You are on page 1of 13

Am J Physiol Gastrointest Liver Physiol 311: G343–G355, 2016.

First published July 14, 2016; doi:10.1152/ajpgi.00372.2015. Themes

Animal models of gastrointestinal and liver diseases. Animal models of acute


and chronic pancreatitis
Xianbao Zhan,1* Fan Wang,1* Yan Bi,2 and Baoan Ji1
1
Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida and 2Department of Gastroenterology and Hepatology,
Mayo Clinic, Jacksonville, Florida
Submitted 23 October 2015; accepted in final form 6 July 2016

Zhan X, Wang F, Bi Y, Ji B. Animal models of gastrointestinal and liver


diseases. Animal models of acute and chronic pancreatitis. Am J Physiol Gastro-
intest Liver Physiol 311: G343–G355, 2016. First published July 14, 2016;
doi:10.1152/ajpgi.00372.2015.—Animal models of pancreatitis are useful for elu-
cidating the pathogenesis of pancreatitis and developing and testing novel inter-
ventions. In this review, we aim to summarize the most commonly used animal
models, overview their pathophysiology, and discuss their strengths and limitations.
We will also briefly describe common animal study procedures and refer readers to
more detailed protocols in the literature. Although animal models include pigs,
dogs, opossums, and other animals, we will mainly focus on rodent models because
of their popularity. Autoimmune pancreatitis and genetically engineered animal
models will be reviewed elsewhere.
fibrosis; inflammation; pancreatic disorder

PANCREATITIS IS THE INFLAMMATORY disease of the pancreas that examination during the early phases of pancreatitis. However,
causes significant morbidity and mortality throughout the a variety of animal models have been developed to facilitate
world (148). There are two main types of the disease: acute our understanding of the pathophysiology of AP. In this re-
pancreatitis and chronic pancreatitis. view, we will discuss several commonly used experimental
models of AP with focus on mice and rats. Although these
Animal Models of Acute Pancreatitis animal models of pancreatitis are widely used, researchers
should keep in mind that no model fully recapitulates the
With ⬃275,000 hospitalizations in 2009 (an increase of human form of the disease. Therefore, understanding the
more than twofold since 1988) (177), acute pancreatitis (AP) is pathophysiology and limitations of each model will guide
the most common single gastrointestinal diagnosis, resulting in investigators in selecting the specific model for their special
an estimated 2.6 billion dollars per year in inpatient costs purposes. Other considerations important during model selec-
(129). AP also ranks as the fifth leading cause of in-hospital tion include the cost, ease of use, reproducibility, disease
deaths (85). Unfortunately, there are no effective preventive or
severity, and clinically relevance.
therapeutic strategies for this disease due to the lack of a deep
It should also be noted that species, sex, and age can also
understanding of its pathogenesis (80).
affect the severity of pancreatitis. Therefore, age- and sex-
In the United States, alcohol abuse and gallstone disease
account for 70 – 80% of the cases of AP. Drugs, postendo- matched controls should be used to offset those potential
scopic retrograde cholangiopancreatography (ERCP) proce- influences. As for species differences, the cholecystokinin
dures, infections, and lipid metabolic disorders can also cause (CCK) analog JMV-180 acts as a partial agonist in rats but a
AP (127). Eighty-five to 90% of AP cases are self-limiting and full agonist in mice (72). Unlike rodent pancreatic acinar cells,
respond well to conservative treatment. However, the remain- human counterparts don’t respond to CCK stimulation (71).
ing 10 –15% of cases with severe acute pancreatitis (SAP) are Age-dependent effects on experimental AP in mice have also
accompanied by local or systemic complications and organ been observed (113). Male rats fed with high-fat diet have
failure and have a grave clinical course. Although the overall shown marked, lower pancreas Mn-superoxide dismutase ac-
mortality in patients with AP is ⬃5%, the mortality of SAP is tivity and higher oxidative damage than females (50). These
between 25 and 50% (13). observations support the role of male hormones in the regula-
The pathophysiology of this disease is not well understood, tion of oxidative stress and pancreatitis. The evidence for
and most of our current knowledge about the pathophysiology female hormones in the development of pancreatitis is even
of this disease is from animal studies. This is partially due to more extensive. The choline-deficient, ethionine-supplemented
the relative inaccessibility of human pancreatic tissue for (CDE) diet induces acute hemorrhagic pancreatic necrosis only
in young female mice but not in young male mice (137).
The interplay between inflammation and the microbiome
* X. Zhan and F. Wang contributed equally to this work.
Address for reprint requests and other correspondence: B. Ji, Dept. of
suggests that housing conditions can affect the severity and
Cancer Biology, Griffin Bldg. 311, Mayo Clinic, 4500 San Pablo Rd S., natural history of pancreatitis (181). The composition of the
Jacksonville, FL 32224 (e-mail: ji.baoan@mayo.edu). model’s diet can also affect inflammatory signaling (133).
http://www.ajpgi.org 0193-1857/16 Copyright © 2016 the American Physiological Society G343
Downloaded from journals.physiology.org/journal/ajpgi at Univ De Antioquia (200.024.017.012) on February 21, 2021.
Themes
G344 PANCREATITIS MODELS

Analgesics can also interfere with pancreatitis-associated in- creatitis induced by cholinergic agonists has never been widely
flammation; indomethacin prevents postendoscopic retrograde used in experimental settings.
cholangiopancreatography pancreatitis (36). In contrast, opi- Caerulein (ceruletide, also spelled as cerulein) and its struc-
ates may be a cause of pancreatitis (159). Although it has been ture were identified in 1967 by Australian and Italian scientists
shown that caerulein-induced AP is associated with pain in from dried skins of the Australian green tree frog (Litoria
experimental animals, most experiments are carried out with- caerulea) (8). Sequencing of the peptide showed a strong
out any pain-relieving treatment. However, in a recent study, resemblance in the amino acid sequence to the carboxy termi-
orally administered metamizol showed no influence on the nus of CCK (CCK octapeptide: Asp-Tyr[SO3H]-Met-Gly-Trp-
caerulein-induced AP and could be given as an analgesic to Met-Asp-Phe-NH2; caerulein: pyroGlu-Gln-Asp-Tyr[SO3H]-
increase animal welfare in experimental models with induced Thr-Gly-Trp-Met-Asp-Phe-NH2). The seven amino acids of
AP (150). the COOH-terminal of caerulein are identical to those of the
AP is an inflammatory disease of the pancreas. The damage CCK octapeptide, except for a Thr residue that is a substitute
to the pancreas can be evaluated by measuring the serum for Met. Both peptides bear a carboxy terminal amide group,
pancreatic enzymes, such as amylase and lipase, that are but caerulein also has a blocked amino terminal with pyroglu-
released into the blood from the damaged acinar cells. There tamine as the initial amino acid. This blocking group reduces
are commercially kits available that can measure these en- susceptibility to inactivation by amino peptidases (173). Addi-
zymes. However, the levels of these enzymes do not necessar- tionally, sulfation of the tyrosine in caerulein is necessary for
ily reflect the severity of pancreatitis. A histological examina- optimal physiological activity (76). In rodents, CCK receptors
tion by an experienced pathologist in a blinded manner is are present both on pancreatic acinar cells and within the neural
highly recommended to assess interstitial edema, acinar cell system. Physiological levels of CCK in plasma act via stimu-
death (apoptosis, necrosis, and autophagy), parenchymal loss, lation of the vagal afferent pathways. In contrast, supraphysi-
hemorrhage, fat necrosis, inflammatory cell infiltration, and fat ological plasma CCK levels act on intrapancreatic neurons and
and fibrotic tissue replacement. However, no consensus is act directly to a larger extent on rodent pancreatic acini (124).
available regarding the criteria for scoring, and the parameters Caerulein is a potent pancreatic secretion stimulant. The
used by different investigators to evaluate the severity of AP dosages used for secretion studies are 1–5 ng/kg through
have yet to be standardized, but most of these parameters are rapid intravenous injection, 0.25–1 ng·kg⫺1·min⫺1 through
described in detail by Nevalainen et al. (118) and Zhang and intravenous infusion, and 50 –100 ng/kg through subcutane-
Rouse (182). Additionally, pancreatic edema can be evaluated ous injection (16, 33). At higher doses (20 ng·kg⫺1·min⫺1),
by whole pancreas/body weight (body wt) ratio, wet/dry pan- caerulein causes zymogen activation, large vacuole accumula-
creas ratio, or (wet⫺dry)/dry weight ratio. tion, endoplasmic reticulum (ER) dilation and segmentation,
Systematic complications involving the lung are also asso- and mitochondrial swelling (155). In 1977, Lampel and Kern
ciated with the severity of the disease. Therefore, the histology (84) discovered that excessive doses (5 ␮g·kg⫺1·h⫺1) of caeru-
of the lung is often included in pancreatitis research. For lein-induced acute interstitial pancreatitis in rats.
studies focusing on the acute respiratory distress syndrome Supramaximal doses of caerulein induce a significant in-
(ARDS) in severe AP, detailed measurements of histological crease in serum pancreatic enzymes, pancreatic interstitial
changes in parenchymal tissue, altered integrity of the alveolar edema, and inflammatory cell infiltration. It has been tested in
capillary barrier, inflammation, and abnormal pulmonary rats, mice, dogs, and hamsters, and all of the animals survived
function should be included. The analytical approaches, the induction of pancreatitis (172). The changes caused by
pathological features, and common measurements have been caerulein infusion occur within 15 min after infusion. Cyto-
described in a recent review (3). In severe models of AP, plasmic vacuoles are the earliest histological alterations. As the
liver/kidney malfunction can also be monitored and has pancreatitis progresses, these vacuoles increase to an enormous
been described (6, 12). size. Electron microscopy showed both intact and degenerative
Finally, cytokine expression levels in the pancreatic tissue granules inside the vacuoles. Interstitial inflammation and
and serum can also be used to reflect the severity of local and acinar cell necrosis were prominent 6 h after injection and
systematic inflammation. It is worth emphasizing that the reached a maximum after 12 h. These changes usually resolved
severity of inflammation and damage to the pancreas is not within a week. During the regression of pancreatitis, focal
evenly distributed. Using a large piece of tissue will decrease atrophy was a remarkable histological finding (119).
sampling error. The mechanisms of secretagogue-induced AP have been
Secretagogue-induced AP. In 1895, Mouret (111a) reported extensively exploited over the past few decades. For in vitro
that excessive cholinergic stimulation was associated with the studies, primary pancreatic acini can be isolated from mouse
development of pancreatic acinar cell vacuolization and necro- and rat pancreata by mechanical dissection followed by puri-
sis. Since then, pancreatitis has been reported as a complication fied collagenase digestion (174). These pancreatic acini can
of anticholinesterase insecticide intoxication in humans and then be treated with CCK or other secretagogues. CCK regu-
experimental animals (34, 44). Cholinergic nerve mediation of lates a complex array of cellular functions in pancreatic acinar
low-dose caerulein- and alcohol-induced experimental pancre- cells through its G protein-coupled receptor, which activates a
atitis has also been documented (102), and, because it stimu- variety of intracellular signaling mechanisms. CCK couples
lates the release of acetylcholine from pancreatic nerves, scor- through heterotrimeric G proteins to activate phospholipase C,
pion venom has been found to induce AP in humans and dogs increasing inositol trisphosphate and releasing intracellular
(14, 46, 104). Similarly, carbachol (a cholinergic mimetic) Ca2⫹. This pathway and protein kinase C activation leads to
administration to rats produces a form of edematous pancre- the secretion of digestive enzymes by exocytosis. CCK also
atitis (21, 38). However, due to systematic toxicity, the pan- activates Nuclear Factor of Activated T cells (NFATs), ERKs,

