pancreatitis parallels all classical symptoms and the question under investigation is of importance when choosing a model. One of the main symptoms of CP is visceral pain and in order to improve the pain treatment and obtain more knowledge about the physiology behind the pancreatitis associated visceral pain, animal models of pancreatitis associated visceral pain are needed.

Footnotes

P- Reviewer: Chow WK S- Editor: Gou SX L- Editor: A E- Editor: Zhang DN

References
1.  Peery AF, Dellon ES, Lund J, Crockett SD, McGowan CE, Bulsiewicz WJ, Gangarosa LM, Thiny MT, Stizenberg K, Morgan DR. Burden of gastrointestinal disease in the United States: 2012 update. Gastroenterology. 2012;143:1179-87.e1-3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1355]  [Cited by in F6Publishing: 1373]  [Article Influence: 114.4]  [Reference Citation Analysis (1)]
2.  DiMagno MJ, DiMagno EP. Chronic pancreatitis. Curr Opin Gastroenterol. 2012;28:523-531.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
3.  Khokhar AS, Seidner DL. The pathophysiology of pancreatitis. Nutr Clin Pract. 2004;19:5-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
4.  Su KH, Cuthbertson C, Christophi C. Review of experimental animal models of acute pancreatitis. HPB (Oxford). 2006;8:264-286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 104]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
5.  Aghdassi AA, Mayerle J, Christochowitz S, Weiss FU, Sendler M, Lerch MM. Animal models for investigating chronic pancreatitis. Fibrogenesis Tissue Repair. 2011;4:26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 89]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
6.  Barreto SG, Saccone GT. Pancreatic nociception--revisiting the physiology and pathophysiology. Pancreatology. 2012;12:104-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
7.  Vege SS. Pathogenesis of acute pancreatitis.  Available from: http://www.uptodate.com/contents/pathogenesis-of-acute-pancreatitis#H2 (15-04-2013).  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Chan YC, Leung PS. Acute pancreatitis: animal models and recent advances in basic research. Pancreas. 2007;34:1-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 107]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
9.  Steer ML, Saluja A. Experimental acute pancreatitis: studies of the early events that lead to cell injury. The Pancreas, Biology, Pathobiology and Disease. 2nd ed. New York: Raven Press 1993; 489-499.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Fétaud V, Frossard JL, Farina A, Pastor CM, Bühler L, Dumonceau JM, Hadengue A, Hochstrasser DF, Lescuyer P. Proteomic profiling in an animal model of acute pancreatitis. Proteomics. 2008;8:3621-3631.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
11.  Pini M, Rhodes DH, Castellanos KJ, Hall AR, Cabay RJ, Chennuri R, Grady EF, Fantuzzi G. Role of IL-6 in the resolution of pancreatitis in obese mice. J Leukoc Biol. 2012;91:957-966.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 27]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
12.  Watanabe O, Baccino FM, Steer ML, Meldolesi J. Supramaximal caerulein stimulation and ultrastructure of rat pancreatic acinar cell: early morphological changes during development of experimental pancreatitis. Am J Physiol. 1984;246:G457-G467.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Niederau C, Ferrell LD, Grendell JH. Caerulein-induced acute necrotizing pancreatitis in mice: protective effects of proglumide, benzotript, and secretin. Gastroenterology. 1985;88:1192-1204.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Renner IG, Wisner JR. Exogenous secretin ameliorates ceruletide induced acute pancreatitis in the dog. Dig Dis Sci. 1983;28:946 (abstract).  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Adler G, Kern HF, Scheele GA. Experimental models and concepts in acute pancreatitis. The exocrine pancreas: biology, pathobiology, diseases. New York: Raven 1986; .  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Pantoja JL, Renner IG, Abramson SB, Edmondson HA. Production of acute hemorrhagic pancreatitis in the dog using venom of the scorpion, Buthus quinquestriatus. Dig Dis Sci. 1983;28:429-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
