Revision of Experimental acute pancreatitis: In vitro models from Wed, 2015-10-28 10:59

(1) Veterans Affairs Greater Los Angeles Healthcare System and Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, CA 90073 USA (2) Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC and the University of Pittsburgh School of Medicine, Pittsburgh, PA 15224 USA
sohail.husain@chp.edu

Entry Version: 

Version 1.0, July 18, 2015

Citation: 

Mareninova, Olga A. Orabi, Abrahim I. Husain, Sohail Z. (2015). Experimental acute pancreatitis: In vitro models.
Pancreapedia: Exocrine Pancreas Knowledge Base, DOI: 10.3998/panc.2015.27

1. Introduction

Acute pancreatitis is an extremely painful and life-threatening inflammatory disease of the exocrine pancreas (57, 100). A sobering point for both clinicians and researchers is that the treatment of acute pancreatitis remains largely supportive. Further, there is a lack of therapies that target primary mechanisms underlying the initiation or propagation of the disease. Thus reliable, relevant, and, importantly, convenient experimental animal models that resemble the human disease are crucial to developing an understanding of the pathobiology of pancreatitis (34, 35, 104, 105, 108, 110). In this chapter, we will review the current in vitro (i.e. ex vivo) models that serve as surrogates for experimental acute pancreatitis. We will specifically discuss: (1) the standard process of preparing pancreatic acinar cells or pancreatic tissue components; (2) assays for assessing in vitro injury and inflammatory precursors; and (3) the array of non-alcoholic and alcoholic in vitro models of pancreatitis.

The pancreatic acinar cell is the main parenchymal cell of the pancreas. It comprises roughly 90% of the pancreatic parenchyma, and functions to synthesize and secrete digestive enzymes in response to hormonal stimulation (24, 30, 36, 48).

The acinar cell is considered the initiating site of pancreatic injury, leading to pancreatitis, and thus in vitro preparations of acinar cells have been used for decades to define the molecular events that occur during the early stages of the disease (1, 49, 134). The advantage of these models are that they provide a high throughput (or at least rapid) system to examine whether cellular pathways or molecular targets modulate injurious in vitro corollaries to in vivo events during pancreatitis, including aberrant Ca2+ signaling, activation of digestive proteases, NF-kB activation, mitochondrial dysfunction, and cell death through apoptosis or necrosis. A disadvantage is that these systems lack the full inflammatory or systemic components and, therefore, subsequent in vivo validation of in vitro findings is crucial.

Recent use of adenovirus-mediated gene transfer has enabled researchers to manipulate acinar cell function in the presence of pathological agents (89, 146). Another powerful genetic approach for studying pancreatitis in vitro is to isolate acinar cells from the pancreas of gene-targeted knockout or transgenic mice (43, 51, 82, 144).

2. Acinar Cell Preparations

Single, Double, and Large Cluster Acinar Cell Preparations

Isolated pancreatic acini and acinar cells can be prepared from rat, mouse, and guinea pig pancreas using a collagenase digestion protocol (13, 93, 101, 116, 140). Depending on the stringency of the isolation protocol, single, double, and large cluster acinar cells are obtained (Figure 1A-B). The greatest determinant in the stringency of acinar cell preparation is the concentration and duration of collagenase digestion. Nonetheless, there are several collagenases to choose from, including Sigma Type II (142), IV or V (80), and Worthington type IV (29). A newer collagenase P from Roche can be used to prepare smaller acini for electrophysiology (75, 147). Liberase (Roche) is another option for acinar cell isolation. Some authorities use collagenase NB1 (Serva) to perform human islet cell isolation, which also yields acinar cells (and duct cells) for experimental use (12, 63). Acinar size and integrity are highly dependent on the type of collagenase used and the application of shearing forces (99). After digestion of pancreatic tissue, acinar cells can be purified away from ducts, islets, and blood vessels by filtration and bovine serum albumin (BSA) density sedimentation. Following this method, acini can be maintained in culture for 24-48 hours but start to lose their polarity and secretory capability after several hours.

Lobules

To assess the direct as well as indirect effects of agonists on acinar cell secretion, in vitro preparations should ideally contain not only acinar cells but also nerves and islets.


Figure 1. In vitro preparations of the pancreas include (A) single acinar cell preparations, (B) acini, (C) pancreatic lobules, (D) pancreatic organoids, or (E) pancreatic slices. Adapted from (99), (93), (113), (10), (45), respectively. Republished with permission.

For this reason pancreatic lobules are useful (Figure 1C). In the original description by Scheele and colleagues, pancreatic lobules were spread apart by injecting Krebs-Ringer bicarbonate (KRB) buffer into the loose connective tissue of the pancreas and then individually excised by micro-dissection under a stereomicroscope (27, 113). This procedure minimizes damage to acinar cells since most of the surgical trauma is limited to ducts and vessels. The excised lobules preserve the overall acinar architecture of the tissue and their small size and allows for easy penetration of oxygen and solutes from the incubation medium. Following this method, lobules can be maintained for several hours in culture (6, 7, 66, 114).

Organoids

The most recent advance in studying pancreatic physiology in vitro involves the generation of pancreatic organoids (10, 46) (Figure 1D). By definition, organoids are three-dimensional organ buds which arise from stem cells. With the use of growth factors, stem cell populations used to develop organoids can be coaxed into forming balls of terminally differentiated cells that self-organize into distinctive layers. As described by Boj and colleagues, pancreatic organoids can be rapidly generated from resected pancreatic tumors and biopsies following manual digestion with collagenase II and seeded in growth factor-reduced Matrigel (10). Conditioning the medium with the growth factor R-spondin promotes a predominantly duct cell population. These pancreatic organoids survive cryopreservation and exhibit ductal- and disease stage-specific characteristics. Further, pancreatic organoids from wild-type mice accurately recapitulated physiologically relevant aspects of disease progression in vitro. Following orthotopic transplantation, pancreatic organoids were capable of regenerating normal ductal architectures. This technique is particularly useful for studying duct cell phenotypes (10).

Pancreas Slice

To preserve the integrity of the pancreatic milieu for at least two days in culture, the novel method of culturing pancreas slices is useful (44, 45) (Figure 1E). This technique allows for both in situ imaging of cellular events relevant to pancreatitis and genetic manipulation. To obtain a pancreas slice, Gaisano and colleagues gently infused a low melting agarose gel into the pancreatic duct of an anesthetized mouse via a transduodenal puncture and cannulation of the common bile duct (44, 45). The pancreas was then excised and trimmed. The agarose renders the pancreas firm enough to then slice, using a vibratome, at a thickness of 80-140 µm. Moreover, agarose is porous and thus provides free exchange of tissue with buffer, ensuring optimal health in culture for up to two days. Further, this technique permits transfection of cells as well as real time imaging.

Acinar cell lines

The most commonly used cell lineto study the exocrine pancreas is the rat pancreatic acinar cell line AR42J (Figure 2). These cells were derived from a transplantable tumor for the rat exocrine pancreas. The AR42J cells differ from primary pancreatic acinar cells in at least two ways: (1) they proliferate rapidly; and (2) although they synthesize, store, and secrete digestive enzymes, they express atypical receptors and conduct atypical inositol phosphate metabolism and cytoskeleton rearrangement (33). Dexamethasone favors their differentiation toward the acinar phenotype, including agonist-stimulated Ca2+ signaling (5, 15, 67, 124). The cell line is incubated for 48-72 hours in culture medium supplemented with 100 nM dexamethasone prior to experimental treatment or induction. AR42J cells are easily cultured in a RPMI 1640 medium supplemented with glutamine, FBS, and antibiotics at 370C under a humidified condition of 95% air and 5% CO2. AR42J cells can be routinely plated at a density of 105 cells/ml in 75 cm2 flasks and cultured for 7-10 days. 