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00372.2015 • www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi at Univ De Antioquia (200.024.017.012) on February 21, 2021.
Themes
PANCREATITIS MODELS G345
JNKs, and p38 MAPK, which are involved in secretion and aliquot powder, and the solubility of caerulein from different
growth (58, 175). In addition to Gq, CCK receptors also providers may vary. If the powder does not completely dis-
activate G12/13, Rho, and Rac to regulate the cytoskeleton and solve, then it will result in a low concentration of caerulein and
secretion (17, 88). may fail to induce pancreatitis.
CCK elicits a concentration-dependent effect on pancreatic The secretagogue overstimulation model of experimental
enzyme secretion. At low physiological concentrations, CCK pancreatitis is so far the most commonly used AP model. The
stimulates pancreatic acinar cell secretion. However, at patho- model has several advantages, including noninvasiveness, high
physiological concentrations, the secretion of the pancreas is reproducibility, and applicability in multiple species. In addi-
inhibited due to the altered influx of intracellular calcium, tion, the in vivo experiment can be paralleled with in vitro
diminished pancreatic enzyme secretion into the duct, patho- acinar cell studies, which makes this model extremely useful
logical basal-lateral secretion/leakage of active enzymes, pre- for the investigation of intracellular signal transduction events,
mature intracellular zymogen activation, actin cytoskeleton protease activation cascades, and cell death pathways involved
reorganization, ER stress, activation of NF-␬B and apoptosis, in the early phase of AP. Moreover, this is a great model for the
and autophagy. Increased vascular permeability and increased investigation of recovery and regeneration of damaged tissue
hydrostatic pressure may contribute to the extensive edema after the toxic substances have been discontinued. However,
associated with pancreatitis. Cytokine storms and active pan- this model has several major disadvantages. Even with maxi-
creatic enzymes lead to a systemic inflammatory response mum dosage of caerulein, only mild, self-limited AP but not
syndrome (SIRS), which includes extrapancreatic damage, severe necrotizing pancreatitis is recapitulated, which limits its
such as pancreatitis-related lung injury (18, 73, 98, 103, 112, use in the study of severe pancreatitis, which carries the highest
142, 157). morbidity and mortality. In addition, secretagogue overstimu-
In addition to the direct effect of CCK on acinar cells, CCK lation is not a common cause of AP in humans, although AP
also acts via the vagal afferent pathways to mediate pancreatic cases have been reported in patients who accidentally received
secretion. Atropine, a muscarinic receptor antagonist, com- high concentrations of secretagogue (14), were exposed to
pletely abolishes pancreatic enzyme responses to physiological cholinesterase inhibitors such as insecticides (149), or were
doses of CCK in rats, which suggests that CCK acts on a exposed to Trinidadian scorpion toxin (14). Moreover, major
presynaptic site along the cholinergic pathway (95). In healthy differences exist between the human and rodent pancreas: CCK
human subjects, CCK infusions produce plasma CCK levels plays a major role in regulating exocrine pancreatic secretion in
similar to those seen postprandially and stimulate pancreatic rodents; however, human pancreatic acinar cells are mostly
secretion by an atropine-sensitive pathway. These observations regulated by cholinergic pathways that involve neurogenic
suggest that both in experimental animals and in humans CCK stimulation (71, 124).
cholinergic neural pathways, rather than pancreatic acini, rep- Basic amino acid-induced AP. In an effort to study the
resent the primary targets in which CCK acts to stimulate effects of arginine on normal tissues, Mizunuma et al. (110)
pancreatic enzyme secretion (2). Cholinergic stimulation of unexpectedly found that single intraperitoneal injection of
pancreatic amylase secretion is mediated in mice by a mixture L-arginine (500 mg/100 g body wt) in rats selectively destroyed
of M1 and M3 muscarinic acetylcholine receptors. Like CCK pancreatic acinar cells. The model was further studied by Tani
receptors, these receptors are selectively coupled to G proteins et al. (154) in 1990. From then on, several groups have used
of the Gq family, which mediate the breakdown of phosphati- this rat model. However, with this protocol the same dose of
dyl inositol lipids (47). Similarly, muscarinic-receptor agonists L-arginine failed to induce reproducible pancreatitis in either
stimulate trypsinogen and NF-␬B activation, two key signaling Balb/c or C57Bl/6 mice. In 2007, Saluja and colleagues (29)
pathways in the pathogenesis of pancreatitis. Interestingly, developed a mouse model of acute necrotizing pancreatitis
ethanol enhances carbachol-induced protease activation and using intraperitoneal applications of L-arginine hydrochloride
accelerates Ca2⫹ waves in isolated rat pancreatic acini. These solution (8% in normal saline, pH 7.0).
observations support the key roles of the cholinergic system in The dosage of L-arginine on AP development is species
the mechanisms of alcoholic pancreatitis (59, 102, 123, 185). dependent. In mice, L-arginine has no effect on pancreatic
The cholinergic signaling may be of particular importance in histology when administered at doses lower than 4 g/kg and
humans because there are no functional CCK receptors on when administered twice (1 h apart). However, in rats, intra-
human pancreatic acinar cells (71). peritoneal administration of 2 g/kg L-lysine induced severe
There are great variations among the protocols used in acute necrotizing pancreatitis (20).
different laboratories. Although jugular or tail vein infusion In rats given single intraperitoneal injections of L-arginine at
was originally used, it was soon replaced by intraperitoneal 500 mg/100 g body wt, serum levels of amylase, lipase, and
injection due to its convenience (119). Typically, caerulein (50 anionic trypsin reached their peaks after 12–24 h but returned
␮g/kg body wt; dissolved in phosphate-buffered saline or 0.9% to normal levels after 24 – 48 h. Histological examinations
saline in a volume of 100 ␮l) is injected intraperitoneally every revealed a number of small vesicles within acinar cells after 6
hour for 8 –12 injections. The pancreata are usually harvested h, which were identified as markedly swollen mitochondria by
1 h after the last injection or 24 h after the first injection. For electron microscopy. Interstitial edema appeared after 12 h,
regeneration studies, the pancreata can be collected after 2–7 and acinar cell necrosis was seen after 24 h. The extent and
days. For early signaling studies, the pancreata can be removed severity of necrotic changes of pancreatic exocrine tissue with
at a desired time after a single dose of injected caerulein. inflammatory cell infiltration was maximal after 72 h. After 7
Caerulein induces parallel activation of both NF-␬B and days without treatment, pancreatic acinar cells began to regen-
trypsinogen in the pancreas in vivo as early as 15 min after erate, and pancreatic architecture appeared almost normal after
injection (64). Of note, caerulein is usually provided in 1-mg 14 days without treatment (154). In this model, pancreatic

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00372.2015 • www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi at Univ De Antioquia (200.024.017.012) on February 21, 2021.
Themes
G346 PANCREATITIS MODELS

necrosis was significantly increased in a concentration-depen- atrophy with acinar cell regeneration. In 1975, Lombardi and
dent manner and was accompanied by lung injury (29, 154, colleagues (100, 138) reported that female mice fed with the
169). CDE diet (a choline-deficient diet enriched with 0.5% ethio-
In the mouse model, the mice become sluggish after admin- nine, a derivative of methionine) developed severe necrotizing
istration of the first dose of L-arginine (4 g/kg) in saline, and pancreatitis and had a mortality rate of 100% after 4 days.
their lethargy increases with the administration of the second The onset of CDE diet-induced AP is variable, but it usually
dose. The administration of L-arginine results in significantly takes ⬃2–3 days to develop. This form of pancreatitis is
increased plasma amylase levels after 48 h and normalizes at characterized by massive fat necrosis of the exocrine paren-
96 h. Histologically, acinar cell vacuoles appear as early as 6 chyma, intense hemorrhaging, and an inflammatory reaction of
h after treatment. Twenty-four hours after injection, zymogen the stroma throughout the peritoneal cavity (99, 100, 120, 138).
granules disappear in some areas. After 72 h, there is an The earliest changes are increased zymogen granules in pan-
accumulation of interstitial fluid and necrosis of acinar cells. creatic acinar cells. Pancreatic inflammation and necrosis occur
After 96 and 120 h, the majority of the pancreas is replaced by before activated proteolytic enzymes can be detected within the
inflammatory and fibrotic cells, and only small areas of intact pancreas. The necrosis of acinar cells develops 48 h after
acinar cells are visible. However, pancreatic islet cells remain treatment, and hemorrhaging starts 60 h after initiation of the
unaffected by L-arginine treatment (29). diet. In the animals who survived the AP, their pancreatic
Although the exact mechanisms of L-arginine pancreatitis function started to recover after 2–3 wk, and morphological
are not clear, the imbalance of amino acids and the subsequent resolution usually took 4 – 6 wk.
decrease of protein synthesis, the derangement of transamina- The exact mechanisms of CDE on AP are unclear. However,
tion and the urea cycle, and metabolic acidosis in the acinar the obvious sex differences in this model suggest that estrogen
cells may all be involved. L-Arginine metabolites are also may play a role in the pathogenesis of AP and have been
involved in pancreatitis development as high doses of L-orni- evidenced by the fact that male mice pretreated with estrogen
thine, a downstream product of L-arginine by L-arginase, have can also develop AP on CDE diet. Additionally, zymogen-
also been reported to cause AP (136). Partial inhibition of granule secretion blockage and intraparenchymal trypsinogen
L-arginase ameliorates L-arginine-induced pancreatitis (19). activation can be a result of a synergistic action of choline
Oxidative stress may also contribute to L-arginine pancreatitis deficiency with the basic toxicity of ethionine toward the acinar
because reducing oxidative stress ameliorates pancreatic injury cells. Ethionine may be toxic to the pancreas by interfering
in this model of pancreatitis (61, 62). Additionally, hyperse- with RNA, protein, and phospholipid metabolism (100). Au-
cretion from the significant enhancement of neural sympathetic tophagy and crinophagy have also been suggested to be in-
and parasympathetic activity (50 mg) (61) and nitric oxide volved in the vacuole formation.
(152) may also be involved in the development of this disease The severity of the CDE AP model will vary depending on
(151, 153). These effects seem to be L-arginine specific as the sex, age, and weight of the animals. Young, female mice
D-arginine, L-lysine, L-alanine, and glycine failed to elicit AP. are prone to have more severe cases compared with older, male
The L-arginine model has been modified to a single injection mice. Generally, young female CD-1 mice (or NMR1, or Swiss
with varying results. Doses higher than 500 mg/100 g body wt Webster mice; 4 – 6 wk old, 10 –14 g) are used for the CDE AP
can induce death within a few hours (154). A single dose of model. Typically there are two approaches to generate this
500 mg/100 g causes necrosis in up to 70 – 80% of the pancre- model. In one approach, the animals are fed regularly with
atic acinar cells within 3 days, which increases to 90% after regular chow and then switched to the CDE diet. The mortality
three further doses over 10 days (154). Splitting the dose to two of the animals depends on the length of time the animal is on
injections can also be used to delay the onset of AP (169). the CDE diet: 33 h renders 10 –20% mortality, 48 h increases
Single injection carries a higher mortality than double injec- the mortality to 40 –70%, 66 h is associated with 55–75%
tion, and therefore double injection models are more com- mortality, and if the diet is administered for 72 h, then there
monly used. In brief, a sterile L-arginine solution (8% in PBS will be a mortality of over 80%. Another approach involves
or normal saline, pH 7) is administered intraperitoneally to animal starvation for 1 day followed by feeding with the CDE
nonfasted mice at a dose of 400 mg/100 g body wt or to rats at diet at 3 g per mouse per day for 1–5 days depending on the
a dose of 250 mg/100 g body wt. One hour later, a second dose desired severity of AP. In this approach, the animals tend to eat
of L-arginine is administered. more of the CDE diet during the first 24 h and develop AP
L-Arginine-induced AP provides a model for necrotizing faster because they have been starved for 24 h (120).
pancreatitis that carries a high morbidity and mortality. It is This model is a noninvasive AP model. Despite the differ-
highly reproducible, and the severity of pancreatic acinar ences in pathogenesis of the pancreatitis induced in this model
necrosis can be controlled by regulating the dose and time of compared with the human disease, the gross and histological
L-arginine. However, it is unlikely that human AP is caused by appearance of the pancreatic and peripancreatic inflammation
basic amino acid overdose. and the clinical and biochemical course of the diet-induced
CDE diet-induced AP. Pancreatic damage caused by low- pancreatitis resemble the human disease. Both the model and
choline diets was originally reported in 1937 (54). In 1950, the human disease share several pathophysiological features,
ethionine was first reported to induce AP in rats by Farber and including necrosis, systemic hypoxia, and hemoconcentration.
Popper (40) and Goldberg et al. (49). Since then, ethionine- Therefore, the model may be suitable for studying these patho-
induced pancreatitis has been shown to occur in many other physiological aspects of this disease. The severity and mortal-
species of experimental animals, including mice, hamsters, ity of the model can be modified by changing the duration of
cats, dogs, and monkeys (22, 30, 166). Ethionine-induced CDE diet administration; for example, all of the mice fed with
pancreatitis is characterized mainly by local necrosis and the CDE diet for 5 days died of hemorrhagic pancreatitis;