17.  Siech M, Heinrich P, Letko G. Development of acute pancreatitis in rats after single ethanol administration and induction of a pancreatic juice edema. Int J Pancreatol. 1991;8:169-175.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Weber H, Merkord J, Jonas L, Wagner A, Schröder H, Käding U, Werner A, Dummler W. Oxygen radical generation and acute pancreatitis: effects of dibutyltin dichloride/ethanol and ethanol on rat pancreas. Pancreas. 1995;11:382-388.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Werner J, Laposata M, Fernández-del Castillo C, Saghir M, Iozzo RV, Lewandrowski KB, Warshaw AL. Pancreatic injury in rats induced by fatty acid ethyl ester, a nonoxidative metabolite of alcohol. Gastroenterology. 1997;113:286-294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 144]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
20.  Wedgwood KR, Adler G, Kern H, Reber HA. Effects of oral agents on pancreatic duct permeability. A model of acute alcoholic pancreatitis. Dig Dis Sci. 1986;31:1081-1088.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 17]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
21.  Friedman HS, Lowery R, Shaughnessy E, Scorza J. The effects of ethanol on pancreatic blood flow in awake and anesthetized dogs. Proc Soc Exp Biol Med. 1983;174:377-382.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  THAL A. Studies on pancreatitis. II. Acute pancreatic necrosis produced experimentally by the arthus sensitization reaction. Surgery. 1955;37:911-917.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Janigan DT, Nevalainen TJ, MacAulay MA, Vethamany VG. Foreign serum-induced pancreatitis in mice. I. A new model of acute pancreatitis. Lab Invest. 1975;33:591-607.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Nevalainen TJ. Pancreatic injury caused by intraductal injection of foreign serum in rat. Virchows Arch B Cell Pathol. 1978;27:89-98.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Kanno H, Nose M, Itoh J, Taniguchi Y, Kyogoku M. Spontaneous development of pancreatitis in the MRL/Mp strain of mice in autoimmune mechanism. Clin Exp Immunol. 1992;89:68-73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 83]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
26.  Lombardi B, Estes LW, Longnecker DS. Acute hemorrhagic pancreatitis (massive necrosis) with fat necrosis induced in mice by DL-ethionine fed with a choline-deficient diet. Am J Pathol. 1975;79:465-480.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Denham W, Yang J, Fink G, Denham D, Carter G, Ward K, Norman J. Gene targeting demonstrates additive detrimental effects of interleukin 1 and tumor necrosis factor during pancreatitis. Gastroenterology. 1997;113:1741-1746.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 120]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
28.  Suzuki S, Miyasaka K, Jimi A, Funakoshi A. Induction of acute pancreatitis by cerulein in human IL-6 gene transgenic mice. Pancreas. 2000;21:86-92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 37]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
29.  Gloor B, Todd KE, Lane JS, Rigberg DA, Reber HA. Mechanism of increased lung injury after acute pancreatitis in IL-10 knockout mice. J Surg Res. 1998;80:110-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
30.  Gerard C, Frossard JL, Bhatia M, Saluja A, Gerard NP, Lu B, Steer M. Targeted disruption of the beta-chemokine receptor CCR1 protects against pancreatitis-associated lung injury. J Clin Invest. 1997;100:2022-2027.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Grady EF, Yoshimi SK, Maa J, Valeroso D, Vartanian RK, Rahim S, Kim EH, Gerard C, Gerard N, Bunnett NW. Substance P mediates inflammatory oedema in acute pancreatitis via activation of the neurokinin-1 receptor in rats and mice. Br J Pharmacol. 2000;130:505-512.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Frossard JL, Saluja A, Bhagat L, Lee HS, Bhatia M, Hofbauer B, Steer ML. The role of intercellular adhesion molecule 1 and neutrophils in acute pancreatitis and pancreatitis-associated lung injury. Gastroenterology. 1999;116:694-701.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Fu K, Tomita T, Sarras MP, De Lisle RC, Andrews GK. Metallothionein protects against cerulein-induced acute pancreatitis: analysis using transgenic mice. Pancreas. 1998;17:238-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 31]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