Figure 2. Morphological characteristics of the AR42J acinar cell line. AR42J cells primed with dexamethasone (100 nM) and visualized by (A) light microscopy using a 20X objective or (B) electron microscopy (arrow heads point to zymogen granules). Adapted from (33) and (20), respectively. Republished with permission.

A less common derived acinar cell line is the 266-6. This cell line is derived from young adult mouse tumors induced with elastase I/SV-40 T-antigen fusion gene. Robert Hammer first described the line in 1985 (97). 266-6 cells retain a partially differentiated phenotype and express several digestive enzymes. They respond to carbachol and cholecystokinin (CCK) but do not respond to substance P, secretin, or vasoactive intestinal peptide (VIP). The culture method is the same as that described for AR42J cells, except that there is no dexamethasone priming.

3. Assays for In Vitro Surrogates of Pancreatitis

Ca2+ Signaling

Pancreatic acinar cells have served as an epithelial cell model for examining Ca2+ signaling for decades (Figure 3). Consistent with the polarized nature of acinar cells, Ca2+ signals in these cells exhibit highly organized spatial characteristics (103). Most agonist-stimulated Ca2+ signals in acinar cells initiate in the apical region and propagate to the basolateral region (31, 48, 52). Single cell imaging of Ca2+ signals involves the use of fluorescent Ca2+ dyes and confocal microscopy. A number of Ca2+ sensing dyes are available, depending on the needs of the researcher (37, 77, 94, 127). The simplest dyes exhibit signature fluorescent properties upon binding Ca2+; they are excited by a certain wavelength of light and emit photons at a certain emission wavelength (i.e. Fluo-3AM, Fluo-4AM). Ratiometric dyes (i.e. Fura-2), on the other hand, exhibit distinct spectral shifts upon Ca2+ binding, such that the Ca2+-free form is excited maximally at 380 nm while the Ca2+ bound form is excited maximally at 340 nm. Both states emit peak fluorescence at 510 nm. 


Figure 3. Typical Ca2+ transients upon stimulation with supraphysiologic concentrations of carbachol (1 μM) or physiologic concentrations of caerulein (10 pM). Changes in whole cell Ca2+ were measured by time-lapse confocal microscopy using the Ca2+ dye Fluo-4AM (5 μM). Images are represented in pseudocolor with a color scale. (A) From left to right; bright field view of an acinus labeled at the apical and basolateral regions of interest. Upon stimulation with physiologic carbachol (1 μM; Ach analog), subsequent images show the initiation of the Ca2+ signal in the apical region followed by propagation to the basal region. (B) Each paneled image (1-4), corresponds to a frame along a representative tracing of change in fluorescence over time for each region of interest. (C-D) Oscillating Ca2+ signals are observed in response to low-dose caerulein (10 pM; CCK analog). These figures were originally published in the J Biol Chem (96) and (106). Republished with permission.

Cells are loaded with the Ca2+ dye of choice, allowed to adhere to glass coverslips, and excited with the agonist of choice, while collecting real time images usually with a laser scanning confocal microscope (93).

Intra-Acinar Protease Activation

Premature intra-cellular activation of digestive proteases has long been thought to represent an early, initiating event in the pathogenesis of pancreatitis. The traditional method for examining intra-acinar protease activation involves probing pancreatic acinar cell lysates with a fluorogenic substrate for the protease of interest (58, 110, 118). The readout is obtained from a fluorimeter (e.g. a fluorescent plate reader or cuvette system, also termed a fluorimeter) in the form of a kinetic plot. These data can be normalized to total protein content or total DNA in order to control for cell loading. Since the initial description of these fluorogenic substrates in 1983 (64, 65), bisamide derivatives of rhodamine 110 have been used as a sensitive and selective substrate for activated protease measurements. Proteolytic selectivity is achieved by using specific benzyloxycarbonyl-peptides. The tripeptide derivative bis-(CBZ-Ile-Pro-Arg)-R110 (BZiPAR) has been successfully used by some groups to measure trypsinogen activation by live microscopy (54, 55, 62, 105).

NF-κB Translocation

NF-κB activation is thought to be an early and critical component of the inflammatory response during acute pancreatitis (104). Traditional methods for examining NF-κB activity in vitro include protein determination of NF-κB pathway markers (i.e. phosphorylated IκB; p65 nuclear translocation; IKK upregulation), electromobility shift assay (EMSA), and immunohistochemistry for phosphorylated p65 (51, 125). Newer techniques include the transfection (or usually infection via viral vectors in pancreatic cells) of NF-κB-luciferase reporters (Figure 4). With these techniques, binding of NF-κB subunits to a nuclear response element drives transcription of the luminescent protein luciferase. The commonly used luciferase reporters are firefly (21) and renilla (68) luciferases. The development of secreted luciferases such as gaussia (Gluc), secreted alkaline phosphatase (SEAP), and cypridina allows for serial determination from the media of NF-κB activity over time (3, 41, 87, 126).

Mitochondrial Damage

Mitochondrial dysfunction has been shown to play a critical role in the pathogenesis of pancreatic acinar cell injury, resulting in pancreatitis (73). Manifestations of mitochondrial dysfunction in pancreatitis include loss of mitochondrial inner membrane potential (DYm), generation of reactive oxygen species (ROS), release of the apoptosis, or programmed cell death mediator cytochrome c into the cytosol, and failure of ATP production; the events lead to varying degrees of acinar cell necrosis or apoptosis (92). Recent data show that preventing mitochondrial damage improves several aspects of pancreatitis and ameliorates disease severity (85, 119).

The effect of pancreatitis on DYm can be measured in isolated acinar cells using the DYm-sensitive fluorescence probe tetramethylrhodamine methyl ester (TMRM), which is a lipophilic cation dye whose accumulation in mitochondria is proportional to the amount of DYm. After preincubation with an agonist, cells are loaded with 1 μM TMRM for 10-20 min at 370C and transferred to a fluorimeter to measure fluorescence intensity at 543 nm/570 nm (90, 119). DYm can also be detected using another DYm-sensitive fluorescence probe JC-1, which exists as a green monomer at low DYm. Because JC-1 forms red fluorescent J-aggregates at higher potentials, the ratio between red (550 nm/600 nm) and green (485 nm/535 nm) fluorescence is used to monitor changes in DYm. A loss of DYm leads to depletion of intracellular ATP and subsequent necrosis. ATP levels in pancreatic acinar cells can be detected using a luciferin/luciferase luminescence-based assay that is normalized to protein content.


Figure 4. Schematic of the NF-κB-luciferase adenoviral construct. The NF-κB-luciferase adenoviral construct contains six tandem-repeat transcription factor response elements, a minimal promoter, and a luciferase coding region. Binding of NF-κB subunits to a nuclear response element drives transcription of the luminescent protein luciferase. Originally published in the J Biol Chem (95). Republished with permission.

Permeabilization of the mitochondrial outer membrane occurs through opening of the mitochondrial permeability transition pore (MPTP), and the event is integral to apoptosis in pancreatitis. MPTP opening and subsequent mitochondrial outer membrane permeabilization result in the release of the mitochondrial resident protein cytochrome c into the cytosol. The technique to detect cytochrome c release within acinar cells relies on examining immunoblots against cytochrome c from cellular fractions of mitochondria-enriched membrane versus cytosolic fractions (74, 91).

