Comprehensive Physiology Wiley Online Library

Cholecystokinin (CCK) Regulation of Pancreatic Acinar Cells: Physiological Actions and Signal Transduction Mechanisms

Full Article on Wiley Online Library



ABSTRACT

Pancreatic acinar cells synthesize and secrete about 20 digestive enzymes and ancillary proteins with the processes that match the supply of these enzymes to their need in digestion being regulated by a number of hormones (CCK, secretin and insulin), neurotransmitters (acetylcholine and VIP) and growth factors (EGF and IGF). Of these regulators, one of the most important and best studied is the gastrointestinal hormone, cholecystokinin (CCK). Furthermore, the acinar cell has become a model for seven transmembrane, heterotrimeric G protein coupled receptors to regulate multiple processes by distinct signal transduction cascades. In this review, we briefly describe the chemistry and physiology of CCK and then consider the major physiological effects of CCK on pancreatic acinar cells. The majority of the review is devoted to the physiologic signaling pathways activated by CCK receptors and heterotrimeric G proteins and the functions they affect. The pathways covered include the traditional second messenger pathways PLC‐IP3‐Ca2+, DAG‐PKC, and AC‐cAMP‐PKA/EPAC that primarily relate to secretion. Then there are the protein‐protein interaction pathways Akt‐mTOR‐S6K, the three major MAPK pathways (ERK, JNK, and p38 MAPK), and Ca2+‐calcineurin‐NFAT pathways that primarily regulate non‐secretory processes including biosynthesis and growth, and several miscellaneous pathways that include the Rho family small G proteins, PKD, FAK, and Src that may regulate both secretory and nonsecretory processes but are not as well understood. © 2019 American Physiological Society. Compr Physiol 9:535‐564, 2019.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Two‐dimensional structure of CCK1R showing membrane spanning topology and sites of posttranslational modification. The amino terminal is outside the cell membrane and the carboxy terminal is inside. Sites of glycosylation are indicated by a Y and sites of phosphorylation by P in a circle. There is a site of palmitoylation in the carboxyl terminal tail indicated by two Cs anchored to the membrane. The natural CCK peptide is indicated as a brown oval docked to the receptor. Reproduced, with permission, from reference (62).
Figure 2. Figure 2. CCK1R signals through multiple G proteins to activate different signaling cascades. Whether β‐Arrestin and Gβ/γ play a role in pancreatic acinar cells is unclear (368).
Figure 3. Figure 3. Ca2+ oscillations induced by pmol/L concentrations of CCK that are initially independent of extracellular Ca2+. Ca2+ was removed and EGTA added 1 min before CCK was added. Reproduced, with permission, from reference (323).
Figure 4. Figure 4. CCK initiates Ca2+ waves that travel around an acini. (A) Ca2+ signaling shown for a single cell was typical of all cells in an acinar cluster; however, they did not occur synchronously. (B) Pseudocolor image of Ca2+ concentration shows that superfusion with 20 pmol/L CCK initiates a signal in one cell that then propagates in each cell around the cluster. After a basal period, a new cycle is initiated in the same cell. Reproduced, with permission, from reference (372).
Figure 5. Figure 5. Intracellular Ca2+ signaling in pancreatic acinar cells is initiated by the binding of Acetylcholine to Muscarinic M3 receptors (M3R) and by Cholecystokinin (CCK) to CCK receptors, predominately the CCK1R in mice and rats. Both receptors are classical seven transmembrane domain receptors coupled to guanine nucleotide‐binding proteins (G proteins). Activation of both receptors leads to stimulation of Gαq and increased activity of phospholipase C‐β (PLC) which cleaves the membrane phospholipid phosphatidylinositol,4,5,‐bisphosphate (PIP2) into diacylglycerol and inositol 1,4,5‐trisphosphate (IP3). IP3 diffuses through the cytoplasm and interacts with InsP3 receptors (IP3R), largely type‐2 and ‐3 present predominantly on the apical endoplasmic reticulum (ER) resulting in Ca2+ release into the cytoplasm. Ca2+ release from IP3R acts a co‐agonist to increase further IP3R activity and also acts on Ryanodine receptors (RyR) to induce Ca2+ release. Depletion of Ca2+ within the ER results in Ca2 + influx from the extracellular space. The ER Ca2+ sensor has been identified as stromal interaction molecule‐1 (Stim‐1). Following ER depletion, Ca2+ is released from an EF hand present in a domain of Stim‐1 within the ER lumen and this results in aggregation of several Stim‐1 molecules. Aggregation of Stim‐1 is sufficient to gate plasma membrane Ca2+ channels and leads to Ca2+ influx. Good candidates for the channel pore are the proteins Orai‐1 and TRPC3. CCK receptor stimulation also stimulates ADP‐ribosyl cyclase activity resulting in the