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00372.2015 • www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi at Univ De Antioquia (200.024.017.012) on February 21, 2021.
Themes
PANCREATITIS MODELS G347
however, when the CDE diet was given for only 1 day, the despite the marked lethality associated with the 5% sodium
incidence of fatal hemorrhagic pancreatitis was reduced to taurocholate treatment, no obvious pulmonary pathology was
55– 65% (120). detected after 24 h postinduction (176).
Several pitfalls and problems have to be considered to obtain Bile salts exert detergent properties and primarily aid in the
valuable data. Due to the mortality associated with the amount absorption of fat and fat-soluble vitamins (65). It was initially
of the CDE diet ingested, the CDE diet implementation re- believed that pancreatic injury resulted from the detergent
quires careful monitoring of dietary intake and animal status. It properties of bile acids. Currently, bile acids are considered
is also important to use large numbers of animals in each crucial agonists that interact with several proteins, including
experimental group. Young mice are affected more severely the nuclear receptor farnesoid X receptor (FXR) and G protein-
than adult mice and females more than males (101). Thereby coupled bile acid receptor-1 (Gpbar1) to regulate many cellular
age- and sex-matched animals should be used for homogeneity functions (39, 48). Bile acids can act on both duct cells and
and reproducibility of CDE diet-induced AP. Another point to acinar cells of the exocrine pancreas. Bile acids have dual
remember is that liver damage and hypoglycemia are also effects on the ductal secretion, and at low concentrations they
observed in this model. Therefore, this model is not ideal for induce a dose-dependent elevation of intracellular Ca2⫹ con-
studying multiple organ distress syndromes because the CDE centration via an inositol 1,4,5-triphosphate receptor and a
diet can trigger those syndromes by mechanisms that are phospholipase C-mediated pathway. This may help wash out
unrelated to the severity of AP. The high cost of the diet also the toxic bile acids and thus protect the acinar cells. At high
discourages the use of this model (120). Because of these concentrations, bile acids induce a toxic, sustained intracellular
limitations, this model has been rarely used in recent years. Ca2⫹ concentration and decrease intracellular ATP, which
Retrograde ductal infusion-induced AP. Biliary pancreatitis inhibits all of the acid-base transporters. Pathological Ca2⫹-
is one of the most common forms of AP. One theory is that dependent trypsinogen and calcineurin activation causes cell
biliary obstruction permits the reflux of bile into the pancreatic death in isolated pancreatic acinar cells (112). Loss of the flux
duct. The first experimental, retrograde ductal infusion-induced defense mechanism may also lead to high concentrations of
AP model was established in 1856 by Bernard (15), who bile acids to reach the acinar cells. These effects at least in part
injected bile and olive oil into a canine pancreas through the are mediated by Gpbar1. Genetic deletion of Gpbar1 signifi-
ampulla of Vater. Since then, various bile salts have been cantly reduces biliary but not secretagogue-induced experi-
reported to induce AP in different species. Ductal infusion of mental AP, suggesting its role in the pathophysiology of the
sodium glycodeoxycholic dose dependently induces edema- biliary pancreatitis (130).
tous pancreatitis and necrotizing pancreatitis in rats (156). An One of the most commonly used retrograde ductal-infusion
infusion containing a combination of enterokinase with sodium protocols in rats was described by Aho et al. (5). First, a
glycodeoxycholic induced necrotic pancreatitis with systemic midline laparotomy is performed, the biliopancreatic duct is
complications and rapid mortality (156). In 1992, Schmidt et then cannulated, and fluid (saline or saline containing bile acid
al. (145) modified the rat duct infusion model by using the and blue dye) is infused. Flow rates higher than 10 ␮l/min
combined actions of very low concentrations of glycodeoxy- should be avoided to minimize any artifacts or injury due to the
cholic acid (5–10 mM) administered via ductal infusion and rapid increase in intrapancreatic pressure. Detailed methods for
caerulein (5 ␮g·kg⫺1·h⫺1 for 6 h) administered intravenously. experimental acute biliary pancreatitis in mice induced by
This model features a moderate onset of moderate injury retrograde infusion of bile acids have been published by
throughout the pancreas that lasts 24 h and provides the Perides et al. (132).
potential for modulating disease severity (41). Recently, Lauk- Biliary AP accounts for 30 –50% of all clinical cases of AP.
karinen et al. (87) established a retrograde ductal infusion- This model uses clinically relevant etiological insults of acute
induced AP model in mice by infusing sodium taurocholate biliary pancreatitis, and its severity can be manipulated by
into the mouse pancreatic duct. In the head of the pancreas, altering the concentration, volume, and infusion pressure of the
evidence of pancreatitis was observed 12–24 h after infusion of injected bile acid. This model is useful for mechanistic studies
20 –50 ␮l of 2–5% sodium taurocholate. The damage in the employing genetically modified mouse strains. The disadvan-
pancreas was concentration and volume dependent. There was tage is that it is technically challenging to control the constant
no lethality found. In contrast, in a separate study using mice pressure when retrograding ductal infusion, which is critical
(176), 2–5% sodium taurocholate solution was injected at a for determining the severity of the induced pancreatitis. Pumps
volume of 2 ml/kg (⬃50 ␮l/mouse). All of the animals receiv- may be needed to control the constant low pressure of infusion
ing 2 or 3% sodium taurocholate survived the experiment. to produce a standard degree of pancreatic injury and to avoid
Animals receiving 4 and 5% sodium taurocholate showed a high perfusion pressure-induced artifact (87).
significant increase of fatty tissue necrosis when compared Recently, Husain and coworkers (75) developed another
with animals receiving the 2% solution. Death within the first clinically relevant mouse model of AP. In this model, the
24 h occurred in 10% of the animals receiving the 4% solution. radiocontrast agent iohexol was injected to the common bile
And death rate reached 60% when 5% taurocholate solution duct and pancreatic duct of Swiss Webster mice by retrograde
was used. infusion. Iohexol infusion causes acute damage and inflamma-
Histological changes of the pancreas after 24 h of treatment tion in the pancreas. Mechanistically, radiocontrast agents
include large areas of acinar cell necrosis adjacent to morpho- cause a pancreatic inflammatory response through the activa-
logically unaltered acinar lobes. Leukocyte infiltration, fatty- tion of NF-␬B, calcium signaling, and calcineurin pathways.
tissue necrosis, and hemorrhage are typical. Taurocholate in- These signaling pathways seem to be pancreatic acinar cell
fusion predominantly affects the pancreatic head. The pancre- specific because they were not elicited in HEK293 or COS7
atic tail usually presents with extensive edema. Surprisingly, cells. Calcineurin deficiency or inhibitors improved the pan-

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00372.2015 • www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi at Univ De Antioquia (200.024.017.012) on February 21, 2021.
Themes
G348 PANCREATITIS MODELS

creatitis associated with this model, suggesting that targeting tis in animals suggests that the provision of ethanol may only
these pathways may prevent post-ERCP pancreatitis in patients increase the predisposition to pancreatitis. In 1999, Pandol et
(75). al. (126) found that an ethanol diet sensitized rats to the
Duct-ligation induced AP. This model is based on the pancreatitis caused by CCK-8. After intragastric feeding with
theory that duct obstruction will block pancreatic fluid and either a control or an ethanol diet for 2 or 6 wk, rats were then
enzyme flow and therefore lead to AP development. Initially infused for 6 h with either saline or CCK-8 at a dose of 3,000
attempted in the early 19th century (81), duct ligation has pmol·kg⫺1·h⫺1, which by itself did not induce pancreatitis. All
been used in various animals including dogs, rabbits, opos- measures of pancreatitis, as well as NF-␬B activity and cyto-
sums, rats, and mice. Opossums have a pancreatic main duct kines were significantly increased only in rats treated with
that drains into a long common bile duct, and both enter the ethanol plus CCK-8 (126). In a later study, rats were fed with
duodenum at the papilla. The extraduodenal common bile control and ethanol-containing Lieber-DeCarli diets (discussed
duct and the pancreatic duct are easily dissected in this in detail below: Alcohol-related CP models) for 6 wk followed
animal and as a result make it easier to study the effects of by four hourly intraperitoneal injections of caerulein at 0.5
ligation of specific ducts on the pathogenesis of AP. Liga- ␮g/kg. Caerulein alone induced only minor pathological
tion of the pancreas duct or common bile duct in opossum changes in control-fed rats. In contrast, in ethanol-fed rats,
induced hemorrhagic AP with a 14-day mortality of 100% caerulein induced marked acinar cell vacuolization, dilation of
(147). However, most other laboratory animals do not de- the ER, and occasional patchy cellular necrosis (102).
velop AP with surgical ligation of the pancreatic duct alone, Both oxidative and nonoxidative metabolisms of ethanol
but long-term ligation causes chronic atrophy of the exo- (OME and NOME, respectively) mechanisms are used to
crine pancreas (122, 167). In mice, the combination of bile degrade alcohol in the exocrine pancreas. NOME combines
infusion into the pancreas followed by bile duct ligation also ethanol with fatty acids to yield lipophilic fatty acid ethyl esters
causes a more severe, necrotizing pancreatitis (89). A sim- (FAEEs) via diverse FAEE synthase enzymes (55). FAEEs can
ilar level of severity seen after obstructing the pancreatic induce high sustained toxic calcium concentrations in pancre-
duct adjacent to duodenum (which also blocks the bile duct) atic acinar cells and lead to acinar necrosis (27). To test the
compared with ligation of both pancreatic duct and bile duct effects of FAEE in the initiation of AP, adult CD1 mice
adjacent to liver suggest that bile reflux may not be neces- received two intraperitoneal injections of ethanol (1.35 g/kg)
sary to induce AP (92). In another study in mice, the and POA (palmitoleic acid, a monounsaturated fatty acid, 150
pancreatic duct was blocked by placing a small metal clip mg/kg) at 1-h intervals. Two hundred microliters of normal
across the lower end of the duct. A suture was incorporated saline were injected immediately prior to ethanol/POA injec-
within the clip during its placement for reversing biliary tions to avoid local damage by ethanol to peritoneal organs at
obstruction as needed (82). Combining duct ligation with the injection site. Mice received either saline, ethanol (1.35
both secretory stimulation or minimized arterial blood pro- g/kg), or POA (150 mg/kg) were used as controls (67). At 24
duced a more severe form of AP (135). h after application, the combination of ethanol and POA
Pancreatic duct obstruction rapidly changes the physio- induced pancreatic damages including extensive acinar cell
logical response of the exocrine pancreas to a calcium edema, neutrophil infiltration, and necrosis. The combination
signaling pattern that has been associated with premature of ethanol/POA also elicited alveolar membrane thickening
digestive enzyme activation and the onset of pancreatitis and inflammatory cell infiltration in the lung but damages in
(111). It is postulated that intrapancreatic digestive enzyme the liver, kidney, or heart are minimal (67). A similar pheno-
activation accounts for the major pathology of this model. type was achieved with the use of C57BL/6J mice (170). Mice
Altered intracellular targeting of endocytosed proteases are more tolerant to the treatment after one instead of two doses
might be one mechanism by which digestive zymogens of treatment (107).
reach an intracellular compartment where premature activa- Coxsackie B virus-induced AP. A wide variety of infections
tion can occur (93). have been associated with AP. These infections include viruses
The duct ligation-induced model closely mimics gallstone- (mumps, coxsackie, hepatitis B, cytomegalovirus, varicella-
induced AP (1). It avoids the use of agents at nonphysiologi- zoster virus, herpes simplex virus), bacteria (Mycoplasma,
cally relevant concentration and dosage that could produce Legionella, Leptospira, Salmonella), fungi (Aspergillus), and
unwanted systemic effects. However, the complexity, technical parasites (Toxoplasma, Cryptosporidium, Ascaris) (128).
difficulty, and limited reproducibility have made the duct Among these factors, coxsackie B virus-induced AP has been
ligation model less popular for investigating AP. Duodenal well documented and studied (24, 60, 139).
wall necrosis and pancreatic peritoneal sepsis often add com- Coxsackie virus-type B virus is a single-stranded RNA
plexity to the model. Furthermore, the severity of AP varies picornavirus with six identified serotypes (serotypes 1– 6).
depending on the species and duration of obstruction (23, 122). Mice infected with coxsackie viruses B1, B3, B4, or B5
There are also species-dependent effects in this model. In produce a severe form of pancreatitis consisting of the degen-
duct-ligated rats, apoptosis is the main mechanism of cell eration of acinar cells, loss of zymogen granules, infiltration of
death. By contrast, necrosis is the predominant mechanism in mononuclear and plasma cells, and the replacement of exocrine
duct-ligated opossums (56, 79). tissue with fatty tissue. In contrast, coxsackie viruses B2 and
Alcohol-related AP. Although alcoholism is one of the major B6 do not cause these changes. Coxsackie viruses B3 and B4
causes of human pancreatitis in the United States, a continuous exhibit more rapid action in the tissue and more severe lesions
intragastric infusion of ethanol to rats over several weeks than B1 and B5. In contrast, none of the coxsackie B viruses
produced only ethanol-induced liver injury, not alcohol-in- examined elicited detectable microscopic changes in the islets
duced pancreatitis. The failure to produce alcoholic pancreati- of Langerhans (86). In newborn mice infected with coxsackie