The mitochondria within acinar cells are highly susceptible to oxidative damage from ROS, and they in turn also serve as primary generators of ROS when the electronic transport chain within the inner mitochondrial membrane is perturbed (usually with loss of DYm). ROS can act as a molecular trigger of cytochrome c release and death responses in pancreatic acinar cells, thus also demonstrating the cross-talk in the mitochondria between necrosis and apoptosis triggers (85). Intracellular ROS levels (both mitochondrial and non-mitochondrial) are detected using 2,7-dichlorofluorescein (DCF) (91). ROS that is selectively generated by the mitochondria can be monitored by labeling the cells with the mitochondrial ROS-sensitive rhodamine-based fluorescent dye DHR123. Mitochondrial localization of DHR123 can be confirmed by co-staining the cells with the mitochondrial specific marker MitoTracker Red (CMXRos). Proper analysis of ROS production in living cells requires the combined use of several fluorescent ROS probes in parallel experiments, assessment of non-ROS related parameters that can induce artifacts (e.g. DY, pH), and the inclusion of adequate control conditions. For example, a common positive control that is known to cause the generation of mitochondrial ROS is rotenone, which inhibits complex I of the electron transport chain. A negative control is the mitochondrial uncoupler carbonyl cyanide m-chlorophenylhydrazone (CCCP), which blocks mitochondrial ROS production.

Cell Injury

The three most common assays used to assess acinar cell injury include: (1) lactate dehydrogenase (LDH) release; (2) propidium iodide (PI) uptake; and (3) reduction of MTT(3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide). LDH catalyzes the interconversion of pyruvate to lactate and NADH to NAD+ (72). Elevated levels of LDH are indicative of tissue injury and breakdown. LDH can be measured using colorimetric assays supplied by Promega (cat #G1780) (93). PI is a high affinity DNA-binding dye that is effectively excluded from live cells (59, 79, 123). Dead or dying cells have compromised plasma membranes and thereby allow the leakage of PI, which then enters the nucleus and binds to DNA. MTT reduction is a measure of mitochondrial function and cell viability (8, 16, 81). MTT is reduced to insoluble formazan by mitochondrial dehydrogenases. Water insoluble formazan can be solubilized using isopropanol or other solvents. The dissolved material is measured spectrophotometrically, yielding absorbance as a function of the concentration of the converted dye.

4. Non-Alcholic Models

Secretagogues

The peptide hormone CCK, or its analog caerulein, has been used in in vitro models to reproducibly induce acute pancreatitis-like responses in acinar cells (14, 58, 63, 111, 112, 134, 140). Pancreatic acinar cells express high and low affinity CCK receptors (CCKRs), which are activated by low and high concentrations of CCK, respectively (76, 139). Low concentrations in the picomolar range bind to high affinity CCK receptors and maximally stimulate physiological acinar cell enzyme secretion (138). High, or supra-physiological, concentrations in the nanomolar range bind to low affinity CCK receptors and result in a relative reduction in the secretory response, a phenomenon that is thought to be pathological to the cell because it leads to the retention of the prematurely activated proteases and their missorting (62).

The activation of digestive proteases requires a rise in cytosolic Ca2+, which occurs through release from intracellular Ca2+ pools (primarily the endoplasmic reticulum) that are gated by IP3 receptors and ryanodine receptors (48, 60, 105, 110). Another consequence of supraphysiological CCK, seen both in vitro and in vivo, is the emergence of large intra-acinar vacuoles (39, 105, 120).

There are other CCK analogues which do not lead to protease activation or pancreatitis, even at high concentrations because they elicit distinct phenotypic responses and distinct cell signals. They include the O-phenyl-methyl-ester analog of CCK (OPE) and JMV-180 (76, 139). These agonists can serve as physiological controls to differentiate between pathological signals. The agonist bombesin (also known as gastrin-related peptide) causes intra-acinar protease activation but no acinar cell injury because, unlike CCK, bombesin does not cause activated proteases to be retained in the acinar cell (34). Other secretagogues that stimulate acinar cell enzyme secretion include secretin, VIP, and pituitary adenylate cyclase-activating peptide (PACAP) (48, 115, 117).

Several investigations have questioned whether CCK hyperstimulation is relevant to human acinar cells (109, 112). Whereas CCK receptors are abundant on murine acinar cells, they have little to no expression in the human acinar cell (50, 133). Except for a notable recent report (86), CCK failed to elicit a Ca2+ signal or a secretory response in isolated human acini (50, 78, 122). By contrast, acetylcholine or its long-acting analog carbachol stimulates robust physiological and pathological (at high millimolar concentrations) responses in acinar cells from mouse, rat, and man (70, 98). Several clinical correlates of pancreatitis are associated with cholinergic overload, from exposure to scorpion toxin or organophosphates (which would prevent the degradation of acetylcholine by inhibiting endogenous acetylcholinesterases) (107, 121, 128, 132).

Bile Acids

The most common cause of acute pancreatitis is impaction of gallstones or sludge in the distal common bile duct, a situation called biliary pancreatitis (4, 69, 71, 129). There are two hotly debated and non-mutually exclusive theories for biliary pancreatitis: (1) increased pressure in the pancreatic duct and (2) reflux of bile into the pancreatic duct (61). The latter can be recapitulated in vitro by exogenous administration of bile or its components. Bile is composed predominantly of the bile acids taurocholate (TC), taurochenodeoxycholate (TCDC), taurodeoxycholate (TDC), while taurolithocholic acid 3-sulfate (TLCS) comprises a small fraction of bile (26, 135). However, TLCS is most commonly used in vitro because it is the least hydrophilic and, therefore, most potent of the naturally occurring bile acids. It induces Ca2+ signals at low micromolar concentrations that are below the critical micellar concentration (42). Bile acids can be transported into pancreatic acinar cells through specific transporter, or they can bind to their cognate receptors, including the transmembrane G protein-coupled receptor TGR5 (also known as the G-protein coupled bile acid receptor 1, or GPBAR1) (53, 102). Bile acid administration triggers aberrant acinar cell Ca2+ signals leading to trypsinogen activation and cell death (47, 83, 84, 130). Rescuing ATP depletion by patching ATP into isolated acinar cells prevents necrotic cell death due to the bile acids (11, 130, 131).

Fatty Acids

Recent investigations into the role of obesity during acute pancreatitis have revealed that accumulation of intra-pancreatic fat is associated with greater tendency towards pancreatic necrosis during acute pancreatitis and that acute pancreatitis is associated with multisystem organ failure in obese individuals (23, 88, 108). These findings provided the rationale to examine a direct role for fatty acids in acinar cell pathobiology in vitro. Unsaturated fatty acids, in particular, appear to exert a proinflammatory role; they trigger pathological intracellular Ca2+ signals, inhibit mitochondrial complexes I and V, and cause necrosis. Saturated fatty acids have no such effect.

5. Alcoholic Models

Alcohol is a major etiology of acute pancreatitis (28, 145). Chronic ethanol exposure appears to sensitize the pancreas to the pathologic effects of other concomitant stressors in the development of the disease (2, 57, 70, 96).

The mechanism of the sensitizing effect of alcohol is unclear. In vitro exposure to clinically relevant concentrations of ethanol (50-100 mM; for at least an hour of incubation, under sealed conditions) in combination with physiological concentrations of CCK or carbachol have been shown to trigger pathological responses of pancreatitis in acinar cells, including protease activation, intracellular activation of NF-kB, expression of pro-inflammatory cytokines, vacuolization, and necrosis (25, 32, 56, 96).

One mechanism of ethanol’s toxic effects is through the actions of its metabolites, including the oxidative (acetaldehyde) and non-oxidative (fatty acid ethyl ester, FAEEs) metabolites (9, 17, 22, 136, 137, 143). Several studies have now demonstrated that both pathways in ethanol metabolism are evident in the pancreas and that exposure of pancreatic acinar cells to ethanol alone results in accumulation of both acetaldehyde and FAEEs (17, 18, 38). The non-oxidative metabolites FAEEs increase acinar cell lysosomal fragility and induce a rise in intracellular Ca2+ (19, 40, 141), along with premature intracellular digestive enzyme activation, acinar cell vacuolization, and loss of DYm, ATP depletion, and cell necrosis (17, 131).