formation of two additional Ca2+ mobilizing second messengers; nicotinic acid adenine dinucleotide phosphate (NAADP) and cyclic‐ADP ribose (cADPr). The particular cyclase responsible has not been defined in pancreas but good candidates include CD38 and CD157. cADPr is generally thought to act on RyR, while the target of NAADP is currently the subject of intense research. Candidates include the RyR and Two Pore Channel (TPC). In addition to the ER, Ca2+ can also be released from a store, which accumulates Ca2+ in a manner dependent on a proton gradient‐ known as the “acidic store.” This pool likely represents the endolysosomal compartment. This pool has been reported to be responsive to IP3, cADPr and NAADP and may represent the store initially released following receptor stimulation. Ca2+ is removed from the cytoplasm by the concerted action of SERCA (ER Ca2+‐ATPase), PMCA (PM Ca2+‐ATPase), and the action of MCU (mitochondrial uniporter). Figure and legend reproduced, with permission, from reference (368).
Figure 6. Figure 6. Activation of PKC isoforms in pancreatic acinar cells. CCK and ACh activate PLCβ and EGF activates PLCγ all of which lead to production of diacylglycerol (DAG) from PIP2. CCK also induces DAG formation from phosphatidylcholine (PC). Ca2+ and DAG together activate the classical isoforms PKC α, β, and γ; DAG activates the novel isoforms PKC δ, ϵ, η, and θ; while unconventional isoforms PKC ζ and λ do not require Ca2+ or DAG. Reproduced, with permission, from Pancreapedia, DAG–PKC Signaling Pathway, 2013.
Figure 7. Figure 7. Parallel nature of MAPK cascades in pancreas. Modified and updated, with permission, from reference (281).
Figure 8. Figure 8. Activation of ERK signaling pathway by CCK and EGF in pancreatic acinar cells. CCK1R signals through PKC and the activation by phosphorylation of Raf isoforms. Raf sequentially phosphorylates and activates MEK1/2 and ERK1/2. By contrast, EGFR autophosphorylates and then binds Shc, Grb2 and SOS with the latter being a GEF for Ras; active Ras binds and activates Raf and the kinase cascade ending in ERK. Activated ERK phosphorylates cytoplasmic substrates and downstream kinases, which phosphorylates transcription factors thereby activating expression of specific genes. Reproduced, with permission, from reference (345).
Figure 9. Figure 9. General and simplified diagram of mTORC1 pathway set in pancreatic acinar cell, where the pathway is regulated by CCK and insulin. Red arrows indicate activation; Black arrows indicate inhibition; Green arrow indicates translocation. Biological processes regulated by mTORC1 are shown at the bottom of the figure, along with the key proteins mediating the effect. Reproduced, with permission, from reference (273).
Figure 10. Figure 10. Activation of the Ca2+‐calcineurin‐NFAT signaling pathway by CCK and cholinergic agonists in pancreatic acinar cells. The initial steps in the pathway involve receptor‐mediated elevation of intracellular Ca2+. Ca2+ binds to calmodulin (CaM) and the complex to the Ca2 + activated phosphatase calcineurin. Calcineurin can dephosphorylate a number of substrates but the most generally important are NFATS which when dephosphorylated move into the nucleus and bind alone or together with coactivators such as AP‐1 to activate specific genes including FGF21, Socs3, Rgs2, Hbegf and Rcan1 with the latter feeding back to inhibit calcineurin. Reproduced, with permission, from the Pancreapedia, Calcium–Calcineurin–NFAT Signaling Pathway, 2012.
Figure 11. Figure 11. Rho Activation pathway by CCK and ACh. The CCK1 and muscarinic M3 receptors activate Gα12/13, which activate a Rho GEF of which several present in the pancreas are shown. Rho with bound GTP can activate mDia1 or ROCK (Rho kinase) thereby regulating the actin cytoskeleton or activate SRF (serum response factor) and activate gene expression. GTP bound Rho is inactivated by a Rho GAP of which several present in pancreas are shown. Botulinum exotoxin can permanently inactivate Rho. Reproduced, with permission, from Pancreapedia, Galpha12/13‐Rho Signaling Pathway, 2010.
Figure 12. Figure 12. Activation of cAMP pathway in pancreatic acinar and duct cells. Secretin, VIP and CCK bind to their receptors which activate adenylyl cyclase (AC) through Gαs while Somatostatin acts on SomstostatinR2 to activate Gαi which inhibits AC. Cholera toxin activates Gαs while forskolin directly activates AC and Pertussis toxin inhibits Gαi. AC produces cyclic AMP (cAMP) which is broken down by phosphodiesterase (PDE) which can be inhibited by isobutylmethylxanthine (IBMX). cAMP activates PKA which can phosphorylate a number of proteins including CREB, CFTR and the IP3 receptor. cAMP also activates EPAC1 which acts as a GEF to activate the small G protein Rap1 present on zymogen granules. Reproduced, with permission, from Pancreapedia, cAMP Signaling Pathway, 2012.