34.  Halangk W, Lerch MM, Brandt-Nedelev B, Roth W, Ruthenbuerger M, Reinheckel T, Domschke W, Lippert H, Peters C, Deussing J. Role of cathepsin B in intracellular trypsinogen activation and the onset of acute pancreatitis. J Clin Invest. 2000;106:773-781.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 431]  [Cited by in F6Publishing: 393]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
35.  Umans L, Serneels L, Overbergh L, Stas L, Van Leuven F. alpha2-macroglobulin- and murinoglobulin-1- deficient mice. A mouse model for acute pancreatitis. Am J Pathol. 1999;155:983-993.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Bhatia M, Saluja AK, Singh VP, Frossard JL, Lee HS, Bhagat L, Gerard C, Steer ML. Complement factor C5a exerts an anti-inflammatory effect in acute pancreatitis and associated lung injury. Am J Physiol Gastrointest Liver Physiol. 2001;280:G974-G978.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Frossard JL, Saluja AK, Mach N, Lee HS, Bhagat L, Hadenque A, Rubbia-Brandt L, Dranoff G, Steer ML. In vivo evidence for the role of GM-CSF as a mediator in acute pancreatitis-associated lung injury. Am J Physiol Lung Cell Mol Physiol. 2002;283:L541-L548.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Mizuma K, Schröder T, Kaarne M, Korpela H, Lempinen M. Serum phospholipase A2 in diet-induced pancreatitis. Eur Surg Res. 1984;16:156-161.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Hegyi P, Rakonczay Z, Sári R, Góg C, Lonovics J, Takács T, Czakó L. L-arginine-induced experimental pancreatitis. World J Gastroenterol. 2004;10:2003-2009.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Varga IS, Matkovics B, Czako L, Hai DQ, Kotorman M, Takacs T, Sasvari M. Oxidative stress changes in L-arginine-induced pancreatitis in rats. Pancreas. 1997;14:355-359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 22]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
41.  Pfeffer RB, Stasior O, Hinton JW. The clinical picture of the sequential development of acute hemorrhagic pancreatitis in the dog. Surg Forum. 1957;8:248-251.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Nevalainen TJ, Seppä A. Acute pancreatitis caused by closed duodenal loop in the rat. Scand J Gastroenterol. 1975;10:521-527.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Orda R, Hadas N, Orda S, Wiznitzer T. Experimental acute pancreatitis. Inducement by taurocholate sodium-trypsin injection into a temporarily closed duodenal loop in the rat. Arch Surg. 1980;115:327-329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
44.  Widdison AL, Alvarez C, Reber HA. The low-pressure duct perfusion model of acute pancreatitis. Eur Surg Res. 1992;24 Suppl 1:55-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
45.  Olazábal A. Effect of prostaglandins E2 and I2 and of indomethacin on deoxycholic acid-induced damage to the rat bile-pancreatic duct. Gastroenterology. 1983;84:928-934.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Aho HJ, Nevalainen TJ, Aho AJ. Experimental pancreatitis in the rat. Development of pancreatic necrosis, ischemia and edema after intraductal sodium taurocholate injection. Eur Surg Res. 1983;15:28-36.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Musa BE, Nelson AW, Gillette EL, Ferguson HL, Lumb WV. A model to study acute pancreatitis in the dog. J Surg Res. 1976;21:51-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
48.  Schmidt J, Rattner DW, Lewandrowski K, Compton CC, Mandavilli U, Knoefel WT, Warshaw AL. A better model of acute pancreatitis for evaluating therapy. Ann Surg. 1992;215:44-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 546]  [Cited by in F6Publishing: 620]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
49.  Barzilai A, Ryback BJ, Medina JA, Toth L, Dreiling DA. The morphological changes of the pancreas in hypovolemic shock and the effect of pretreatment with steroids. Int J Pancreatol. 1987;2:23-32.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Redha F, Uhlschmid G, Ammann RW, Freiburghaus AU. Injection of microspheres into pancreatic arteries causes acute hemorrhagic pancreatitis in the rat: a new animal model. Pancreas. 1990;5:188-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