6. Summary

In summary, we have described methods for the isolation of pancreatic acinar cells, lobules, organoids, and slices. In addition, we have provided a description of assays for critical surrogates of pancreatitis in vitro. Lastly, we have given an overview of the various types of secretagogues and naturally occurring agonists that can be used to stimulate pancreatic acinar cells in vitro for the purpose of studying pathologic surrogates of pancreatitis. The use of such tools is helping researchers, not only to elucidate the molecular mechanisms mediating acute pancreatitis, but also to test novel therapeutic agents on acinar cells, that could reduce cell damage caused by pancreatitis.

7. References

  1. Amsterdam A and Jamieson JD. Structural and functional characterization of isolated pancreatic exocrine cells. Proc Natl Acad Sci USA 69(10): 3028-3032, 1972. PMID: 4342974.
  2. Apte MV, Pirola RC and Wilson JS. Individual susceptibility to alcoholic pancreatitis. Journal of Gastroenterology and Hepatology 23(s1): S63-S68, 2008. PMID: 18336667.
  3. Badr CE, Niers JM, Tjon-Kon-Fat LA, Noske DP, Wurdinger T and Tannous BA. Real-time monitoring of nuclear factor kappaB activity in cultured cells and in animal models. Mol Imaging 8(5): 278-290, 2009. PMID: 19796605.
  4. Bai HX, Lowe ME and Husain SZ. What have we learned about acute pancreatitis in children? J Pediatr Gastroenterol Nutr 52(3): 262-270, 2011. PMID: 21336157.
  5. Barnhart DC, Sarosi GA, Jr., Romanchuk G and Mulholland MW. Calcium signaling induced by angiotensin II in the pancreatic acinar cell line AR42J. Pancreas 18(2): 189-196, 1999. PMID: 10090417.
  6. Barreto SG, Carati CJ, Toouli J and Saccone GT. The islet-acinar axis of the pancreas: more than just insulin. Am J Physiol Gastrointest Liver Physiol 299(1): G10-22, 2010. PMID: 20395539.
  7. Barreto SG, Woods CM, Carati CJ, Schloithe AC, Jaya SR, Toouli J, et al. Galanin inhibits caerulein-stimulated pancreatic amylase secretion via cholinergic nerves and insulin. Am J Physiol Gastrointest Liver Physiol 297(2): G333-339, 2009. PMID: 19497960.
  8. Berridge MV, Herst PM and Tan AS. Tetrazolium dyes as tools in cell biology: new insights into their cellular reduction. Biotechnol Annu Rev 11: 127-152, 2005. PMID: 16216776.
  9. Best CA and Laposata M. Fatty acid ethyl esters: toxic non-oxidative metabolites of ethanol and markers of ethanol intake. Front Biosci 8: e202-217, 2003. PMID: 12456329.
  10. Boj SF, Hwang CI, Baker LA, Chio, II, Engle DD, Corbo V, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 160(1-2): 324-338, 2015. PMID: 25557080.
  11. Booth DM MJ, Mukherjee R, Awais M, Neoptolemos JP, Gerasimenko OV, Tepikin AV, et al. Reactive oxygen species induced by bile acid induce apoptosis and protect against necrosis in pancreatic acinar cells. Gastroenterology 140(7): 9, 2011. PMID: 21354148.
  12. Bottino R, Bertera S, Grupillo M, Melvin PR, Humar A, Mazariegos G, et al. Isolation of human islets for autologous islet transplantation in children and adolescents with chronic pancreatitis. J Transplant 2012: 642787, 2012. PMID: 22461976.
  13. Burnham DB, McChesney DJ, Thurston KC and Williams JA. Interaction of cholecystokinin and vasoactive intestinal polypeptide on function of mouse pancreatic acini in vitro. J Physiol 349: 475-482, 1984. PMID: 6204039.
  14. Burnham DB and Williams JA. Effects of high concentrations of secretagogues on the morphology and secretory activity of the pancreas: a role for microfilaments. Cell Tissue Res 222(1): 201-212, 1982. PMID: 6174234.
  15. Christophe J. Pancreatic tumoral cell line AR42J: an amphicrine model. Am J Physiol 266(6 Pt 1): G963-971, 1994. PMID: 7517639.
  16. Cory AH, Owen TC, Barltrop JA and Cory JG. Use of an aqueous soluble tetrazolium/formazan assay for cell growth assays in culture. Cancer Commun 3(7): 207-212, 1991. PMID: 1867954.
  17. Criddle DN, Murphy J, Fistetto G, Barrow S, Tepikin AV, Neoptolemos JP, et al. Fatty acid ethyl esters cause pancreatic calcium toxicity via inositol trisphosphate receptors and loss of ATP synthesis. Gastroenterology 130(3): 781-793, 2006. PMID: 16530519.
  18. Criddle DN, Raraty MG, Neoptolemos JP, Tepikin AV, Petersen OH and Sutton R. Ethanol toxicity in pancreatic acinar cells: mediation by nonoxidative fatty acid metabolites. Proc Natl Acad Sci USA 101(29): 10738-10743, 2004. PMID: 15247419.
  19. Criddle DN, Sutton R and Petersen OH. Role of Ca2+ in pancreatic cell death induced by alcohol metabolites. J Gastroenterol Hepatol 21 Suppl 3: S14-17, 2006. PMID: 16958662.
  20. De Lisle RC, Norkina O, Roach E and Ziemer D. Expression of pro-Muclin in pancreatic AR42J cells induces functional regulated secretory granules. Am J Physiol Cell Physiol 289(5): C1169-1178, 2005. PMID: 15987769.
  21. de Wet JR, Wood KV, DeLuca M, Helinski DR and Subramani S. Firefly luciferase gene: structure and expression in mammalian cells. Mol Cell Biol 7(2): 725-737, 1987. PMID: 3821727.
  22. Dolai S, Liang T, Lam PP, Fernandez NA, Chidambaram S and Gaisano HY. Effects of ethanol metabolites on exocytosis of pancreatic acinar cells in rats. Gastroenterology 143(3): 832-843 e831-837, 2012. PMID: 22710192.
  23. Durgampudi C, Noel P, Patel K, Cline R, Trivedi RN, DeLany JP, et al. Acute lipotoxicity regulates severity of biliary acute pancreatitis without affecting its initiation. Am J Pathol 184(6): 1773-1784, 2014. PMID: 24854864.
  24. Eisses JF, Davis AW, Tosun AB, Dionise ZR, Chen C, Ozolek JA, et al. A computer-based automated algorithm for assessing acinar cell loss after experimental pancreatitis. PloS one 9(10): e110220, 2014. PMID: 25343460.
  25. Fernandez-Sanchez M, del Castillo-Vaquero A, Salido GM and Gonzalez A. Ethanol exerts dual effects on calcium homeostasis in CCK-8-stimulated mouse pancreatic acinar cells. BMC Cell Biol 10: 77, 2009. PMID: 19878551.
  26. Fisher MM and Yousef IM. Sex differences in the bile acid composition of human bile: studies in patients with and without gallstones. Can Med Assoc J 109(3): 190-193, 1973. PMID: 4728947.
  27. Flowe KM, Welling TH and Mulholland MW. Gastrin-releasing peptide stimulation of amylase release from rat pancreatic lobules involves intrapancreatic neurons. Pancreas 9(4): 513-517, 1994. PMID: 7524066.