Figure 1. Two‐dimensional structure of CCK1R showing membrane spanning topology and sites of posttranslational modification. The amino terminal is outside the cell membrane and the carboxy terminal is inside. Sites of glycosylation are indicated by a Y and sites of phosphorylation by P in a circle. There is a site of palmitoylation in the carboxyl terminal tail indicated by two Cs anchored to the membrane. The natural CCK peptide is indicated as a brown oval docked to the receptor. Reproduced, with permission, from reference (62).


Figure 2. CCK1R signals through multiple G proteins to activate different signaling cascades. Whether β‐Arrestin and Gβ/γ play a role in pancreatic acinar cells is unclear (368).


Figure 3. Ca2+ oscillations induced by pmol/L concentrations of CCK that are initially independent of extracellular Ca2+. Ca2+ was removed and EGTA added 1 min before CCK was added. Reproduced, with permission, from reference (323).


Figure 4. CCK initiates Ca2+ waves that travel around an acini. (A) Ca2+ signaling shown for a single cell was typical of all cells in an acinar cluster; however, they did not occur synchronously. (B) Pseudocolor image of Ca2+ concentration shows that superfusion with 20 pmol/L CCK initiates a signal in one cell that then propagates in each cell around the cluster. After a basal period, a new cycle is initiated in the same cell. Reproduced, with permission, from reference (372).


Figure 5. Intracellular Ca2+ signaling in pancreatic acinar cells is initiated by the binding of Acetylcholine to Muscarinic M3 receptors (M3R) and by Cholecystokinin (CCK) to CCK receptors, predominately the CCK1R in mice and rats. Both receptors are classical seven transmembrane domain receptors coupled to guanine nucleotide‐binding proteins (G proteins). Activation of both receptors leads to stimulation of Gαq and increased activity of phospholipase C‐β (PLC) which cleaves the membrane phospholipid phosphatidylinositol,4,5,‐bisphosphate (PIP2) into diacylglycerol and inositol 1,4,5‐trisphosphate (IP3). IP3 diffuses through the cytoplasm and interacts with InsP3 receptors (IP3R), largely type‐2 and ‐3 present predominantly on the apical endoplasmic reticulum (ER) resulting in Ca2+ release into the cytoplasm. Ca2+ release from IP3R acts a co‐agonist to increase further IP3R activity and also acts on Ryanodine receptors (RyR) to induce Ca2+ release. Depletion of Ca2+ within the ER results in Ca2 + influx from the extracellular space. The ER Ca2+ sensor has been identified as stromal interaction molecule‐1 (Stim‐1). Following ER depletion, Ca2+ is released from an EF hand present in a domain of Stim‐1 within the ER lumen and this results in aggregation of several Stim‐1 molecules. Aggregation of Stim‐1 is sufficient to gate plasma membrane Ca2+ channels and leads to Ca2+ influx. Good candidates for the channel pore are the proteins Orai‐1 and TRPC3. CCK receptor stimulation also stimulates ADP‐ribosyl cyclase activity resulting in the formation of two additional Ca2+ mobilizing second messengers; nicotinic acid adenine dinucleotide phosphate (NAADP) and cyclic‐ADP ribose (cADPr). The particular cyclase responsible has not been defined in pancreas but good candidates include CD38 and CD157. cADPr is generally thought to act on RyR, while the target of NAADP is currently the subject of intense research. Candidates include the RyR and Two Pore Channel (TPC). In addition to the ER, Ca2+ can also be released from a store, which accumulates Ca2+ in a manner dependent on a proton gradient‐ known as the “acidic store.” This pool likely represents the endolysosomal compartment. This pool has been reported to be responsive to IP3, cADPr and NAADP and may represent the store initially released following receptor stimulation. Ca2+ is removed from the cytoplasm by the concerted action of SERCA (ER Ca2+‐ATPase), PMCA (PM Ca2+‐ATPase), and the action of MCU (mitochondrial uniporter). Figure and legend reproduced, with permission, from reference (368).