51.  Pfeffer RB, Lazzarini-Robertson A, Safadi D, Mixter G, Secoy CF, Hinton JW. Gradations of pancreatitis, edematous, through hemorrhagic, experimentally produced by controlled injection of microspheres into blood vessels in dogs. Surgery. 1962;51:764-769.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Sjövall S, Holmin T, Evander A, Stenram U. Splenic and gastro-duodenal vein occlusion--influence on the pancreatic gland and on the outcome of experimental pancreatitis. Int J Pancreatol. 1988;3:143-149.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Hoffmann TF, Leiderer R, Waldner H, Arbogast S, Messmer K. Ischemia reperfusion of the pancreas: a new in vivo model for acute pancreatitis in rats. Res Exp Med (Berl). 1995;195:125-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 61]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
54.  Broe PJ, Zuidema GD, Cameron JL. The role of ischemia in acute pancreatitis: studies with an isolated perfused canine pancreas. Surgery. 1982;91:377-382.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Baxter JN, Jenkins SA, Day DW, Roberts NB, Cowell DC, Mackie CR, Shields R. Effects of somatostatin and a long-acting somatostatin analogue on the prevention and treatment of experimentally induced acute pancreatitis in the rat. Br J Surg. 1985;72:382-385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 67]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
56.  Popper HL, Necheles H. Edema of the pancreas. Surg Gynecol Obstet. 1942;74:123.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Popper HL, Necheles H, Russell KC. Transition of pancreatic edema into pancreatic necrosis. Surg Gynecol Obstet. 1948;87:79-82.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Senninger N, Moody FG, Coelho JC, Van Buren DH. The role of biliary obstruction in the pathogenesis of acute pancreatitis in the opossum. Surgery. 1986;99:688-693.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Chen JW, Thomas A, Woods CM, Schloithe AC, Toouli J, Saccone GT. Sphincter of Oddi dysfunction produces acute pancreatitis in the possum. Gut. 2000;47:539-545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 41]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
60.  Neuschwander-Tetri BA, Burton FR, Presti ME, Britton RS, Janney CG, Garvin PR, Brunt EM, Galvin NJ, Poulos JE. Repetitive self-limited acute pancreatitis induces pancreatic fibrogenesis in the mouse. Dig Dis Sci. 2000;45:665-674.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Elsässer HP, Haake T, Grimmig M, Adler G, Kern HF. Repetitive cerulein-induced pancreatitis and pancreatic fibrosis in the rat. Pancreas. 1992;7:385-390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
62.  Segersvärd R, Sylván M, Lempinen M, Larsson J, Permert J. Impact of chronic and acute high-fat feeding on acute experimental pancreatitis complicated by endotoxinaemia. Scand J Gastroenterol. 2004;39:74-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
63.  Vaquero E, Molero X, Tian X, Salas A, Malagelada JR. Myofibroblast proliferation, fibrosis, and defective pancreatic repair induced by cyclosporin in rats. Gut. 1999;45:269-277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 36]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
64.  Sparmann G, Merkord J, Jäschke A, Nizze H, Jonas L, Löhr M, Liebe S, Emmrich J. Pancreatic fibrosis in experimental pancreatitis induced by dibutyltin dichloride. Gastroenterology. 1997;112:1664-1672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 87]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
65.  Gukovsky I, Lugea A, Shahsahebi M, Cheng JH, Hong PP, Jung YJ, Deng QG, French BA, Lungo W, French SW. A rat model reproducing key pathological responses of alcoholic chronic pancreatitis. Am J Physiol Gastrointest Liver Physiol. 2008;294:G68-G79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 59]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
66.  Deng X, Wang L, Elm MS, Gabazadeh D, Diorio GJ, Eagon PK, Whitcomb DC. Chronic alcohol consumption accelerates fibrosis in response to cerulein-induced pancreatitis in rats. Am J Pathol. 2005;166:93-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 86]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
67.  Perides G, Tao X, West N, Sharma A, Steer ML. A mouse model of ethanol dependent pancreatic fibrosis. Gut. 2005;54:1461-1467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 77]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
68.  Ohashi S, Nishio A, Nakamura H, Asada M, Tamaki H, Kawasaki K, Fukui T, Yodoi J, Chiba T. Overexpression of redox-active protein thioredoxin-1 prevents development of chronic pancreatitis in mice. Antioxid Redox Signal. 2006;8:1835-1845.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