  28. Frey CF, Zhou H, Harvey DJ and White RH. The incidence and case-fatality rates of acute biliary, alcoholic, and idiopathic pancreatitis in California, 1994-2001. Pancreas 33(4): 336-344, 2006. PMID: 17079936.
  29. Frick TW, Fernandez-del-Castillo C, Bimmler D and Warshaw AL. Elevated calcium and activation of trypsinogen in rat pancreatic acini. Gut 41(3): 339-343, 1997. PMID: 9378389.
  30. Frossard J-L, Steer ML and Pastor CM. Acute pancreatitis. The Lancet 371(9607): 143-152, 2008. PMID: 18191686.
  31. Giovannucci DR, Bruce JIE, Straub SV, Arreola J, Sneyd J, Shuttleworth TJ, et al. Cytosolic Ca2+ and Ca2+-activated Cl- current dynamics: insights from two functionally distinct mouse exocrine cells. J Physiol 540(2): 469-484, 2002. PMID: 11956337.
  32. Gonzalez A, Nunez AM, Granados MP, Pariente JA and Salido GM. Ethanol impairs CCK-8-evoked amylase secretion through Ca2+-mediated ROS generation in mouse pancreatic acinar cells. Alcohol 38(1): 51-57, 2006. PMID: 16762692.
  33. Gonzalez A S-CP, Salido GM (2011) Culture of pancreatic AR42J cell for use as a model for acinar cell function In: The Pancreapedia: Exocrine Pancreas Knowledge Base 2011,  DOI: 10.3998/panc.2011.26.
  34. Grady T, Mah'Moud M, Otani T, Rhee S, Lerch MM and Gorelick FS. Zymogen proteolysis within the pancreatic acinar cell is associated with cellular injury. Am J Physiol 275(5 Pt 1): G1010-1017, 1998. PMID: 9815031.
  35. Grady T, Saluja A, Kaiser A and Steer M. Edema and intrapancreatic trypsinogen activation precede glutathione depletion during caerulein pancreatitis. Am J Physiol 271(1 Pt 1): G20-26, 1996. PMID: 8760102.
  36. Grossman A. An overview of pancreatic exocrine secretion. Comp Biochem Physiol B 78(1): 1-13, 1984. PMID: 6378509.
  37. Grynkiewicz G, Poenie M and Tsien RY. A new generation of Ca2+ indicators with greatly improved fluorescence properties. J Biol Chem 260(6): 3440-3450, 1985. PMID: 3838314.
  38. Gukovskaya AS, Mouria M, Gukovsky I, Reyes CN, Kasho VN, Faller LD, et al. Ethanol metabolism and transcription factor activation in pancreatic acinar cells in rats. Gastroenterology 122(1): 106-118, 2002. PMID: 11781286.
  39. Gukovsky I, Pandol SJ and Gukovskaya AS. Organellar dysfunction in the pathogenesis of pancreatitis. Antioxid Redox Signal 15(10): 2699-2710, 2011. PMID: 21834686.
  40. Haber PS, Wilson JS, Apte MV and Pirola RC. Fatty acid ethyl esters increase rat pancreatic lysosomal fragility. J Lab Clin Med 121(6): 759-764, 1993. PMID: 8505587.
  41. Haridas V, Shrivastava A, Su J, Yu GL, Ni J, Liu D, et al. VEGI, a new member of the TNF family activates nuclear factor-kappa B and c-Jun N-terminal kinase and modulates cell growth. Oncogene 18(47): 6496-6504, 1999. PMID: 10597252.
  42. Hofmann AF and Roda A. Physicochemical properties of bile acids and their relationship to biological properties: an overview of the problem. J Lipid Res 25(13): 1477-1489, 1984. PMID: 6397555.
  43. Hoque R, Farooq A, Ghani A, Gorelick F and Mehal WZ. Lactate reduces liver and pancreatic injury in Toll-like receptor- and inflammasome-mediated inflammation via GPR81-mediated suppression of innate immunity. Gastroenterology 146(7): 1763-1774, 2014. PMID: 24657625.
  44. Huang YC, Gaisano HY and Leung YM. Electrophysiological identification of mouse islet alpha-cells: from isolated single alpha-cells to in situ assessment within pancreas slices. Islets 3(4): 139-143, 2011. PMID: 21623173.
  45. Huang YC, Rupnik M and Gaisano HY. Unperturbed islet alpha-cell function examined in mouse pancreas tissue slices. J Physiol 589(Pt 2): 395-408, 2011. PMID: 21078586.
  46. Huch M, Bonfanti P, Boj SF, Sato T, Loomans CJ, van de Wetering M, et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J 32(20): 2708-2721, 2013. PMID: 24045232.
  47. Husain SZ, Orabi AI, Muili KA, Luo Y, Sarwar S, Mahmood SM, et al. Ryanodine receptors contribute to bile acid-induced pathological calcium signaling and pancreatitis in mice. Am J Physiol Gastrointest Liver Physiol 302(12): G1423-1433, 2012. PMID: 22517774.
  48. Husain SZ, Prasad P, Grant WM, Kolodecik TR, Nathanson MH and Gorelick FS. The ryanodine receptor mediates early zymogen activation in pancreatitis. Proc Natl Acad Sci USA 102(40): 14386-14391, 2005. PMID: 16186498.
  49. Jamieson JD and Palade GE. Intracellular transport of secretory proteins in the pancreatic exocrine cell. II. Transport to condensing vacuoles and zymogen granules. J Cell Biol 34(2): 597-615, 1967. PMID: 6035648.
  50. Ji B, Bi Y, Simeone D, Mortensen RM and Logsdon CD. Human pancreatic acinar cells lack functional responses to cholecystokinin and gastrin. Gastroenterology 121(6): 1380-1390, 2001. PMID: 11729117.
  51. Kang R, Zhang Q, Hou W, Yan Z, Chen R, Bonaroti J, et al. Intracellular Hmgb1 inhibits inflammatory nucleosome release and limits acute pancreatitis in mice. Gastroenterology 146(4): 1097-1107, 2014. PMID: 24361123.
  52. Kasai H and Augustine GJ. Cytosolic Ca2+ gradients triggering unidirectional fluid secretion from exocrine pancreas. Nature 348(6303): 735-738, 1990. PMID: 1701852.
  53. Kim JY, Kim KH, Lee JA, Namkung W, Sun AQ, Ananthanarayanan M, et al. Transporter-mediated bile acid uptake causes Ca2+-dependent cell death in rat pancreatic acinar cells. Gastroenterology 122(7): 1941-1953, 2002. PMID: 12055600.
  54. Kim MS, Lee KP, Yang D, Shin DM, Abramowitz J, Kiyonaka S, et al. Genetic and pharmacologic inhibition of the Ca2+ influx channel TRPC3 protects secretory epithelia from Ca2+-dependent toxicity. Gastroenterology 140(7): 2107-2115, 2115 e2101-2104, 2011. PMID: 21354153.
  55. Kruger B, Albrecht E and Lerch MM. The role of intracellular calcium signaling in premature protease activation and the onset of pancreatitis. Am J Pathol 157(1): 43-50, 2000. PMID: 10880374.
  56. Lam PP, Cosen Binker LI, Lugea A, Pandol SJ and Gaisano HY. Alcohol redirects CCK-mediated apical exocytosis to the acinar basolateral membrane in alcoholic pancreatitis. Traffic 8(5): 605-617, 2007. PMID: 17451559.
  57. Lankisch PG, Apte M and Banks PA. Acute pancreatitis. Lancet, 2015. PMID: 25616312.
  58. Leach SD, Modlin IM, Scheele GA and Gorelick FS. Intracellular activation of digestive zymogens in rat pancreatic acini. Stimulation by high doses of cholecystokinin. J Clin Invest 87(1): 362-366, 1991. PMID: 1985109.