Figure 6. Activation of PKC isoforms in pancreatic acinar cells. CCK and ACh activate PLCβ and EGF activates PLCγ all of which lead to production of diacylglycerol (DAG) from PIP2. CCK also induces DAG formation from phosphatidylcholine (PC). Ca2+ and DAG together activate the classical isoforms PKC α, β, and γ; DAG activates the novel isoforms PKC δ, ϵ, η, and θ; while unconventional isoforms PKC ζ and λ do not require Ca2+ or DAG. Reproduced, with permission, from Pancreapedia, DAG–PKC Signaling Pathway, 2013.


Figure 7. Parallel nature of MAPK cascades in pancreas. Modified and updated, with permission, from reference (281).


Figure 8. Activation of ERK signaling pathway by CCK and EGF in pancreatic acinar cells. CCK1R signals through PKC and the activation by phosphorylation of Raf isoforms. Raf sequentially phosphorylates and activates MEK1/2 and ERK1/2. By contrast, EGFR autophosphorylates and then binds Shc, Grb2 and SOS with the latter being a GEF for Ras; active Ras binds and activates Raf and the kinase cascade ending in ERK. Activated ERK phosphorylates cytoplasmic substrates and downstream kinases, which phosphorylates transcription factors thereby activating expression of specific genes. Reproduced, with permission, from reference (345).


Figure 9. General and simplified diagram of mTORC1 pathway set in pancreatic acinar cell, where the pathway is regulated by CCK and insulin. Red arrows indicate activation; Black arrows indicate inhibition; Green arrow indicates translocation. Biological processes regulated by mTORC1 are shown at the bottom of the figure, along with the key proteins mediating the effect. Reproduced, with permission, from reference (273).


Figure 10. Activation of the Ca2+‐calcineurin‐NFAT signaling pathway by CCK and cholinergic agonists in pancreatic acinar cells. The initial steps in the pathway involve receptor‐mediated elevation of intracellular Ca2+. Ca2+ binds to calmodulin (CaM) and the complex to the Ca2 + activated phosphatase calcineurin. Calcineurin can dephosphorylate a number of substrates but the most generally important are NFATS which when dephosphorylated move into the nucleus and bind alone or together with coactivators such as AP‐1 to activate specific genes including FGF21, Socs3, Rgs2, Hbegf and Rcan1 with the latter feeding back to inhibit calcineurin. Reproduced, with permission, from the Pancreapedia, Calcium–Calcineurin–NFAT Signaling Pathway, 2012.


Figure 11. Rho Activation pathway by CCK and ACh. The CCK1 and muscarinic M3 receptors activate Gα12/13, which activate a Rho GEF of which several present in the pancreas are shown. Rho with bound GTP can activate mDia1 or ROCK (Rho kinase) thereby regulating the actin cytoskeleton or activate SRF (serum response factor) and activate gene expression. GTP bound Rho is inactivated by a Rho GAP of which several present in pancreas are shown. Botulinum exotoxin can permanently inactivate Rho. Reproduced, with permission, from Pancreapedia, Galpha12/13‐Rho Signaling Pathway, 2010.