69.  Leung PS, Chan YC. Role of oxidative stress in pancreatic inflammation. Antioxid Redox Signal. 2009;11:135-165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 184]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
70.  Delaney CP, McGeeney KF, Dervan P, Fitzpatrick JM. Pancreatic atrophy: a new model using serial intra-peritoneal injections of L-arginine. Scand J Gastroenterol. 1993;28:1086-1090.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
71.  Weaver C, Bishop AE, Polak JM. Pancreatic changes elicited by chronic administration of excess L-arginine. Exp Mol Pathol. 1994;60:71-87.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
72.  Yamaguchi T, Kihara Y, Taguchi M, Nagashio Y, Tashiro M, Nakamura H, Otsuki M. Persistent destruction of the basement membrane of the pancreatic duct contributes to progressive acinar atrophy in rats with experimentally induced pancreatitis. Pancreas. 2005;31:365-372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 15]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
73.  Lieber CS, DeCarli LM. Alcoholic liver injury: experimental models in rats and baboons. Adv Exp Med Biol. 1975;59:379-393.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Lieber CS, Decarli LM. Animal models of ethanol dependence and liver injury in rats and baboons. Fed Proc. 1976;35:1232-1236.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Sarles H, Lebreuil G, Tasso F, Figarella C, Clemente F, Devaux MA, Fagonde B, Payan H. A comparison of alcoholic pancreatitis in rat and man. Gut. 1971;12:377-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 104]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
76.  Tsukamoto H, Towner SJ, Yu GS, French SW. Potentiation of ethanol-induced pancreatic injury by dietary fat. Induction of chronic pancreatitis by alcohol in rats. Am J Pathol. 1988;131:246-257.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Ohashi K, Kim JH, Hara H, Aso R, Akimoto T, Nakama K. WBN/Kob rats. A new spontaneously occurring model of chronic pancreatitis. Int J Pancreatol. 1990;6:231-247.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Hashimoto T, Yamada T, Yokoi T, Sano H, Ando H, Nakazawa T, Ohara H, Nomura T, Joh T, Itoh M. Apoptosis of acinar cells is involved in chronic pancreatitis in Wbn/Kob rats: role of glucocorticoids. Pancreas. 2000;21:296-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
79.  Mori M, Fu X, Chen L, Zhang G, Higuchi K. Hereditary pancreatitis model WBN/Kob rat strain has a unique haplotype in the Pdwk1 region on chromosome 7. Exp Anim. 2009;58:409-413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
80.  Archer H, Jura N, Keller J, Jacobson M, Bar-Sagi D. A mouse model of hereditary pancreatitis generated by transgenic expression of R122H trypsinogen. Gastroenterology. 2006;131:1844-1855.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 83]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
81.  Ohmuraya M, Hirota M, Araki K, Baba H, Yamamura K. Enhanced trypsin activity in pancreatic acinar cells deficient for serine protease inhibitor kazal type 3. Pancreas. 2006;33:104-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 41]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
82.  Dimagno MJ, Lee SH, Hao Y, Zhou SY, McKenna BJ, Owyang C. A proinflammatory, antiapoptotic phenotype underlies the susceptibility to acute pancreatitis in cystic fibrosis transmembrane regulator (-/-) mice. Gastroenterology. 2005;129:665-681.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 55]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
83.  Rogers CS, Stoltz DA, Meyerholz DK, Ostedgaard LS, Rokhlina T, Taft PJ, Rogan MP, Pezzulo AA, Karp PH, Itani OA. Disruption of the CFTR gene produces a model of cystic fibrosis in newborn pigs. Science. 2008;321:1837-1841.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 611]  [Cited by in F6Publishing: 558]  [Article Influence: 34.9]  [Reference Citation Analysis (0)]
84.  Lin F, Hiesberger T, Cordes K, Sinclair AM, Goldstein LS, Somlo S, Igarashi P. Kidney-specific inactivation of the KIF3A subunit of kinesin-II inhibits renal ciliogenesis and produces polycystic kidney disease. Proc Natl Acad Sci USA. 2003;100:5286-5291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 445]  [Cited by in F6Publishing: 431]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