  59. Lecoeur H. Nuclear apoptosis detection by flow cytometry: influence of endogenous endonucleases. Exp Cell Res 277(1): 1-14, 2002. PMID: 12061813.
  60. Leite MF, Burgstahler AD and Nathanson MH. Ca2+ waves require sequential activation of inositol trisphosphate receptors and ryanodine receptors in pancreatic acini. Gastroenterology 122(2): 415-427, 2002. PMID: 11832456.
  61. Lerch MM and Aghdassi AA. The role of bile acids in gallstone-induced pancreatitis. Gastroenterology 138(2): 429-433, 2010. PMID: 20034603.
  62. Lerch MM and Gorelick FS. Early trypsinogen activation in acute pancreatitis. Med Clin North Am 84(3): 549-563, viii, 2000. PMID: 10872413.
  63. Lewarchik CM, Orabi AI, Jin S, Wang D, Muili KA, Shah AU, et al. The ryanodine receptor is expressed in human pancreatic acinar cells and contributes to acinar cell injury. Am J Physiol Gastrointest Liver Physiol 307(5): G574-581, 2014. PMID: 25012845.
  64. Leytus SP, Patterson WL and Mangel WF. New class of sensitive and selective fluorogenic substrates for serine proteinases. Amino acid and dipeptide derivatives of rhodamine. Biochem J 215(2): 253-260, 1983. PMID: 6228222.
  65. Leytus SP, Toledo DL and Mangel WF. Theory and experimental method for determining individual kinetic constants of fast-acting, irreversible proteinase inhibitors. Biochim Biophys Acta 788(1): 74-86, 1984. PMID: 6204689.
  66. Linari G, Nencini P and Nucerito V. Cadmium inhibits stimulated amylase secretion from isolated pancreatic lobules of the guinea-pig. Pharmacol Res Society 43(3): 219-223, 2001. PMID: 11401412.
  67. Logsdon CD, Moessner J, Williams JA and Goldfine ID. Glucocorticoids increase amylase mRNA levels, secretory organelles, and secretion in pancreatic acinar AR42J cells. J Cell Biol 100(4): 1200-1208, 1985. PMID: 2579957.
  68. Lorenz WW, McCann RO, Longiaru M and Cormier MJ. Isolation and expression of a cDNA encoding Renilla reniformis luciferase. Proc Natl Acad Sci U S A 88(10): 4438-4442, 1991. PMID: 1674607.
  69. Lowenfels AB, Sullivan T, Fiorianti J and Maisonneuve P. The epidemiology and impact of pancreatic diseases in the United States. Curr Gastroenterol Rep 7(2): 90-95, 2005. PMID: 15802095.
  70. Lugea A, Gong J, Nguyen J, Nieto J, French SW and Pandol SJ. Cholinergic mediation of alcohol-induced experimental pancreatitis. Alcohol Clin Exp Res 34(10): 1768-1781, 2010. PMID: 20626730.
  71. Ma MH, Bai HX, Park AJ, Latif SU, Mistry PK, Pashankar D, et al. Risk factors associated with biliary pancreatitis in children. J Pediatr Gastroenterol Nutr 54(5): 651-656, 2012. PMID: 22002481.
  72. Madern D. Molecular evolution within the L-malate and L-lactate dehydrogenase super-family. J Mol Evol 54(6): 825-840, 2002. PMID: 12029364.
  73. Maleth J, Rakonczay Z, Jr., Venglovecz V, Dolman NJ and Hegyi P. Central role of mitochondrial injury in the pathogenesis of acute pancreatitis. Acta Physiol 207(2): 226-235, 2013. PMID: 23167280.
  74. Mareninova OA, Sung KF, Hong P, Lugea A, Pandol SJ, Gukovsky I, et al. Cell death in pancreatitis: caspases protect from necrotizing pancreatitis. J Biol Chem 281(6): 3370-3381, 2006. PMID: 16339139.
  75. Means AL, Meszoely IM, Suzuki K, Miyamoto Y, Rustgi AK, Coffey RJ, Jr., et al. Pancreatic epithelial plasticity mediated by acinar cell transdifferentiation and generation of nestin-positive intermediates. Development 132(16): 3767-3776, 2005. PMID: 16020518.
  76. Miller LJ and Lybrand TP. Molecular Basis of Agonist Binding to the Type A Cholecystokinin Receptor. Pharmacology and Toxicology 91(6): 282-285, 2002. PMID: 12688369.
  77. Minta A, Kao JP and Tsien RY. Fluorescent indicators for cytosolic calcium based on rhodamine and fluorescein chromophores. J Biol Chem 264(14): 8171-8178, 1989. PMID: 2498308.
  78. Miyasaka K, Shinozaki H, Jimi A and Funakoshi A. Amylase secretion from dispersed human pancreatic acini: neither cholecystokinin a nor cholecystokinin B receptors mediate amylase secretion in vitro. Pancreas 25(2): 161-165, 2002. PMID: 12142739.
  79. Moore A, Donahue CJ, Bauer KD and Mather JP. Simultaneous measurement of cell cycle and apoptotic cell death. Methods Cell Biol 57: 265-278, 1998. PMID: 9648110.
  80. Mooren FC, Turi S, Gunzel D, Schlue W-R, Domschke W, Singh J, et al. Calcium-magnesium interactions in pancreatic acinar cells. FASEB J. 15(3): 659-672, 2001. PMID: 11259384.
  81. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 65(1-2): 55-63, 1983. PMID: 6606682.
  82. Muili KA, Ahmad M, Orabi AI, Mahmood SM, Shah AU, Molkentin JD, et al. Pharmacological and genetic inhibition of calcineurin protects against carbachol-induced pathological zymogen activation and acinar cell injury. Am J Physiol Gastrointest Liver Physiol 302(8): G898-905, 2012. PMID: 22323127.
  83. Muili KA, Jin S, Orabi AI, Eisses JF, Javed TA, Le T, et al. Pancreatic acinar cell NF-kappaB activation due to bile acid exposure is dependent on calcineurin. J Biol Chem, 2013. PMID: 23744075.
  84. Muili KA, Wang D, Orabi AI, Sarwar S, Luo Y, Javed TA, et al. Bile acids induce pancreatic acinar cell injury and pancreatitis by activating calcineurin. J Biol Chem 288(1): 570-580, 2013. PMID: 23148215.
  85. Mukherjee R, Mareninova OA, Odinokova IV, Huang W, Murphy J, Chvanov M, et al. Mechanism of mitochondrial permeability transition pore induction and damage in the pancreas: inhibition prevents acute pancreatitis by protecting production of ATP. Gut, 2015. PMID: 26071131.
  86. Murphy JA, Criddle DN, Sherwood M, Chvanov M, Mukherjee R, McLaughlin E, et al. Direct activation of cytosolic Ca2+ signaling and enzyme secretion by cholecystokinin in human pancreatic acinar cells. Gastroenterology 135(2): 632-641, 2008. PMID: 18555802.
  87. Nakajima Y, Kobayashi K, Yamagishi K, Enomoto T and Ohmiya Y. cDNA cloning and characterization of a secreted luciferase from the luminous Japanese ostracod, Cypridina noctiluca. Biosci Biotechnol Biochem 68(3): 565-570, 2004. PMID: 15056888.
  88. Navina S, Acharya C, DeLany JP, Orlichenko LS, Baty CJ, Shiva SS, et al. Lipotoxicity causes multisystem organ failure and exacerbates acute pancreatitis in obesity. Sci Transl Med 3(107): 107ra110, 2011. PMID: 22049070.
  89. Nicke B, Tseng MJ, Fenrich M and Logsdon CD. Adenovirus-mediated gene transfer of RasN17 inhibits specific CCK actions on pancreatic acinar cells. Am J Physiol 276(2 Pt 1): G499-506, 1999. PMID: 9950825.