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00372.2015 • www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi at Univ De Antioquia (200.024.017.012) on February 21, 2021.
Themes
PANCREATITIS MODELS G349
B4 virus by intraperitoneal inoculation for 1–2 days, cytone- trypsin activity is the rapid induction of acinar cell death via
crosis consistent with a picornaviral infection was observed apoptosis and causes fat replacement in the pancreas (45).
(60). Mice with coxsackievirus B3 infection developed necro- Pancreatic fibrosis is a histopathological feature of CP. It is
tizing AP. This form of pancreatitis eventually led to complete now recognized that pancreatic fibrosis is mediated by the
atrophy of the exocrine pancreas with the islets of Langerhans activation of pancreatic stellate cells (PSCs) that normally
and pancreatic ducts spared (165). These mice also developed reside in the periacinar region of the pancreas in a quiescent
focal myocarditis (51). The viral infection also interacted with state (9). The mechanisms of PSC activation have been studied
other modifying factors. For example, it was observed that and reviewed previously (11). It seems that fibrosis is an active
infection of the virulent coxsackievirus B3 strain 28 in com- process mediated by the activation of PSCs. Acinar cell apo-
bination with alcohol feeding resulted in a more severe form of ptosis that is induced by intracellular trypsin caused extensive
pancreatitis. Furthermore, infection with the avirulent strain fat replacement (45). In contrast, Ras-induced acinar cell se-
(GA) plus alcohol feeding caused severe pancreatitis, whereas nescence led to a dramatic desmoplastic reaction in the pan-
the infection alone did not result in any obvious pathological creas (74).
effects in the pancreas (70). Alcohol also impaired pancreatic In humans, acute and recurrent AP can develop into CP.
regeneration following viral-induced injury in a dose- and These diseases share similar genetic and environmental caus-
duration-dependent fashion (25). ative factors. Therefore, AP, recurrent AP, and CP are now
considered as a disease continuum (171). Indeed, many re-
searchers have tried to establish CP models by using modified
Animal Models of Chronic Pancreatitis
experimental protocols that cause AP.
Progressive inflammation of the pancreas leads to chronic Models based on repetitive induction of AP. In caerulein-
pancreatitis (CP), which is defined by the loss of parenchymal induced AP, the pancreas will recover from a single episode of
(exocrine and endocrine) cells, inflammatory cell infiltration, caerulein challenge within a week. Expression of TGF-␤1, a
fat replacement, stellate cell activation and fibrosis, calcifica- key profibrogenic factor, is increased in acinar and stromal
tion, and nerve enlargement. Clinical manifestations include cells of the rat pancreas in this model, indicating that repetitive
pain, maldigestion, and possible diabetes. Although pain is treatment with caerulein may cause fibrosis, a key feature of
common, exocrine insufficiency occurs late in the course of the CP (53). Indeed, repeated caerulein insult could induce exo-
disease because the pancreas has a great functional reserve. crine deficiency, fibrosis, and diabetes (115). As in AP models,
Steatorrhea occurs only when lipase secretion is reduced to less this is the most commonly used and reproducible model for
than 10% of normal (162). In the absence of established CP. The procedures vary in different studies and can be used in
diagnostic criteria, early diagnosis of human CP is challenging combination with many other insults. In one early study, rats
and often based on a combination of clinical presentation, were treated with water immersion stress for 5 h and two
imaging results, and pancreatic function test results (35, 105). intraperitoneal injections of caerulein (20 ␮g/kg body wt) once
There is also no consensus on the criteria for evaluating animal a week for 16 wk. Pancreatic atrophy, marked fibrosis, fatty
changes, and destruction of the lobular architecture were dem-
models of CP. Because the purpose of pancreatitis research is
onstrated microscopically; diabetes was also present (52).
to eliminate the inflammatory response, prevent fatty/fibrotic
Acute pancreatic injury induced in mice by twice-weekly
replacement, and preserve both exocrine and endocrine pan-
caerulein treatment (50 ␮g·kg⫺1·h⫺1 ⫻ 6 h) for 10 wk in-
creatic functions, the parameters used to evaluate CP include
creased procollagen expression and progressive accumulation
pancreatic weight, blood insulin/glucose levels, pancreatic am-
of the extracellular matrix surrounding acinar units and in
ylase content, inflammatory cell including macrophage and interlobular spaces. Atrophy, transdifferentiation of acinar
lymphocyte infiltrations, ␣-SMA expression (a marker for units to ductlike tubular complexes, and dilatation of intra-
early stellate cell activation), and areas of fatty/fibrotic replace- acinar lumina also developed (117). A more closely spaced
ment (45, 69, 74, 140, 141, 183). treatment strategy in which AP was induced three times weekly
The pathogenesis of CP is poorly understood. Accumulated instead of twice weekly produced an even more rapid accumu-
evidence suggests that the incidence of CP is increasing (77). lation of periacinar fibrosis and altered acinar architecture 6 wk
Alcohol is the major cause of CP in the Western world. Other after the initial AP induction (116, 158). After the insults were
etiologies include smoking, obstructive lesions, other toxic stopped, the pancreatic fibrosis mostly resolved in 3– 6 wk,
agents, and genetic factors (178). Ethanol has both direct and suggesting that the processes responsible for matrix resolution
indirect toxic effects on the integrity of pancreatic acinar cells are potently active in the pancreas (116). Additionally, the
that are mediated by the ethanol metabolite acetaldehyde (7). addition of lipopolysaccharide (LPS) increased the amount of
Oxidative stress caused by ethanol or nicotine can lead to the fibrosis (121). This may be because pancreatic stellate cells
peroxidation of the lipid bilayer of the cell membrane, which express a variety of Toll-like receptors (TLRs) and respond to
consecutively disintegrates the membrane (108). Different eti- TLR ligands (e.g., LPS), leading to the activation of signaling
ologies may have different pathogenic mechanisms, and di- pathways and proinflammatory responses (109).
verse signaling leads to various phenotypic changes. For ex- Cyclosporin is a drug that binds to cyclophilin to inhibit
ample, prolonged activity of NF-␬B results in inflammatory calcineurin and increases TGF-␤ expression. Although cyclo-
cell infiltration, activation of stellate cells, loss of acinar cells, sporin accumulates at high concentrations in the pancreas, it
and fibrosis, which are characteristics of CP (66). Ras activa- only produces minor morphological or functional derange-
tion leads to acinar cell senescence and generates inflammation ments. However, the combination of cyclosporin and caerulein
and massive fibrosis resembling the histological features of CP exacerbates the chronic inflammatory response in the pancreas
(28, 74, 97). In contrast, the primary response to intracellular (161), but the doses and duration of caerulein treatment and the

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00372.2015 • www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi at Univ De Antioquia (200.024.017.012) on February 21, 2021.
Themes
G350 PANCREATITIS MODELS

timing of tissue collection seem to be important in determining Alcohol-related CP models. In 1967, Maki et al. (106)
the phenotype observed. In one study, pancreatitis was induced studied the influence of the oral administration of alcohol on
nine times at intervals of ⬃20 days; 3 days after the last the rat pancreas and the interaction of alcohol with various
injection of caerulein, inflammation was still observed. How- dietary compositions. Alcohol with a concentration of 15%
ever, 6 wk later, the pancreas fully recovered (37). Using was added to the drinking water of the study animals for 2 mo.
higher frequencies of caerulein applications will likely lead to The alcohol intake caused increased serum amylase levels,
more a dramatic formation of pancreatic fibrosis. pancreatic edema, and vacuolization of acinar cells, but fat
Serial intraperitoneal injections of arginine with initial dose necrosis or pancreatic necrosis was not noted. Interestingly, the
of 500 mg/100 g body wt followed by three injections at 250 rats that were fed on a high-carbohydrate diet showed minimal
mg/100 g over 10 days was also used to produce CP. After 24 changes in the pancreas compared with those fed on high-
h the pancreas had severe edematous AP. By day 5 there was protein and high-fat diets and with those fed on low-protein
up to 90% acinar destruction with adipose tissue replacement, and high-fat diets. In 1971, Sarles et al. (144) fed rats with 20%
but ductal and islet cells appeared undamaged. These changes ethanol for 20 –30 mo. The average daily consumption of
were still present 6 mo after injection (31). In another study, alcohol was 4 g/rat. More than half of the animals developed
male rats were intraperitoneally injected daily (350 mg/100 g pancreatic lesions similar to those of human CP. The patho-
body wt) with L-arginine from 1 to 4 wk. At week 1, light logical changes included reduced acini, duct multiplication,
microscopy revealed areas of focal acinar cell degeneration. At protein plugs, sometimes calcified ducts, and sclerosis. Beta-
the end of week 2, acinar necrosis was evident throughout most cell adenomata of the islets of Langerhans were also observed
pancreatic lobules. At week 4, only isolated, single acinar cells in four of the rats exposed to ethanol.
remained within a fibrous connective tissue matrix contiguous Currently, the most popular protocol used to imitate the
with ducts, blood vessels, intrapancreatic nerves, and islets effects of alcohol on various organs was developed by Lieber
(169). Instead of fibrosis, Yamaguchi et al. (180) showed that and DeCarli in 1975 for alcohol liver injury (96). The feeding
85 to 90% of the acinar tissue was replaced by fatty tissue and regime was based on the supplementation of ethanol into a
dilated pancreatic ducts on day 54 after the first intraperitoneal liquid diet (referred as Lieber-DeCarli diet), and it allows an
arginine injection. increased total alcohol intake. Isocaloric substitution of carbo-
Mice fed with an intermittent CDE diet (3 days of CDE diet hydrates by ethanol (36% of total calories in rats) resulted in
alternating with 3 days of normal diet) for a prolonged period the production of fatty liver disease, alcoholic hepatitis, and
of 24 wk can develop some histological features of CP. The eventually cirrhosis. This diet leads to severe organ damage in
fibrosis in this model is mild (69). the liver but pancreatic morphology changes are mild. Rather
In mice and rats, complete pancreatic duct obstruction than CP, only mild functional insufficiency develops in most
mainly results in atrophy of the organ and does not lead to the cases. These observations correlate with the clinical finding
typical morphology of CP. Several months after pancreatic that only a minority (less than 10%) of alcoholics ever develop
duct ligation, there is intralobular fatty replacement of the clinical CP (94). Therefore, alcohol alone is a very weak
exocrine pancreas (168). Therefore, duct ligation alone does inducer of CP, and other factors are necessary for the onset of
not cause typical CP. However, complete pancreatic duct CP. However, the Lieber-DeCarli administration causes a num-
obstruction and daily caerulein hyperstimulation caused large ber of changes that may predispose the gland to damage. These
amounts of interstitial collagen expression after 72 h (114). changes include an increased concentration of digestive en-
Recently, a modified-CP model by branch duct ligation in zymes and the lysosomal enzyme cathepsin B, which is im-
combination with caerulein administration has been developed portant in intracellular trypsinogen activation. It is believed
and could be a valuable model (146). In this study, CP was that alcohol “sensitizes” or “primes” the pancreas to pancre-
induced by ligation of the pancreatic duct at the junction atitis; current studies are focused on the mechanisms respon-
between the gastric and the duodenal lobe sparing the bile duct sible for the sensitizing effects of alcohol, and Lieber-DeCarli
and its concomitant artery. These animals then received a diet is usually used in combination with many other factors
single injection of caerulein (50 ␮g/kg body wt) 2 days after (genetic, environmental, or dietary) to induce CP (10, 125).
duct ligation. Severe necrotizing pancreatitis developed in the The pancreata from rats fed ethanol for 9 –12 mo are more
ligated part of the pancreas 3 days after surgery. Model- susceptible to caerulein-induced activation of chymotrypsino-
associated mortality ranged from 10 to 15% and always oc- gen than the pancreata from pair-fed control animals (134).
curred within 48 h of caerulein injection. Areas of pancreatic The combination of alcohol feeding with caerulein injections
necrosis were replaced by fibrotic and fatty tissues, and max- exacerbates pancreatitis with increased fibrosis and loss of
imal fibrosis was detected after 21 days. parenchyma. Additionally, calcifications indicating severe CP
The mortality rate is usually high in acute hemorrhagic can be observed. Although pancreatic function was not directly
pancreatitis that is induced by retrograde infusion of sodium tested, there was evidence that digestive enzyme synthesis is
taurocholate into the pancreatic duct system of the rats. In reduced after chronic ethanol feeding (32, 131).
animals surviving 72 h, there was marked acinar atrophy and Combining LPS and the alcohol model can be adopted
pancreatic fibrosis (5). because of its potential clinical relevance. In one study, alco-
However, with a single retrograde intraductal infusion of 40 hol-fed rats receiving a single LPS injection displayed more
␮l/100 g body wt of 3% sodium taurocholate, the pancreas severe acinar vacuolization, acinar cell necrosis, inflammatory
appeared to be histologically normal 42 days after intraductal infiltrates, and hemorrhaging. Negligible lesions were found in
infusion (180). The volume, concentration, pressure, and du- the control diet-fed plus LPS group. These lesions mimic some
ration may be factors that are complicated in these phenotypic of the features of AP (164). When the rats fed Lieber-DeCarli
changes. liquid diets for 10 wk were challenged with three repeated