85.  Harding HP, Zeng H, Zhang Y, Jungries R, Chung P, Plesken H, Sabatini DD, Ron D. Diabetes mellitus and exocrine pancreatic dysfunction in perk-/- mice reveals a role for translational control in secretory cell survival. Mol Cell. 2001;7:1153-1163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 933]  [Cited by in F6Publishing: 927]  [Article Influence: 40.3]  [Reference Citation Analysis (0)]
86.  Marrache F, Tu SP, Bhagat G, Pendyala S, Osterreicher CH, Gordon S, Ramanathan V, Penz-Osterreicher M, Betz KS, Song Z. Overexpression of interleukin-1beta in the murine pancreas results in chronic pancreatitis. Gastroenterology. 2008;135:1277-1287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 82]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
87.  Aho HJ, Nevalainen TJ, Lindberg RL, Aho AJ. Experimental pancreatitis in the rat. The role of phospholipase A in sodium taurocholate-induced acute haemorrhagic pancreatitis. Scand J Gastroenterol. 1980;15:1027-1031.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 49]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
88.  Mundlos S, Adler G, Schaar M, Koop I, Arnold R. Exocrine pancreatic function in oleic acid-induced pancreatic insufficiency in rats. Pancreas. 1986;1:29-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
89.  Henry JP, Steinberg WM. Pancreatic function tests in the rat model of chronic pancreatic insufficiency. Pancreas. 1993;8:622-626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 4]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
90.  Yamaguchi T, Nakamura H, Kihara Y, Taguchi M, Yoshikawa H, Otsuki M. Long-term overexpression of membrane type-1 matrix metalloproteinase and matrix metalloproteinase-2 in oleic acid-induced pancreatitis in rats. Pancreas. 2002;24:348-356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
91.  Tashiro M, Nakamura H, Taguchi M, Yamaguchi T, Yoshikawa H, Fukumitsu K, Kihara Y, Otsuki M. Oleic acid-induced pancreatitis alters expression of transforming growth factor-beta1 and extracellular matrix components in rats. Pancreas. 2003;26:197-204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
92.  Tomita T, Rhodes J, Falscroft J, Doull V, Kimmel JR, Pollock HG. Endocrine pancreas in the rat model of exocrine pancreatic insufficiency. Pancreas. 1988;3:568-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
93.  Steinberg WM, Burns MK, Henry JP, Nochomovitz LE, Anderson KK. Cerulein induces hyperplasia of the pancreas in a rat model of chronic pancreatic insufficiency. Pancreas. 1987;2:176-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
94.  Kataoka K, Sasaki T, Yorizumi H, Sakagami J, Kashima K. Pathophysiologic studies of experimental chronic pancreatitis in rats induced by injection of zein-oleic acid-linoleic acid solution into the pancreatic duct. Pancreas. 1998;16:289-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
95.  Yamaguchi T, Kihara Y, Yamamoto M, Otsuki M. Transient stasis of pancreatic fluid flow together with mild injury of the pancreatic duct cause chronic pancreatitis. Dig Dis Sci. 2011;56:2326-2332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
96.  Churg A, Richter WR. Early changes in the exocrine pancreas of the dog and rat after ligation of the pancreatic duct. A light and electron microscopic study. Am J Pathol. 1971;63:521-546.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Watanabe S, Abe K, Anbo Y, Katoh H. Changes in the mouse exocrine pancreas after pancreatic duct ligation: a qualitative and quantitative histological study. Arch Histol Cytol. 1995;58:365-374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 67]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
98.  Boerma D, Straatsburg IH, Offerhaus GJ, Gouma DJ, van Gulik TM. Experimental model of obstructive, chronic pancreatitis in pigs. Dig Surg. 2003;20:520-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 23]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
99.  Tanaka T, Ichiba Y, Fujii Y, Itoh H, Kodama O, Dohi K. New canine model of chronic pancreatitis due to chronic ischemia with incomplete pancreatic duct obstruction. Digestion. 1988;41:149-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 18]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
100.  Isaksson G, Lundquist I, Ihse I. Effects on the exocrine and endocrine pancreas of duct occlusion with two different tissue glues in the rat. Eur Surg Res. 1983;15:136-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