  90. Odinokova I. V. SN, Gukovskaya A. S., Mareninova O.A. Isolation of pancreatic mitochondria and measurement of their functional parameters. In: Pancreapedia: Exocrine Pancreas Knowledge Base, 2011. DOI: 10.3998/panc.2011.25.
  91. Odinokova IV, Sung KF, Mareninova OA, Hermann K, Evtodienko Y, Andreyev A, et al. Mechanisms regulating cytochrome c release in pancreatic mitochondria. Gut 58(3): 431-442, 2009. PMID: 18596195.
  92. Odinokova IV, Sung KF, Mareninova OA, Hermann K, Gukovsky I and Gukovskaya AS. Mitochondrial mechanisms of death responses in pancreatitis. Journal of gastroenterology and hepatology 23 Suppl 1: S25-30, 2008. PMID: 18336659.
  93. Orabi AI, Muili KA, Wang D, Jin S, Perides G and Husain SZ. Preparation of pancreatic acinar cells for the purpose of calcium imaging, cell injury measurements, and adenoviral infection. J Vis Exp(77), 2013. PMID: 23851390.
  94. Orabi AI, Nathanson, Michael H. and Husain, Sohail Z. (2011) Measuring Ca2+ dynamics in pancreatic acini using confocal microscopy. In: Pancreapedia: Exocrine Pancreas Knowledge Base  DOI: 10.3998/panc.2011.30.
  95. Orabi AI, Sah S, Javed TA, Lemon KL, Good ML, Guo P, et al. Dynamic Imaging of Pancreatic Nuclear Factor kappaB (NF-kappaB) Activation in Live Mice Using Adeno-associated Virus (AAV) Infusion and Bioluminescence. The Journal of biological chemistry 290(18): 11309-11320, 2015. PMID: 25802340.
  96. Orabi AI, Shah AU, Muili K, Luo Y, Mahmood SM, Ahmad A, et al. Ethanol enhances carbachol-induced protease activation and accelerates Ca2+ waves in isolated rat pancreatic acini. J Biol Chem 286(16): 14090-14097, 2011. PMID: 21372126.
  97. Ornitz DM, Palmiter RD, Hammer RE, Brinster RL, Swift GH and MacDonald RJ. Specific expression of an elastase-human growth hormone fusion gene in pancreatic acinar cells of transgenic mice. Nature 313(6003): 600-602, 1985. PMID: 3844051.
  98. Owyang C and Logsdon CD. New insights into neurohormonal regulation of pancreatic secretion. Gastroenterology 127(3): 957-969, 2004. PMID: 15362050.
  99. Park MK, Lee M and Petersen OH. Morphological and functional changes of dissociated single pancreatic acinar cells: testing the suitability of the single cell as a model for exocytosis and calcium signaling. Cell calcium 35(4): 367-379, 2004. PMID: 15036953.
  100. Peery AF, Dellon ES, Lund J, Crockett SD, McGowan CE, Bulsiewicz WJ, et al. Burden of gastrointestinal disease in the United States: 2012 update. Gastroenterology 143(5): 1179-1187 e1171-1173, 2012. PMID: 22885331.
  101. Peikin SR, Rottman AJ, Batzri S and Gardner JD. Kinetics of amylase release by dispersed acini prepared from guinea pig pancreas. Am J Physiol 235(6): E743-749, 1978. PMID: 736135.
  102. Perides G, Laukkarinen JM, Vassileva G and Steer ML. Biliary acute pancreatitis in mice is mediated by the G-protein-coupled cell surface bile acid receptor Gpbar1. Gastroenterology 138(2): 715-725, 2010. PMID: 19900448.
  103. Petersen OH and Tepikin AV. Polarized calcium signaling in exocrine gland cells. Annu Rev Physiol 70: 273-299, 2008. PMID: 17850212.
  104. Rakonczay Z, Jr., Hegyi P, Takacs T, McCarroll J and Saluja AK. The role of NF-kappaB activation in the pathogenesis of acute pancreatitis. Gut 57(2): 259-267, 2008. PMID: 17675325.
  105. Raraty M, Ward J, Erdemli G, Vaillant C, Neoptolemos JP, Sutton R, et al. Calcium-dependent enzyme activation and vacuole formation in the apical granular region of pancreatic acinar cells. Proc Natl Acad Sci U S A 97(24): 13126-13131, 2000. PMID: 11087863.
  106. Reed AM, Husain SZ, Thrower E, Alexandre M, Shah A, Gorelick FS, et al. Low extracellular pH induces damage in the pancreatic acinar cell by enhancing calcium signaling. J Biol Chem 286(3): 1919-1926, 2011. PMID: 21084290.
  107. Roeyen G, Chapelle T, Jorens P, de Beeck BO and Ysebaert D. Necrotizing pancreatitis due to poisoning with organophosphate pesticides. Acta Gastroenterol Belg 71(1): 27-29, 2008. PMID: 18396746.
  108. Sah RP, Dawra RK and Saluja AK. New insights into the pathogenesis of pancreatitis. Curr Opin Gastroenterol 29(5): 523-530, 2013. PMID: 23892538.
  109. Saluja A, Logsdon C and Garg P. Direct versus indirect action of cholecystokinin on human pancreatic acinar cells: is it time for a judgment after a century of trial? Gastroenterology 135(2): 357-360, 2008. PMID: 18616945.
  110. Saluja AK, Bhagat L, Lee HS, Bhatia M, Frossard JL and Steer ML. Secretagogue-induced digestive enzyme activation and cell injury in rat pancreatic acini. Am J Physiol 276(4 Pt 1): G835-842, 1999. PMID: 10198325.
  111. Saluja AK, Donovan EA, Yamanaka K, Yamaguchi Y, Hofbauer B and Steer ML. Cerulein-induced in vitro activation of trypsinogen in rat pancreatic acini is mediated by cathepsin B. Gastroenterology 113(1): 304-310, 1997. PMID: 9207291.
  112. Saluja AK, Lerch MM, Phillips PA and Dudeja V. Why does pancreatic overstimulation cause pancreatitis? Annu Rev Physiol 69: 249-269, 2007. PMID: 17059357.
  113. Scheele GA and Palade GE. Studies on the guinea pig pancreas. Parallel discharge of exocrine enzyme activities. J Biol Chem 250(7): 2660-2670, 1975. PMID: 1123325.
  114. Schloithe AC, Sutherland K, Woods CM, Blackshaw LA, Davison JS, Toouli J, et al. A novel preparation to study rat pancreatic spinal and vagal mechanosensitive afferents in vitro. Neurogastroenterol Motil 20(9): 1060-1069, 2008. PMID: 18482253.
  115. Schmidt WE, Meyer-Alber A, Waschulewski IH, Fetz I, Hocker M, Kern HF, et al. Serine/threonine phosphatases play a role in stimulus-secretion coupling in pancreatic acinar cells. Z Gastroenterol 32(4): 226-231, 1994. PMID: 7517088.
  116. Schultz GS, Sarras MP, Jr., Gunther GR, Hull BE, Alicea HA, Gorelick FS, et al. Guinea pig pancreatic acini prepared with purified collagenase. Exp Cell Res 130(1): 49-62, 1980. PMID: 6256185.
  117. Shah AU, Grant WM, Latif SU, Mannan ZM, Park AJ and Husain SZ. Cyclic-AMP Accelerates Calcium Waves in Pancreatic Acinar Cells. Am J Physiol Gastrointest Liver Physiol 294(6): G1328-1334, 2008. PMID: 18388188.
  118. Shah AU, Sarwar A, Orabi AI, Gautam S, Grant WM, Park AJ, et al. Protease Activation during in vivo Pancreatitis is Dependent upon Calcineurin Activation. Am J Physiol Gastrointest Liver Physiol, 2009. PMID: 19713471.