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00372.2015 • www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi at Univ De Antioquia (200.024.017.012) on February 21, 2021.
Themes
PANCREATITIS MODELS G351
doses of LPS (3 mg/kg intravenously per week for 3 wk), ACKNOWLEDGMENTS
significantly greater pancreatic injury and pancreatic fibrosis We thank Kelly E. Viola for critical editing.
were seen in the alcohol-fed rats but not in control diet group.
In in vitro studies, alcohol and LPS exerted a synergistic effect GRANTS
on PSC activation, and pancreata exposed to alcohol were The project described was supported by P50 CA102701 and K12 CA90628
more sensitive to LPS-induced damage. This may be caused by from the National Cancer Institute and W81XWH-15-1-0257 from the Depart-
increased sensitivity to necrotic cell death rather than apoptotic ment of Defense. This publication was also made possible by the CTSA Grant
UL1 TR000135 from the National Center for Advancing Translational Sci-
cell death (42). Withdrawal of alcohol led to the resolution of ences (NCATS), a component of the National Institutes of Health. The content
pancreatic lesions including increased PSC apoptosis and de- is solely the responsibility of the authors and does not necessarily represent the
creased fibrosis (163). official views of the National Institutes of Health or the Department of
Smoking is a strong and independent risk factor of pancre- Defense.
atitis. Cigarette smoke dose dependently potentiates the
DISCLOSURES
amount of pancreatic injury generated by ethanol alone (63,
179). Pancreatic stellate cells can be activated by clinically No conflicts of interest, financial or otherwise, are declared by the author (s).
relevant concentrations of cigarette smoke components (90).
AUTHOR CONTRIBUTIONS
In a cyclosporin model of alcoholic CP developed by Guk-
ovsky et al. (57), the addition of cyclosporin and caerulein in X.Z., F.W., Y.B., and B.J. drafted manuscript; Y.B. and B.J. conception and
rats fed with ethanol led to a decrease of the pancreatic design of research; Y.B. and B.J. edited and revised manuscript; B.J. approved
final version of manuscript.
parenchyma by ⬃86%. The combination markedly increased
NF-␬B activation and cytokine/chemokine expression, colla- REFERENCES
gen, fibronectin, the activities of matrix metalloproteinases,
and the activation of pancreatic stellate cells. 1. Acosta JM, Ledesma CL. Gallstone migration as a cause of acute
pancreatitis. N Engl J Med 290: 484 –487, 1974.
It has been found that, in animals fed on a high-protein and 2. Adler G, Beglinger C, Braun U, Reinshagen M, Koop I, Schafmayer
high-fat diet, ethanol increased the amount of the excretory A, Rovati L, Arnold R. Interaction of the cholinergic system and
enzymes in the pancreas (26). In Tsukamoto’s study (160), rats cholecystokinin in the regulation of endogenous and exogenous stimu-
implanted with gastrostomy catheters were fed with increasing lation of pancreatic secretion in humans. Gastroenterology 100: 537–
543, 1991.
doses of ethanol (from 8 g·kg⫺1·day⫺1 to 15 g·kg⫺1·day⫺1) 3. Aeffner F, Bolon B, Davis IC. Mouse models of acute respiratory
plus corn oil unsaturated fat: 5% (low-fat), 25% (high-fat), or distress syndrome: a review of analytical approaches, pathologic features,
35% (extra-high-fat) of total calories. With the low-fat diet, and common measurements. Toxicol Pathol 43: 1074 –1092, 2015.
chronic ethanol feeding produced only mild pancreatic pathol- 4. Aghdassi AA, Mayerle J, Christochowitz S, Weiss FU, Sendler M,
Lerch MM. Animal models for investigating chronic pancreatitis. Fi-
ogy, such as steatosis and interstitial edema. However, ethanol brogenesis Tissue Repair 4: 26, 2011.
with the high-fat and extra-high-fat diets caused increased 5. Aho HJ, Koskensalo SM, Nevalainen TJ. Experimental pancreatitis in
acinar cell apoptosis. Focal lesions of CP were also observed in the rat. Sodium taurocholate-induced acute haemorrhagic pancreatitis.
20 or 30% of ethanol-fed animals given the high-fat or extra- Scand J Gastroenterol 15: 411–416, 1980.
high-fat diet. These lesions included fat necrosis, mononuclear 6. Alhan E, Usta A, Turkyilmaz S, Kural BV, Ercin C. Effects of
glutamine alone on the acute necrotizing pancreatitis in rats. J Surg Res
cell infiltration, fibrosis, acinar atrophy, ductal dilatation, and 193: 161–167, 2015.
intraductal mucinous or proteinaceous plugs (160). Interest- 7. Ammann RW, Heitz PU, Kloppel G. Course of alcoholic chronic
ingly, although pancreatic damage was associated with unsat- pancreatitis: a prospective clinicomorphological long-term study. Gas-
urated fat, medium-chain triglycerides (saturated fat) signifi- troenterology 111: 224 –231, 1996.
8. Anastasi A, Erspamer V, Endean R. Isolation and structure of caeru-
cantly blunted the damage, suggesting that the dietary fat types lein, an active decapeptide from the skin of Hyla caerulea. Experientia
have different roles in pancreatitis (83). 23: 699 –700, 1967.
9. Apte M, Pirola R, Wilson J. The fibrosis of chronic pancreatitis: new
Perspectives insights into the role of pancreatic stellate cells. Antioxid Redox Signal
15: 2711–2722, 2011.
There are many models available for both acute and chronic 10. Apte MV, Norton ID, Wilson JS. Ethanol induced acinar cell injury.
Alcohol Alcohol Suppl 2: 365–368, 1994.
pancreatitis. These models have been discussed in many ex- 11. Apte MV, Wilson JS, Lugea A, Pandol SJ. A starring role for stellate
cellent reviews (4, 43, 68, 78, 91, 143, 184). However, inves- cells in the pancreatic cancer microenvironment. Gastroenterology 144:
tigators studying pancreatitis face several challenges. The first 1210 –1219, 2013.
and most major challenge is the clinical relevance of the 12. Ateyya H, Wagih HM, El-Sherbeeny NA. Effect of tiron on remote
organ injury in rats with severe acute pancreatitis induced by L-arginine.
models. Unfortunately, many models use non-clinically rele- Naunyn Schmiedebergs Arch Pharmacol 389: 873–885, 2016.
vant stimulants, while others fail to reproduce human diseases, 13. Banks PA, Freeman ML. Practice guidelines in acute pancreatitis. Am
even with human-related etiological insults. Thus the mecha- J Gastroenterol 101: 2379 –2400, 2006.
nisms found from these models should be interpreted with 14. Bartholomew C. Acute scorpion pancreatitis in Trinidad. Br Med J 1:
caution. The second is that reproducible rodent models for 666 –668, 1970.
15. Bernard C. Memoir on the pancreas and on the role of pancreatic juice
some forms of human pancreatic inflammatory diseases with in digestive processes, particularly in the digestion of neutral fat. 1856.
known etiologies (e.g., hereditary pancreatitis, cystic fibrosis) Transl. Henderson J. Monogr Physiol Soc 42: 1–131, 1985.
have yet to be developed. Last, current research largely focuses 16. Bertaccini G, De Caro G, Endean R, Erspamer V, Impicciatore M.
on the initiating mechanisms. Effective preventive and thera- The action of caerulein on pancreatic secretion of the dog and biliary
secretion of the dog and the rat. Br J Pharmacol 37: 185–197, 1969.
peutic drugs for the human pancreatitis are still lacking. Clin- 17. Bi Y, Page SL, Williams JA. Rho and Rac promote acinar morpholog-
ically relevant models are urgently needed for developing and ical changes, actin reorganization, and amylase secretion. Am J Physiol
testing interventions. Gastrointest Liver Physiol 289: G561–G570, 2005.

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00372.2015 • www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi at Univ De Antioquia (200.024.017.012) on February 21, 2021.
Themes
G352 PANCREATITIS MODELS