101.  Cano DA, Sekine S, Hebrok M. Primary cilia deletion in pancreatic epithelial cells results in cyst formation and pancreatitis. Gastroenterology. 2006;131:1856-1869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 108]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
102.  Yamamoto M, Otani M, Otsuki M. A new model of chronic pancreatitis in rats. Am J Physiol Gastrointest Liver Physiol. 2006;291:G700-G708.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
103.  Klein ES, Grateron H, Rudick J, Dreiling DA. Pancreatic intraductal pressure: I. A consideration of regulatory factors. Am J Gastroenterol. 1983;78:507-509.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  De Rai P, Franciosi C, Confalonieri GM, Biffi R, Andreoni B, Uggeri F, Malesci A. Effects of somatostatin on acute pancreatitis induced in rats by injection of taurocholate and trypsin into a temporarily closed duodenal loop. Int J Pancreatol. 1988;3:367-373.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Rosendahl J, Bödeker H, Mössner J, Teich N. Hereditary chronic pancreatitis. Orphanet J Rare Dis. 2007;2:1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 78]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
106.  Tyagi P, Thaper S, Bhatia V, Sharma M. Often a missed type of pancreatitis: groove pancreatitis. Indian J Gastroenterol. 2012;31:215-216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
107.  Zen Y, Fujii T, Harada K, Kawano M, Yamada K, Takahira M, Nakanuma Y. Th2 and regulatory immune reactions are increased in immunoglobin G4-related sclerosing pancreatitis and cholangitis. Hepatology. 2007;45:1538-1546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 526]  [Cited by in F6Publishing: 469]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
108.  Okazaki K, Uchida K, Ohana M, Nakase H, Uose S, Inai M, Matsushima Y, Katamura K, Ohmori K, Chiba T. Autoimmune-related pancreatitis is associated with autoantibodies and a Th1/Th2-type cellular immune response. Gastroenterology. 2000;118:573-581.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 410]  [Cited by in F6Publishing: 428]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
109.  Haneji N, Nakamura T, Takio K, Yanagi K, Higashiyama H, Saito I, Noji S, Sugino H, Hayashi Y. Identification of alpha-fodrin as a candidate autoantigen in primary Sjögren’s syndrome. Science. 1997;276:604-607.  [PubMed]  [DOI]  [Cited in This Article: ]
110.  Hahm KB, Im YH, Lee C, Parks WT, Bang YJ, Green JE, Kim SJ. Loss of TGF-beta signaling contributes to autoimmune pancreatitis. J Clin Invest. 2000;105:1057-1065.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 61]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
111.  Sakaguchi Y, Inaba M, Tsuda M, Quan GK, Omae M, Ando Y, Uchida K, Okazaki K, Ikehara S. The Wistar Bonn Kobori rat, a unique animal model for autoimmune pancreatitis with extrapancreatic exocrinopathy. Clin Exp Immunol. 2008;152:1-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
112.  Kountouras J, Zavos C, Chatzopoulos D. A concept on the role of Helicobacter pylori infection in autoimmune pancreatitis. J Cell Mol Med. 2005;9:196-207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 82]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
113.  Meagher C, Tang Q, Fife BT, Bour-Jordan H, Wu J, Pardoux C, Bi M, Melli K, Bluestone JA. Spontaneous development of a pancreatic exocrine disease in CD28-deficient NOD mice. J Immunol. 2008;180:7793-7803.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Haruta I, Yanagisawa N, Kawamura S, Furukawa T, Shimizu K, Kato H, Kobayashi M, Shiratori K, Yagi J. A mouse model of autoimmune pancreatitis with salivary gland involvement triggered by innate immunity via persistent exposure to avirulent bacteria. Lab Invest. 2010;90:1757-1769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
115.  Ammann RW. Diagnosis and management of chronic pancreatitis: current knowledge. Swiss Med Wkly. 2006;136:166-174.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Andrén-Sandberg A, Hoem D, Gislason H. Pain management in chronic pancreatitis. Eur J Gastroenterol Hepatol. 2002;14:957-970.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 77]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
117.  DiMagno EP. Toward understanding (and management) of painful chronic pancreatitis. Gastroenterology. 1999;116:1252-1257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 41]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