  119. Shalbueva N, Mareninova OA, Gerloff A, Yuan J, Waldron RT, Pandol SJ, et al. Effects of Oxidative Alcohol Metabolism on the Mitochondrial Permeability Transition Pore and Necrosis in a Mouse Model of Alcoholic Pancreatitis. Gastroenterology, 2012. PMID: 23103769.
  120. Sherwood MW, Prior IA, Voronina SG, Barrow SL, Woodsmith JD, Gerasimenko OV, et al. Activation of trypsinogen in large endocytic vacuoles of pancreatic acinar cells. Proc Natl Acad Sci USA 104(13): 5674-5679, 2007. PMID: 17363470.
  121. Singh S, Bhardwaj U, Verma SK, Bhalla A and Gill K. Hyperamylasemia and acute pancreatitis following anticholinesterase poisoning. Hum Exp Toxicol 26(6): 467-471, 2007. PMID: 17698941.
  122. Susini C, Estival A, Scemama JL, Ruellan C, Vaysse N, Clemente F, et al. Studies on human pancreatic acini: action of secretagogues on amylase release and cellular cyclic AMP accumulation. Pancreas 1(2): 124-129, 1986. PMID: 2437561.
  123. Suzuki T, Fujikura K, Higashiyama T and Takata K. DNA staining for fluorescence and laser confocal microscopy. J Histochem Cytochem 45(1): 49-53, 1997. PMID: 9010468.
  124. Szmola R and Sahin-Toth M. Pancreatitis-associated chymotrypsinogen C (CTRC) mutant elicits endoplasmic reticulum stress in pancreatic acinar cells. Gut 59(3): 365-372, 2010. PMID: 19951900.
  125. Tando Y, Algul H, Schneider G, Weber CK, Weidenbach H, Adler G, et al. Induction of IkappaB-kinase by cholecystokinin is mediated by trypsinogen activation in rat pancreatic lobules. Digestion 66(4): 237-245, 2002. PMID: 12592100.
  126. Tannous BA, Kim DE, Fernandez JL, Weissleder R and Breakefield XO. Codon-optimized Gaussia luciferase cDNA for mammalian gene expression in culture and in vivo. Mol Ther 11(3): 435-443, 2005. PMID: 15727940.
  127. Thomas D, Tovey SC, Collins TJ, Bootman MD, Berridge MJ and Lipp P. A comparison of fluorescent Ca2+ indicator properties and their use in measuring elementary and global Ca2+ signals. Cell Calcium 28(4): 213-223, 2000. PMID: 11032777.
  128. Tomiyama M, Arai A, Kimura T, Suzuki C, Watanabe M, Kawarabayashi T, et al. Exacerbation of chronic pancreatitis induced by anticholinesterase medications in myasthenia gravis. Eur J Neurol 15(5): e40-41, 2008. PMID: 18325026.
  129. van Geenen EJ, van der Peet DL, Bhagirath P, Mulder CJ and Bruno MJ. Etiology and diagnosis of acute biliary pancreatitis. Nat Rev Gastroenterol Hepatol 7(9): 495-502, 2010. PMID: 20703238.
  130. Voronina S, Longbottom R, Sutton R, Petersen OH and Tepikin A. Bile acids induce calcium signals in mouse pancreatic acinar cells: implications for bile-induced pancreatic pathology. J Physiol 540(Pt 1): 49-55, 2002. PMID: 11927668.
  131. Voronina SG, Barrow SL, Simpson AW, Gerasimenko OV, da Silva Xavier G, Rutter GA, et al. Dynamic changes in cytosolic and mitochondrial ATP levels in pancreatic acinar cells. Gastroenterology 138(5): 1976-1987, 2010. PMID: 20102715.
  132. Votanopoulos KI, Lee TC, Dominguez EP, Choi YU and Sweeney JF. Propoxur induced pancreatitis after inhalation of baygon pesticide. Pancreas 34(3): 379-380, 2007. PMID: 17414064.
  133. Wank SA, Pisegna JR and de Weerth A. Cholecystokinin receptor family. Molecular cloning, structure, and functional expression in rat, guinea pig, and human. Ann N Y Acad Sci 713: 49-66, 1994. PMID: 8185215.
  134. Watanabe O, Baccino FM, Steer ML and Meldolesi J. Supramaximal caerulein stimulation and ultrastructure of rat pancreatic acinar cell: early morphological changes during development of experimental pancreatitis. Am J Physiol 246(4 Pt 1): G457-467, 1984. PMID: 6720895.
  135. Weinman SA and Jalil S. Bile Secretion and Cholestasis. Textbook of Gastroenterology 5th Ed. T. Yamada, Blackwell Publishing Ltd.: 401-428, 2008.
  136. Werner J, Laposata M, Fernandez-del Castillo C, Saghir M, Iozzo RV, Lewandrowski KB, et al. Pancreatic injury in rats induced by fatty acid ethyl ester, a nonoxidative metabolite of alcohol. Gastroenterology 113(1): 286-294, 1997. PMID: 9207289.
  137. Werner J, Saghir M, Warshaw AL, Lewandrowski KB, Laposata M, Iozzo RV, et al. Alcoholic pancreatitis in rats: injury from nonoxidative metabolites of ethanol. Am J Physiol Gastrointest Liver Physiol 283(1): G65-73, 2002. PMID: 12065293.
  138. Williams JA. Intracellular signaling mechanisms activated by cholecystokinin-regulating synthesis and secretion of digestive enzymes in pancreatic acinar cells. Annu Rev Physiol 63: 77-97, 2001. PMID: 11181949.
  139. Williams JA. Receptor biology and intracellular regulatory mechanisms in pancreatic acinar cells. Curr Opin Gastroenterol 18(5): 529-535, 2002. PMID: 17033329.
  140. Williams JA, Korc M and Dormer RL. Action of secretagogues on a new preparation of functionally intact, isolated pancreatic acini. Am J Physiol 235(5): 517-524, 1978. PMID: 215042.
  141. Wilson JS, Korsten MA, Apte MV, Thomas MC, Haber PS and Pirola RC. Both ethanol consumption and protein deficiency increase the fragility of pancreatic lysosomes. J Lab Clin Med 115(6): 749-755, 1990. PMID: 2366035.
  142. Won JH, Cottrell WJ, Foster TH and Yule DI. Ca2+ release dynamics in parotid and pancreatic exocrine acinar cells evoked by spatially limited flash photolysis. Am J Physiol Gastrointest Liver Physiol 293(6): G1166-1177, 2007. PMID: 17901163.
  143. Wu H, Bhopale KK, Ansari GAS and Kaphalia BS. Ethanol-induced cytotoxicity in rat pancreatic acinar AR42J cells: Role of fatty acid ethyl esters. Alcohol Alcohol. 43(1): 1-8, 2008. PMID: 17942438.
  144. Xiao X, Guo P, Prasadan K, Shiota C, Peirish L, Fischbach S, et al. Pancreatic cell tracing, lineage tagging and targeted genetic manipulations in multiple cell types using pancreatic ductal infusion of adeno-associated viral vectors and/or cell-tagging dyes. Nat Protocol 9(12): 2719-2724, 2014. PMID: 25356582.
  145. Yadav D and Lowenfels AB. The epidemiology of pancreatitis and pancreatic cancer. Gastroenterology 144(6): 1252-1261, 2013. PMID: 23622135.
  146. Zhang L, Graziano K, Pham T, Logsdon CD and Simeone DM. Adenovirus-mediated gene transfer of dominant-negative Smad4 blocks TGF-{beta} signaling in pancreatic acinar cells. Am J Physiol Gastrointest Liver Physiol 280(6): G1247-1253, 2001. PMID: 11352818.
  147. Zhao H and Muallem S. Na+, K+, and Cl- transport in resting pancreatic acinar cells. J Gen Physiol 106(6): 1225-1242, 1995. PMID: 8786358.