18. Bi Y, Williams JA. Receptor biology and signal transduction in pancre- 40. Farber E, Popper H. Production of acute pancreatitis with ethionine and
atic acinar cells. Curr Opin Gastroenterol 20: 427–434, 2004. its prevention by methionine. Proc Soc Exp Biol Med 74: 838 –840, 1950.
19. Biczo G, Hegyi P, Berczi S, Dosa S, Hracsko Z, Varga IS, Ivanyi B, 41. Foitzik T, Hotz HG, Eibl G, Buhr HJ. Experimental models of acute
Venglovecz V, Wittmann T, Takacs T, Rakonczay Z Jr. Inhibition of pancreatitis: are they suitable for evaluating therapy? Int J Colorectal Dis
arginase activity ameliorates L-arginine-induced acute pancreatitis in rats. 15: 127–135, 2000.
Pancreas 39: 868 –874, 2010. 42. Fortunato F, Deng X, Gates LK, McClain CJ, Bimmler D, Graf R,
20. Biczo G, Hegyi P, Dosa S, Shalbuyeva N, Berczi S, Sinervirta R, Whitcomb DC. Pancreatic response to endotoxin after chronic alcohol
Hracsko Z, Siska A, Kukor Z, Jarmay K, Venglovecz V, Varga IS, exposure: switch from apoptosis to necrosis? Am J Physiol Gastrointest
Ivanyi B, Alhonen L, Wittmann T, Gukovskaya A, Takacs T, Ra- Liver Physiol 290: G232–G241, 2006.
konczay Z Jr. The crucial role of early mitochondrial injury in L-lysine- 43. Foster JR. A review of animal models of nonneoplastic pancreatic
induced acute pancreatitis. Antioxid Redox Signal 15: 2669 –2681, 2011. diseases. Toxicol Pathol 42: 243–259, 2014.
21. Bilchik AJ, Zucker KA, Adrian TE, Modlin IM. Amelioration of 44. Frick TW, Dalo S, O’Leary JF, Runge W, Borner JW, Baraniewski
cholinergic-induced pancreatitis with a selective cholecystokinin recep- H, Dressel T, Shearen JG, Goodale RL. Effects of insecticide, diazi-
tor antagonist. Arch Surg 125: 1546 –1549, 1990. non, on pancreas of dog, cat and guinea pig. J Environ Pathol Toxicol
22. Boquist L. Morphologic effects of ethionine on the pancreas of the Oncol 7: 1–11, 1987.
Chinese hamster. A light and electron microscopic study of degenerative 45. Gaiser S, Daniluk J, Liu Y, Tsou L, Chu J, Lee W, Longnecker DS,
changes. Acta Pathol Microbiol Scand 76: 91–105, 1969. Logsdon CD, Ji B. Intracellular activation of trypsinogen in transgenic
23. Chan YC, Leung PS. Acute pancreatitis: animal models and recent mice induces acute but not chronic pancreatitis. Gut 60: 1379 –1388,
advances in basic research. Pancreas 34: 1–14, 2007. 2011.
24. Cheever FS, Daniels JB, Hersey EF. A viral agent isolated from a case 46. Gallagher S, Sankaran H, Williams JA. Mechanism of scorpion
of “non-paralytic poliomyelitis” and pathogenic for suckling mice: its toxin-induced enzyme secretion in rat pancreas. Gastroenterology 80:
possible relation to the coxsackie group of viruses. J Exp Med 92: 970 –973, 1981.
153–167, 1950. 47. Gautam D, Han SJ, Heard TS, Cui Y, Miller G, Bloodworth L, Wess
25. Clemens DL, Jerrells TR. Ethanol consumption potentiates viral pan- J. Cholinergic stimulation of amylase secretion from pancreatic acinar
creatitis and may inhibit pancreas regeneration: preliminary findings. cells studied with muscarinic acetylcholine receptor mutant mice. J
Alcohol 33: 183–189, 2004. Pharmacol Exp Ther 313: 995–1002, 2005.
26. Criddle DN. The role of fat and alcohol in acute pancreatitis: a danger- 48. Gioiello A, Cerra B, Mostarda S, Guercini C, Pellicciari R, Macchi-
ous liaison. Pancreatology 15: S6 –S12, 2015. arulo A. Bile acid derivatives as ligands of the farnesoid x receptor:
27. Criddle DN, Raraty MG, Neoptolemos JP, Tepikin AV, Petersen molecular determinants for bile acid binding and receptor modulation.
OH, Sutton R. Ethanol toxicity in pancreatic acinar cells: mediation by Curr Top Med Chem 14: 2159 –2174, 2014.
nonoxidative fatty acid metabolites. Proc Natl Acad Sci USA 101: 49. Goldberg RC, Chaikoff IL, Dodge AH. Destruction of pancreatic
10738 –10743, 2004. acinar tissue by DL-ethionine. Proc Soc Exp Biol Med 74: 869 –872,
28. Daniluk J, Liu Y, Deng D, Chu J, Huang H, Gaiser S, Cruz-
1950.
Monserrate Z, Wang H, Ji B, Logsdon CD. An NF-kappaB pathway-
50. Gomez-Perez Y, Gianotti M, Llado I, Proenza AM. Sex-dependent
mediated positive feedback loop amplifies Ras activity to pathological
effects of high-fat-diet feeding on rat pancreas oxidative stress. Pancreas
levels in mice. J Clin Invest 122: 1519 –1528, 2012.
40: 682–688, 2011.
29. Dawra R, Sharif R, Phillips P, Dudeja V, Dhaulakhandi D, Saluja
51. Gomez RM, Lascano EF, Berria MI. Murine acinar pancreatitis pre-
AK. Development of a new mouse model of acute pancreatitis induced
ceding necrotizing myocarditis after Coxsackievirus B3 inoculation. J
by administration of L-arginine. Am J Physiol Gastrointest Liver Physiol
Med Virol 35: 71–75, 1991.
292: G1009 –G1018, 2007.
52. Goto M, Nakano I, Kimura T, Miyahara T, Kinjo M, Nawata H. New
30. De Almeida AL, Grossman MI. Experimental production of pancreati-
chronic pancreatitis model with diabetes induced by cerulein plus stress
tis with ethionine. Gastroenterology 20: 554 –577, 1952.
31. Delaney CP, McGeeney KF, Dervan P, Fitzpatrick JM. Pancreatic in rats. Dig Dis Sci 40: 2356 –2363, 1995.
atrophy: a new model using serial intra-peritoneal injections of L-argi- 53. Gress T, Muller-Pillasch F, Elsasser HP, Bachem M, Ferrara C,
nine. Scand J Gastroenterol 28: 1086 –1090, 1993. Weidenbach H, Lerch M, Adler G. Enhancement of transforming
32. Deng X, Wang L, Elm MS, Gabazadeh D, Diorio GJ, Eagon PK, growth factor beta 1 expression in the rat pancreas during regeneration
Whitcomb DC. Chronic alcohol consumption accelerates fibrosis in from caerulein-induced pancreatitis. Eur J Clin Invest 24: 679 –685,
response to cerulein-induced pancreatitis in rats. Am J Pathol 166: 1994.
93–106, 2005. 54. Griffith WH, Wade NJ. Choline metabolism. I. The occurrence and
33. Dorigotti L, Glasser AH. Comparative effects of caerulein, pancreozy- prevention of hemorrhagic degeneration in young rats on a low choline
min and secretin on pancreatic blood flow. Experientia 24: 806 –807, diet. Nutrition classics from The Journal of Biological Chemistry 131:
1968. 567–577, 1937. Nutr Rev 32: 112–114, 1974.
34. Dressel TD, Goodale RL Jr, Arneson MA, Borner JW. Pancreatitis as 55. Gukovskaya AS, Mouria M, Gukovsky I, Reyes CN, Kasho VN,
a complication of anticholinesterase insecticide intoxication. Ann Surg Faller LD, Pandol SJ. Ethanol metabolism and transcription factor
189: 199 –204, 1979. activation in pancreatic acinar cells in rats. Gastroenterology 122: 106 –
35. Duggan SN, Ni Chonchubhair HM, Lawal O, O’Connor DB, Conlon 118, 2002.
KC. Chronic pancreatitis: a diagnostic dilemma. World J Gastroenterol 56. Gukovskaya AS, Perkins P, Zaninovic V, Sandoval D, Rutherford R,
22: 2304 –2313, 2016. Fitzsimmons T, Pandol SJ, Poucell-Hatton S. Mechanisms of cell
36. Elmunzer BJ, Scheiman JM, Lehman GA, Chak A, Mosler P, death after pancreatic duct obstruction in the opossum and the rat.
Higgins PD, Hayward RA, Romagnuolo J, Elta GH, Sherman S, Gastroenterology 110: 875–884, 1996.
Waljee AK, Repaka A, Atkinson MR, Cote GA, Kwon RS, McHenry 57. Gukovsky I, Lugea A, Shahsahebi M, Cheng JH, Hong PP, Jung YJ,
L, Piraka CR, Wamsteker EJ, Watkins JL, Korsnes SJ, Schmidt SE, Deng QG, French BA, Lungo W, French SW, Tsukamoto H, Pandol
Turner SM, Nicholson S, Fogel EL. A randomized trial of rectal SJ. A rat model reproducing key pathological responses of alcoholic
indomethacin to prevent post-ERCP pancreatitis. N Engl J Med 366: chronic pancreatitis. Am J Physiol Gastrointest Liver Physiol 294:
1414 –1422, 2012. G68 –G79, 2008.
37. Elsasser HP, Haake T, Grimmig M, Adler G, Kern HF. Repetitive 58. Gurda GT, Crozier SJ, Ji B, Ernst SA, Logsdon CD, Rothermel BA,
cerulein-induced pancreatitis and pancreatic fibrosis in the rat. Pancreas Williams JA. Regulator of calcineurin 1 controls growth plasticity of
7: 385–390, 1992. adult pancreas. Gastroenterology 139: 609 –619, 619.e1–e6, 2010.
38. Evander A, Lundquist I, Ihse I. Hormonal and cholinergic effects on 59. Hai Z, Jiang C, Zhang J, Wang L, Fang K. Relationship between
amylase and lysosomal enzyme activities in pancreatic tissue and ascites carbachol hyperstimulation-induced pancreatic acinar cellular injury and
of rats with acute experimental pancreatitis. J Surg Res 35: 168 –175, trypsinogen or NF-kappaB activation in rats in vitro. J Huazhong Univ
1983. Sci Technolog Med Sci 26: 34 –35, 58, 2006.
39. Fan M, Wang X, Xu G, Yan Q, Huang W. Bile acid signaling and liver 60. Harb JM, Burch GE. Spherical aggregates of coxsackie B4 virus
regeneration. Biochim Biophys Acta 1849: 196 –200, 2015. particles in mouse pancreas. Beitr Pathol 156: 122–127, 1975.

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00372.2015 • www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi at Univ De Antioquia (200.024.017.012) on February 21, 2021.
Themes
PANCREATITIS MODELS G353
61. Hardman J, Jamdar S, Shields C, McMahon R, Redmond HP, 84. Lampel M, Kern HF. Acute interstitial pancreatitis in the rat induced by
Siriwardena AK. Intravenous selenium modulates L-arginine-induced excessive doses of a pancreatic secretagogue. Virchows Arch A Pathol
experimental acute pancreatitis. JOP 6: 431–437, 2005. Anat Histol 373: 97–117, 1977.
62. Hardman J, Shields C, Schofield D, McMahon R, Redmond HP, 85. Lankisch PG, Apte M, Banks PA. Acute pancreatitis. Lancet 386:
Siriwardena AK. Intravenous antioxidant modulation of end-organ 85–96, 2015.
damage in L-arginine-induced experimental acute pancreatitis. Pancrea- 86. Lansdown AB. Pathological changes in the pancreas of mice following
tology 5: 380 –386, 2005. infection with Coxsackie B viruses. Br J Exp Pathol 57: 331–338, 1976.
63. Hartwig W, Werner J, Ryschich E, Mayer H, Schmidt J, Gebhard 87. Laukkarinen JM, Van Acker GJ, Weiss ER, Steer ML, Perides G. A
MM, Herfarth C, Klar E. Cigarette smoke enhances ethanol-induced mouse model of acute biliary pancreatitis induced by retrograde pancre-
pancreatic injury. Pancreas 21: 272–278, 2000. atic duct infusion of Na-taurocholate. Gut 56: 1590 –1598, 2007.
64. Hietaranta AJ, Saluja AK, Bhagat L, Singh VP, Song AM, Steer ML. 88. Le Page SL, Bi Y, Williams JA. CCK-A receptor activates RhoA
Relationship between NF-kappaB and trypsinogen activation in rat pan- through G alpha 12/13 in NIH3T3 cells. Am J Physiol Cell Physiol 285:
creas after supramaximal caerulein stimulation. Biochem Biophys Res C1197–C1206, 2003.
Commun 280: 388 –395, 2001. 89. Le T, Eisses JF, Lemon KL, Ozolek JA, Pociask DA, Orabi AI,
65. Hofmann AF, Small DM. Detergent properties of bile salts: correlation Husain SZ. Intraductal infusion of taurocholate followed by distal
with physiological function. Annu Rev Med 18: 333–376, 1967. common bile duct ligation leads to a severe necrotic model of pancreatitis
66. Huang H, Liu Y, Daniluk J, Gaiser S, Chu J, Wang H, Li ZS, in mice. Pancreas 44: 493–499, 2015.
Logsdon CD, Ji B. Activation of nuclear factor-kappaB in acinar cells 90. Lee AT, Xu Z, Pothula SP, Patel MB, Pirola RC, Wilson JS, Apte
increases the severity of pancreatitis in mice. Gastroenterology 144: MV. Alcohol and cigarette smoke components activate human pancreatic
202–210, 2013. stellate cells: implications for the progression of chronic pancreatitis.
67. Huang W, Booth DM, Cane MC, Chvanov M, Javed MA, Elliott VL, Alcohol Clin Exp Res 39: 2123–2133, 2015.
Armstrong JA, Dingsdale H, Cash N, Li Y, Greenhalf W, Mukherjee 91. Lerch MM, Gorelick FS. Models of acute and chronic pancreatitis.
R, Kaphalia BS, Jaffar M, Petersen OH, Tepikin AV, Sutton R, Gastroenterology 144: 1180 –1193, 2013.
Criddle DN. Fatty acid ethyl ester synthase inhibition ameliorates 92. Lerch MM, Saluja AK, Dawra R, Ramarao P, Saluja M, Steer ML.
ethanol-induced Ca2⫹-dependent mitochondrial dysfunction and acute Acute necrotizing pancreatitis in the opossum: earliest morphological
pancreatitis. Gut 63: 1313–1324, 2014. changes involve acinar cells. Gastroenterology 103: 205–213, 1992.
68. Hyun JJ, Lee HS. Experimental models of pancreatitis. Clin Endosc 47: 93. Lerch MM, Saluja AK, Runzi M, Dawra R, Steer ML. Luminal
212–216, 2014. endocytosis and intracellular targeting by acinar cells during early biliary
69. Ida S, Ohmuraya M, Hirota M, Ozaki N, Hiramatsu S, Uehara H, pancreatitis in the opossum. J Clin Invest 95: 2222–2231, 1995.
Takamori H, Araki K, Baba H, Yamamura K. Chronic pancreatitis in 94. Li J, Guo M, Hu B, Liu R, Wang R, Tang C. Does chronic ethanol
mice by treatment with choline-deficient ethionine-supplemented diet. intake cause chronic pancreatitis?: evidence and mechanism. Pancreas
Exp Anim 59: 421–429, 2010. 37: 189 –195, 2008.
70. Jerrells TR, Chapman N, Clemens DL. Animal model of alcoholic 95. Li Y, Owyang C. Vagal afferent pathway mediates physiological action
pancreatitis: role of viral infections. Pancreas 27: 301–304, 2003. of cholecystokinin on pancreatic enzyme secretion. J Clin Invest 92:
71. Ji B, Bi Y, Simeone D, Mortensen RM, Logsdon CD. Human pancre- 418 –424, 1993.
atic acinar cells lack functional responses to cholecystokinin and gastrin. 96. Lieber CS, DeCarli LM. Alcoholic liver injury: experimental models in
Gastroenterology 121: 1380 –1390, 2001. rats and baboons. Adv Exp Med Biol 59: 379 –393, 1975.
72. Ji B, Kopin AS, Logsdon CD. Species differences between rat and 97. Logsdon CD, Ji B. Ras activity in acinar cells links chronic pancreatitis
mouse CCKA receptors determine the divergent acinar cell response to and pancreatic cancer. Clin Gastroenterol Hepatol 7: S40 –S43, 2009.
the cholecystokinin analog JMV-180. J Biol Chem 275: 19115–19120, 98. Logsdon CD, Ji B. The role of protein synthesis and digestive enzymes
2000. in acinar cell injury. Nat Rev Gastroenterol Hepatol 10: 362–370, 2013.
73. Ji B, Logsdon CD. Digesting new information about the role of trypsin 99. Lombardi AV. A factor analysis of morphogenetic fields in the human
in pancreatitis. Gastroenterology 141: 1972–1975, 2011. dentition. Am J Phys Anthropol 42: 99 –104, 1975.
74. Ji B, Tsou L, Wang H, Gaiser S, Chang DZ, Daniluk J, Bi Y, Grote 100. Lombardi B, Estes LW, Longnecker DS. Acute hemorrhagic pancre-
T, Longnecker DS, Logsdon CD. Ras activity levels control the devel- atitis (massive necrosis) with fat necrosis induced in mice by DL-
opment of pancreatic diseases. Gastroenterology 137: 1072–1082, ethionine fed with a choline-deficient diet. Am J Pathol 79: 465–480,
1082.e1– e6, 2009. 1975.
75. Jin S, Orabi AI, Le T, Javed TA, Sah S, Eisses JF, Bottino R, 101. Lombardi B, Rao NK. Acute hemorrhagic pancreatic necrosis in mice.
Molkentin JD, Husain SZ. Exposure to radiocontrast agents induces Influence of the age and sex of the animals and of dietary ethionine,
pancreatic inflammation by activation of nuclear factor-kappab, calcium choline, methionine, and adenine sulfate. Am J Pathol 81: 87–100, 1975.
signaling, and calcineurin. Gastroenterology 149: 753–764.e11, 2015. 102. Lugea A, Gong J, Nguyen J, Nieto J, French SW, Pandol SJ.
76. Johnson LR, Stening GF, Grossman MI. Effect of sulfation on the Cholinergic mediation of alcohol-induced experimental pancreatitis. Al-
gastrointestinal actions of caerulein. Gastroenterology 58: 208 –216, cohol Clin Exp Res 34: 1768 –1781, 2010.
1970. 103. Lugea A, Tischler D, Nguyen J, Gong J, Gukovsky I, French SW,
77. Jupp J, Fine D, Johnson CD. The epidemiology and socioeconomic Gorelick FS, Pandol SJ. Adaptive unfolded protein response attenuates
impact of chronic pancreatitis. Best Pract Res Clin Gastroenterol 24: alcohol-induced pancreatic damage. Gastroenterology 140: 987–997,
219 –231, 2010. 2011.
78. Kahl S, Mayer JM. Update on experimental acute pancreatitis. Minerva 104. Machado JC, da Silveira Filho JF. [Induction of acute hemorrhagic
Gastroenterol Dietol 58: 355–363, 2012. pancreatitis in dogs by scorpion venom from T serrulatus]. Mem Inst
79. Kaiser AM, Saluja AK, Sengupta A, Saluja M, Steer ML. Relation- Butantan 40 – 41: 1–9, 1976.
ship between severity, necrosis, and apoptosis in five models of experi- 105. Majumder S, Chari ST. Chronic pancreatitis. Lancet 387: 1957–1966,
mental acute pancreatitis. Am J Physiol Cell Physiol 269: C1295–C1304, 2016.
1995. 106. Maki T, Kakizaki G, Sato T, Saito Y, Onuma T. Experimental study
80. Kambhampati S, Park W, Habtezion A. Pharmacologic therapy for on alcoholic pancreatitis. Tohoku J Exp Med 92: 415–421, 1967.
acute pancreatitis. World J Gastroenterol 20: 16868 –16880, 2014. 107. Maleth J, Balazs A, Pallagi P, Balla Z, Kui B, Katona M, Judak L,
81. Kirkbride MB. The islands of Langerhans after ligation of the pancre- Nemeth I, Kemeny LV, Rakonczay Z Jr, Venglovecz V, Foldesi I,
atic ducts. J Exp Med 15: 101–105, 1912. Peto Z, Somoracz A, Borka K, Perdomo D, Lukacs GL, Gray MA,
82. Kirkland JG, Godfrey CB, Garrett R, Kakar S, Yeh BM, Corvera Monterisi S, Zaccolo M, Sendler M, Mayerle J, Kuhn JP, Lerch MM,
CU. Reversible surgical model of biliary inflammation and obstructive Sahin-Toth M, Hegyi P. Alcohol disrupts levels and function of the
jaundice in mice. J Surg Res 164: 221–227, 2010. cystic fibrosis transmembrane conductance regulator to promote devel-
83. Kono H, Nakagami M, Rusyn I, Connor HD, Stefanovic B, Brenner opment of pancreatitis. Gastroenterology 148: 427–439.e16, 2015.
DA, Mason RP, Arteel GE, Thurman RG. Development of an animal 108. Malfertheiner P, Schutte K. Smoking—a trigger for chronic inflamma-
model of chronic alcohol-induced pancreatitis in the rat. Am J Physiol tion and cancer development in the pancreas. Am J Gastroenterol 101:
Gastrointest Liver Physiol 280: G1178 –G1186, 2001. 160 –162, 2006.