118.  Vera-Portocarrero LP, Lu Y, Westlund KN. Nociception in persistent pancreatitis in rats: effects of morphine and neuropeptide alterations. Anesthesiology. 2003;98:474-484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 65]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
119.  Takamido S, Kataoka Y, Tanano A, Cui Y, Ikeura T, Shimatani M, Kubota Y, Okazaki K, Yamada H. Intrapancreatic axonal hyperbranching of dorsal root ganglia neurons in chronic pancreatitis model rats and its relation to pancreatic pain. Pancreas. 2006;33:268-279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 9]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
120.  Wesselmann U, Lai J. Mechanisms of referred visceral pain: uterine inflammation in the adult virgin rat results in neurogenic plasma extravasation in the skin. Pain. 1997;73:309-317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 105]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
121.  Winston JH, He ZJ, Shenoy M, Xiao SY, Pasricha PJ. Molecular and behavioral changes in nociception in a novel rat model of chronic pancreatitis for the study of pain. Pain. 2005;117:214-222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 60]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
122.  Xu GY, Winston JH, Shenoy M, Yin H, Pendyala S, Pasricha PJ. Transient receptor potential vanilloid 1 mediates hyperalgesia and is up-regulated in rats with chronic pancreatitis. Gastroenterology. 2007;133:1282-1292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 106]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
123.  Zhang W, Gao J, Zhao T, Wei L, Wu W, Bai Y, Zou D, Li Z. Proteinase-activated receptor 2 mediates thermal hyperalgesia and is upregulated in a rat model of chronic pancreatitis. Pancreas. 2011;40:300-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
124.  Hughes MS, Shenoy M, Liu L, Colak T, Mehta K, Pasricha PJ. Brain-derived neurotrophic factor is upregulated in rats with chronic pancreatitis and mediates pain behavior. Pancreas. 2011;40:551-556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
125.  Friess H, Shrikhande S, Shrikhande M, Martignoni M, Kulli C, Zimmermann A, Kappeler A, Ramesh H, Büchler M. Neural alterations in surgical stage chronic pancreatitis are independent of the underlying aetiology. Gut. 2002;50:682-686.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 57]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
126.  Büchler M, Weihe E, Friess H, Malfertheiner P, Bockman E, Müller S, Nohr D, Beger HG. Changes in peptidergic innervation in chronic pancreatitis. Pancreas. 1992;7:183-192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 104]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
127.  Zhu ZW, Friess H, Wang L, Zimmermann A, Büchler MW. Brain-derived neurotrophic factor (BDNF) is upregulated and associated with pain in chronic pancreatitis. Dig Dis Sci. 2001;46:1633-1639.  [PubMed]  [DOI]  [Cited in This Article: ]
128.  Vera-Portocarrero LP, Xie JY, Kowal J, Ossipov MH, King T, Porreca F. Descending facilitation from the rostral ventromedial medulla maintains visceral pain in rats with experimental pancreatitis. Gastroenterology. 2006;130:2155-2164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 93]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
129.  Liu PY, Lu CL, Wang CC, Lee IH, Hsieh JC, Chen CC, Lee HF, Lin HC, Chang FY, Lee SD. Spinal microglia initiate and maintain hyperalgesia in a rat model of chronic pancreatitis. Gastroenterology. 2012;142:165-173.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
130.  Zhu Y, Colak T, Shenoy M, Liu L, Pai R, Li C, Mehta K, Pasricha PJ. Nerve growth factor modulates TRPV1 expression and function and mediates pain in chronic pancreatitis. Gastroenterology. 2011;141:370-377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 63]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
131.  Cattaruzza F, Johnson C, Leggit A, Grady E, Schenk AK, Cevikbas F, Cedron W, Bondada S, Kirkwood R, Malone B. Transient receptor potential ankyrin 1 mediates chronic pancreatitis pain in mice. Am J Physiol Gastrointest Liver Physiol. 2013;304:G1002-G1012.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 42]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
132.  Liao XZ, Zhou MT, Mao YF, Xu H, Chen H, Sun JH, Xiong YC. Analgesic effects of gabapentin on mechanical hypersensitivity in a rat model of chronic pancreatitis. Brain Res. 2010;1337:104-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]