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00372.2015 • www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi at Univ De Antioquia (200.024.017.012) on February 21, 2021.
Themes
G354 PANCREATITIS MODELS

109. Masamune A, Kikuta K, Watanabe T, Satoh K, Satoh A, Shimose- 130. Perides G, Laukkarinen JM, Vassileva G, Steer ML. Biliary acute
gawa T. Pancreatic stellate cells express Toll-like receptors. J Gastro- pancreatitis in mice is mediated by the G-protein-coupled cell surface
enterol 43: 352–362, 2008. bile acid receptor Gpbar1. Gastroenterology 138: 715–725, 2010.
110. Mizunuma T, Kawamura S, Kishino Y. Effects of injecting excess 131. Perides G, Tao X, West N, Sharma A, Steer ML. A mouse model of
arginine on rat pancreas. J Nutr 114: 467–471, 1984. ethanol dependent pancreatic fibrosis. Gut 54: 1461–1467, 2005.
111. Mooren F, Hlouschek V, Finkes T, Turi S, Weber IA, Singh J, 132. Perides G, van Acker GJ, Laukkarinen JM, Steer ML. Experimental
Domschke W, Schnekenburger J, Kruger B, Lerch MM. Early acute biliary pancreatitis induced by retrograde infusion of bile acids into
changes in pancreatic acinar cell calcium signaling after pancreatic duct the mouse pancreatic duct. Nat Protoc 5: 335–341, 2010.
obstruction. J Biol Chem 278: 9361–9369, 2003. 133. Philip B, Roland CL, Daniluk J, Liu Y, Chatterjee D, Gomez SB, Ji
111a.Mouret J. Contribution a l’étude des cellules glandulaires (pancreas). J B, Huang H, Wang H, Fleming JB, Logsdon CD, Cruz-Monserrate
Anat Physiol 31: 221–236, 1895. Z. A high-fat diet activates oncogenic Kras and COX2 to induce
112. Muili KA, Wang D, Orabi AI, Sarwar S, Luo Y, Javed TA, Eisses JF, development of pancreatic ductal adenocarcinoma in mice. Gastroenter-
Mahmood SM, Jin S, Singh VP, Ananthanaravanan M, Perides G, ology 145: 1449 –1458, 2013.
Williams JA, Molkentin JD, Husain SZ. Bile acids induce pancreatic 134. Ponnappa BC, Marciniak R, Schneider T, Hoek JB, Rubin E. Ethanol
acinar cell injury and pancreatitis by activating calcineurin. J Biol Chem consumption and susceptibility of the pancreas to cerulein-induced pan-
288: 570 –580, 2013. creatitis. Pancreas 14: 150 –157, 1997.
113. Muller S, Kaiser H, Kruger B, Fitzner B, Lange F, Bock CN, Nizze 135. Popper HL, Necheles H, Russell KC. Transition of pancreatic edema
H, Ibrahim SM, Fuellen G, Wolkenhauer O, Jaster R. Age-dependent into pancreatic necrosis. Surg Gynecol Obstet 87: 79 –82, 1948.
effects of UCP2 deficiency on experimental acute pancreatitis in mice. 136. Rakonczay Z Jr, Hegyi P, Dosa S, Ivanyi B, Jarmay K, Biczo G,
PLoS One 9: e94494, 2014. Hracsko Z, Varga IS, Karg E, Kaszaki J, Varro A, Lonovics J, Boros
114. Murayama KM, Barent BL, Gruber M, Brooks A, Eliason S, Brunt I, Gukovsky I, Gukovskaya AS, Pandol SJ, Takacs T. A new severe
EM, Smith GS. Characterization of a novel model of pancreatic fibrosis acute necrotizing pancreatitis model induced by L-ornithine in rats. Crit
and acinar atrophy. J Gastrointest Surg 3: 418 –425, 1999. Care Med 36: 2117–2127, 2008.
115. Murtaugh LC, Keefe MD. Regeneration and repair of the exocrine 137. Rao KN, Eagon PK, Okamura K, Van Thiel DH, Gavaler JS, Kelly
pancreas. Annu Rev Physiol 77: 229 –249, 2015. RH, Lombardi B. Acute hemorrhagic pancreatic necrosis in mice.
116. Neuschwander-Tetri BA, Bridle KR, Wells LD, Marcu M, Ramm Induction in male mice treated with estradiol. Am J Pathol 109: 8 –14,
GA. Repetitive acute pancreatic injury in the mouse induces procollagen 1982.
alpha1 (I) expression colocalized to pancreatic stellate cells. Lab Invest 138. Rao KN, Tuma J, Lombardi B. Acute hemorrhagic pancreatic necrosis
80: 143–150, 2000. in mice. Intraparenchymal activation of zymogens, and other enzyme
117. Neuschwander-Tetri BA, Burton FR, Presti ME, Britton RS, Janney changes in pancreas and serum. Gastroenterology 70: 720 –726, 1976.
CG, Garvin PR, Brunt EM, Galvin NJ, Poulos JE. Repetitive self- 139. Robertson JS. The pancreatic lesion in adult mice, infected with a strain
limited acute pancreatitis induces pancreatic fibrogenesis in the mouse. of pleurodynia virus. I. Electron microscopical observations. Aust J Exp
Biol Med Sci 32: 393–409, 1954.
Dig Dis Sci 45: 665–674, 2000.
140. Romac JM, Shahid RA, Choi SS, Karaca GF, Westphalen CB, Wang
118. Nevalainen TJ, Aho HJ. Standards of morphological evaluation and
TC, Liddle RA. Pancreatic secretory trypsin inhibitor I reduces the
histological grading in experimental acute pancreatitis. Eur Surg Res 24,
severity of chronic pancreatitis in mice overexpressing interleukin-1␤ in
Suppl 1: 14 –23, 1992.
the pancreas. Am J Physiol Gastrointest Liver Physiol 302: G535–G541,
119. Niederau C, Ferrell LD, Grendell JH. Caerulein-induced acute necro-
2012.
tizing pancreatitis in mice: protective effects of proglumide, benzotript,
141. Sah RP, Dudeja V, Dawra RK, Saluja AK. Cerulein-induced chronic
and secretin. Gastroenterology 88: 1192–1204, 1985.
pancreatitis does not require intra-acinar activation of trypsinogen in
120. Niederau C, Luthen R, Niederau MC, Grendell JH, Ferrell LD.
mice. Gastroenterology 144: 1076 –1085.e2, 2013.
Acute experimental hemorrhagic-necrotizing pancreatitis induced by
142. Sah RP, Garg P, Saluja AK. Pathogenic mechanisms of acute pancre-
feeding a choline-deficient, ethionine-supplemented diet. Methodology atitis. Curr Opin Gastroenterol 28: 507–515, 2012.
and standards. Eur Surg Res 24, Suppl 1: 40 –54, 1992. 143. Saluja AK, Dudeja V. Relevance of animal models of pancreatic cancer
121. Ohashi S, Nishio A, Nakamura H, Asada M, Tamaki H, Kawasaki K, and pancreatitis to human disease. Gastroenterology 144: 1194 –1198,
Fukui T, Yodoi J, Chiba T. Overexpression of redox-active protein 2013.
thioredoxin-1 prevents development of chronic pancreatitis in mice. 144. Sarles H, Lebreuil G, Tasso F, Figarella C, Clemente F, Devaux MA,
Antioxid Redox Signal 8: 1835–1845, 2006. Fagonde B, Payan H. A comparison of alcoholic pancreatitis in rat and
122. Ohshio G, Saluja A, Steer ML. Effects of short-term pancreatic duct man. Gut 12: 377–388, 1971.
obstruction in rats. Gastroenterology 100: 196 –202, 1991. 145. Schmidt J, Rattner DW, Lewandrowski K, Compton CC, Mandavilli
123. Orabi AI, Shah AU, Muili K, Luo Y, Mahmood SM, Ahmad A, Reed U, Knoefel WT, Warshaw AL. A better model of acute pancreatitis for
A, Husain SZ. Ethanol enhances carbachol-induced protease activation evaluating therapy. Ann Surg 215: 44 –56, 1992.
and accelerates Ca2⫹ waves in isolated rat pancreatic acini. J Biol Chem 146. Sendler M, Beyer G, Mahajan UM, Kauschke V, Maertin S, Schur-
286: 14090 –14097, 2011. mann C, Homuth G, Volker U, Volzke H, Halangk W, Wartmann T,
124. Owyang C, Logsdon CD. New insights into neurohormonal regulation Weiss FU, Hegyi P, Lerch MM, Mayerle J. Complement component 5
of pancreatic secretion. Gastroenterology 127: 957–969, 2004. mediates development of fibrosis, via activation of stellate cells, in 2
125. Pandol SJ, Lugea A, Mareninova OA, Smoot D, Gorelick FS, Guk- mouse models of chronic pancreatitis. Gastroenterology 149: 765–
ovskaya AS, Gukovsky I. Investigating the pathobiology of alcoholic 776.e10, 2015.
pancreatitis. Alcohol Clin Exp Res 35: 830 –837, 2011. 147. Senninger N, Moody FG, Coelho JC, Van Buren DH. The role of
126. Pandol SJ, Periskic S, Gukovsky I, Zaninovic V, Jung Y, Zong Y, biliary obstruction in the pathogenesis of acute pancreatitis in the opos-
Solomon TE, Gukovskaya AS, Tsukamoto H. Ethanol diet increases sum. Surgery 99: 688 –693, 1986.
the sensitivity of rats to pancreatitis induced by cholecystokinin octapep- 148. Shaheen NJ, Hansen RA, Morgan DR, Gangarosa LM, Ringel Y,
tide. Gastroenterology 117: 706 –716, 1999. Thiny MT, Russo MW, Sandler RS. The burden of gastrointestinal and
127. Pandol SJ, Saluja AK, Imrie CW, Banks PA. Acute pancreatitis: bench liver diseases, 2006. Am J Gastroenterol 101: 2128 –2138, 2006.
to the bedside. Gastroenterology 132: 1127–1151, 2007. 149. Singh S, Bhardwaj U, Verma SK, Bhalla A, Gill K. Hyperamylasemia
128. Parenti DM, Steinberg W, Kang P. Infectious causes of acute pancre- and acute pancreatitis following anticholinesterase poisoning. Hum Exp
atitis. Pancreas 13: 356 –371, 1996. Toxicol 26: 467–471, 2007.
129. Peery AF, Dellon ES, Lund J, Crockett SD, McGowan CE, Bulsie- 150. Stumpf F, Algul H, Thoeringer CK, Schmid RM, Wolf E, Schneider
wicz WJ, Gangarosa LM, Thiny MT, Stizenberg K, Morgan DR, MR, Dahlhoff M. Metamizol relieves pain without interfering with
Ringel Y, Kim HP, Dibonaventura MD, Carroll CF, Allen JK, Cook cerulein-induced acute pancreatitis in mice. Pancreas 45: 572–578, 2016.
SF, Sandler RS, Kappelman MD, Shaheen NJ. Burden of gastrointes- 151. Takacs T, Czako L, Jarmay K, Farkas G Jr, Mandi Y, Lonovics J.
tinal disease in the United States: 2012 update. Gastroenterology 143: Cytokine level changes in L-arginine-induced acute pancreatitis in rat.
1179 –1187.e1– e3, 2012. Acta Physiol Hung 84: 147–156, 1996.

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00372.2015 • www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi at Univ De Antioquia (200.024.017.012) on February 21, 2021.
Themes
PANCREATITIS MODELS G355
152. Takacs T, Czako L, Morschl E, Laszlo F, Tiszlavicz L, Rakonczay Z 169. Weaver C, Bishop AE, Polak JM. Pancreatic changes elicited by
Jr, Lonovics J. The role of nitric oxide in edema formation in L-arginine- chronic administration of excess L-arginine. Exp Mol Pathol 60: 71–87,
induced acute pancreatitis. Pancreas 25: 277–282, 2002. 1994.
153. Takacs T, Farkas G Jr, Czako L, Jarmay K, Mandi Y, Lonovics J. 170. Wen L, Voronina S, Javed MA, Awais M, Szatmary P, Latawiec D,
Time-course changes in serum cytokine levels in two experimental acute Chvanov M, Collier D, Huang W, Barrett J, Begg M, Stauderman K,
pancreatitis models in rats. Res Exp Med (Berl) 196: 153–161, 1996. Roos J, Grigoryev S, Ramos S, Rogers E, Whitten J, Velicelebi G,
154. Tani S, Itoh H, Okabayashi Y, Nakamura T, Fujii M, Fujisawa T, Dunn M, Tepikin AV, Criddle DN, Sutton R. Inhibitors of ORAI1
Koide M, Otsuki M. New model of acute necrotizing pancreatitis prevent cytosolic calcium-associated injury of human pancreatic acinar
induced by excessive doses of arginine in rats. Dig Dis Sci 35: 367–374, cells and acute pancreatitis in 3 mouse models. Gastroenterology 149:
1990. 481–492.e7, 2015.
155. Tardini A, Anversa P, Bordi C, Bertaccini G, Impicciatore M. 171. Whitcomb DC. Mechanisms of disease: advances in understanding the
Ultrastructural and biochemical changes after marked caerulein stimula- mechanisms leading to chronic pancreatitis. Nat Clin Pract Gastroen-
tion of the exocrine pancreas in the dog. Am J Pathol 62: 35–56, 1971. terol Hepatol 1: 46 –52, 2004.
156. Terry TR, Grant DA, Hermon-Taylor J. Intraduct enterokinase is 172. Willemer S, Elsasser HP, Adler G. Hormone-induced pancreatitis. Eur
lethal in rats with experimental bile-salt pancreatitis. Br J Surg 74: Surg Res 24, Suppl 1: 29 –39, 1992.
40 –43, 1987. 173. Williams JA, Krawitz B, Hou Y. Caerulein. In: Pancreapedia: Exo-
157. Thrower EC, Gorelick FS, Husain SZ. Molecular and cellular mech- crine Pancreas Knowledge Base. doi: 10.3998/panc.2014.15, 2014.
anisms of pancreatic injury. Curr Opin Gastroenterol 26: 484 –489, 174. Williams JA, Korc M, Dormer RL. Action of secretagogues on a new
2010. preparation of functionally intact, isolated pancreatic acini. Am J Physiol
158. Treiber M, Neuhofer P, Anetsberger E, Einwachter H, Lesina M, Endocrinol Metab Gastrointest Physiol 235: E517–E524, 1978.
Rickmann M, Liang S, Kehl T, Nakhai H, Schmid RM, Algul H. 175. Williams JA, Sans MD, Tashiro M, Schafer C, Bragado MJ, Dab-
Myeloid, but not pancreatic, RelA/p65 is required for fibrosis in a mouse rowski A. Cholecystokinin activates a variety of intracellular signal
model of chronic pancreatitis. Gastroenterology 141: 1473–1485, transduction mechanisms in rodent pancreatic acinar cells. Pharmacol
1485.e1– e7, 2011. Toxicol 91: 297–303, 2002.
159. Trivedi CD, Pitchumoni CS. Drug-induced pancreatitis: an update. J
176. Wittel UA, Wiech T, Chakraborty S, Boss B, Lauch R, Batra SK,
Clin Gastroenterol 39: 709 –716, 2005.
Hopt UT. Taurocholate-induced pancreatitis: a model of severe necro-
160. Tsukamoto H, Towner SJ, Yu GS, French SW. Potentiation of
tizing pancreatitis in mice. Pancreas 36: e9 –e21, 2008.
ethanol-induced pancreatic injury by dietary fat. Induction of chronic
177. Yadav D, Lowenfels AB. The epidemiology of pancreatitis and pancre-
pancreatitis by alcohol in rats. Am J Pathol 131: 246 –257, 1988.
atic cancer. Gastroenterology 144: 1252–1261, 2013.
161. Vaquero E, Molero X, Tian X, Salas A, Malagelada JR. Myofibroblast
178. Yadav D, Timmons L, Benson JT, Dierkhising RA, Chari ST.
proliferation, fibrosis, and defective pancreatic repair induced by cyclo-
sporin in rats. Gut 45: 269 –277, 1999. Incidence, prevalence, and survival of chronic pancreatitis: a population-
162. Volzke H, Baumeister SE, Alte D, Hoffmann W, Schwahn C, Simon based study. Am J Gastroenterol 106: 2192–2199, 2011.
P, John U, Lerch MM. Independent risk factors for gallstone formation 179. Yadav D, Whitcomb DC. The role of alcohol and smoking in pancre-
in a region with high cholelithiasis prevalence. Digestion 71: 97–105, atitis. Nat Rev Gastroenterol Hepatol 7: 131–145, 2010.
2005. 180. Yamaguchi T, Kihara Y, Taguchi M, Nagashio Y, Tashiro M,
163. Vonlaufen A, Phillips PA, Xu Z, Zhang X, Yang L, Pirola RC, Nakamura H, Otsuki M. Persistent destruction of the basement mem-
Wilson JS, Apte MV. Withdrawal of alcohol promotes regression while brane of the pancreatic duct contributes to progressive acinar atrophy in
continued alcohol intake promotes persistence of LPS-induced pancreatic rats with experimentally induced pancreatitis. Pancreas 31: 365–372,
injury in alcohol-fed rats. Gut 60: 238 –246, 2011. 2005.
164. Vonlaufen A, Xu Z, Daniel B, Kumar RK, Pirola R, Wilson J, Apte 181. Zambirinis CP, Pushalkar S, Saxena D, Miller G. Pancreatic cancer,
MV. Bacterial endotoxin: a trigger factor for alcoholic pancreatitis? inflammation, and microbiome. Cancer J 20: 195–202, 2014.
Evidence from a novel, physiologically relevant animal model. Gastro- 182. Zhang J, Rouse RL. Histopathology and pathogenesis of caerulein-,
enterology 133: 1293–1303, 2007. duct ligation-, and arginine-induced acute pancreatitis in Sprague-Daw-
165. Vuorinen T, Kallajoki M, Hyypia T, Vainionpaa R. Coxsackievirus ley rats and C57BL6 mice. Histol Histopathol 29: 1135–1152, 2014.
B3-induced acute pancreatitis: analysis of histopathological and viral 183. Zhao HF, Ito T, Gibo J, Kawabe K, Oono T, Kaku T, Arita Y, Zhao
parameters in a mouse model. Br J Exp Pathol 70: 395–403, 1989. QW, Usui M, Egashira K, Nawata H. Anti-monocyte chemoattractant
166. Wachstein M, Meisel E. Equal effectiveness of L and D-ethionine in protein 1 gene therapy attenuates experimental chronic pancreatitis in-
producing tissue damage in rats and mice. Proc Soc Exp Biol Med 82: duced by dibutyltin dichloride in rats. Gut 54: 1759 –1767, 2005.
70 –72, 1953. 184. Zhao JB, Liao DH, Nissen TD. Animal models of pancreatitis: can it be
167. Walker NI. Ultrastructure of the rat pancreas after experimental duct translated to human pain study? World J Gastroenterol 19: 7222–7230,
ligation. I. The role of apoptosis and intraepithelial macrophages in 2013.
acinar cell deletion. Am J Pathol 126: 439 –451, 1987. 185. Zheng H, Chen D, Zhang J, Tian Y. Involvement of M3 cholinergic
168. Watanabe S, Abe K, Anbo Y, Katoh H. Changes in the mouse exocrine receptor signal transduction pathway in regulation of the expression of
pancreas after pancreatic duct ligation: a qualitative and quantitative chemokine MOB-1, MCP-1 genes in pancreatic acinar cells. J Huazhong
histological study. Arch Histol Cytol 58: 365–374, 1995. Univ Sci Technolog Med Sci 24: 140 –143, 157, 2004.

AJP-Gastrointest Liver Physiol • doi:10.1152/ajpgi.00372.2015 • www.ajpgi.org


Downloaded from journals.physiology.org/journal/ajpgi at Univ De Antioquia (200.024.017.012) on February 21, 2021.

You might also like