Figure 12. Activation of cAMP pathway in pancreatic acinar and duct cells. Secretin, VIP and CCK bind to their receptors which activate adenylyl cyclase (AC) through Gαs while Somatostatin acts on SomstostatinR2 to activate Gαi which inhibits AC. Cholera toxin activates Gαs while forskolin directly activates AC and Pertussis toxin inhibits Gαi. AC produces cyclic AMP (cAMP) which is broken down by phosphodiesterase (PDE) which can be inhibited by isobutylmethylxanthine (IBMX). cAMP activates PKA which can phosphorylate a number of proteins including CREB, CFTR and the IP3 receptor. cAMP also activates EPAC1 which acts as a GEF to activate the small G protein Rap1 present on zymogen granules. Reproduced, with permission, from Pancreapedia, cAMP Signaling Pathway, 2012.
References
 1. Adler G , Beglinger C , Braun U , Reinshagen M , Koop I , Schafmayer A , Rovati L , Arnold R . Interaction of the cholinergic system and cholecystokinin in the regulation of endogenous and exogenous stimulation of pancreatic secretion in humans. Gastroenterology 100: 537‐543, 1991.
 2. Aktories K , Hall A . Botulinum ADP‐ribosyltransferase C3: A new tool to study low molecular weight GTP‐binding proteins. Trends Pharmacol Sci 10: 415‐418, 1989.
 3. Alexandre M , Thrower EC . PKC. Pancreapedia: Exocrine Pancreas Knowledge Base, DOI: 10.3998/panc.2012.13, 2012.
 4. Amsterdam A , Jamieson JD . Structural and functional characterization of isolated pancreatic exocrine cells. Proc Natl Acad Sci U S A 69: 3028‐3032, 1972.
 5. Antal CE , Newton AC . Tuning the signalling output of protein kinase C. Biochem Soc Trans 42: 1477‐1483, 2014.
 6. Aparicio IM , Garcia‐Marin LJ , Andreolotti AG , Bodega G , Jensen RT , Bragado MJ . Hepatocyte growth factor activates several transduction pathways in rat pancreatic acini. Biochim Biophys Acta 1643: 37‐46, 2003.
 7. Argent BE , Gray MA , MSteward MC , Case RM . Cell physiology of pancreatic ducts. In: Physiol of the Gastrointest Tract, edited by Johnson LR. Boston: Academic Press, 2012, pp. 1399‐1423.
 8. Ashby MC , Camello‐Almaraz C , Gerasimenko OV , Petersen OH , Tepikin AV . Long distance communication between muscarinic receptors and Ca2+ release channels revealed by carbachol uncaging in cell‐attached patch pipette. J Biol Chem 278: 20860‐20864, 2003.
 9. Aslan JE , McCarty OJ . Rho GTPases in platelet function. J Thromb Haemost 11: 35‐46, 2013.
 10. Bar‐Peled L , Sabatini DM . Regulation of mTORC1 by amino acids. Trends Cell Biol 24: 400‐406, 2014.
 11. Bartolome A , Guillen C . Role of the mammalian target of rapamycin (mTOR) complexes in pancreatic beta‐cell mass regulation. Vitam Horm 95: 425‐469, 2014.
 12. Bastani B , Yang L , Baldassare JJ , Pollo DA , Gardner JD . Cellular distribution of isoforms of protein kinase C (PKC) in pancreatic acini. Biochim Biophys Acta 1269: 307‐315, 1995.
 13. Baumler MD , Nelson DW , Ney DM , Groblewski GE . Loss of exocrine pancreatic stimulation during parenteral feeding suppresses digestive enzyme expression and induces Hsp70 expression. Am J Physiol Gastrointest Liver Physiol 292: G857‐G866, 2007.
 14. Beglinger C , Fried M , Whitehouse I , Jansen JB , Lamers CB , Gyr K . Pancreatic enzyme response to a liquid meal and to hormonal stimulation. Correlation with plasma secretin and cholecystokinin levels. J Clin Invest 75: 1471‐1476, 1985.
 15. Bentzinger CF , Romanino K , Cloetta D , Lin S , Mascarenhas JB , Oliveri F , Xia J , Casanova E , Costa CF , Brink M , Zorzato F , Hall MN , Ruegg MA . Skeletal muscle‐specific ablation of raptor, but not of rictor, causes metabolic changes and results in muscle dystrophy. Cell Metab 8: 411‐424, 2008.
 16. Berna MJ , Hoffmann KM , Tapia JA , Thill M , Pace A , Mantey SA , Jensen RT . CCK causes PKD1 activation in pancreatic acini by signaling through PKC‐delta and PKC‐independent pathways. Biochim Biophys Acta 1773: 483‐501, 2007.
 17. Berridge MJ . Rapid accumulation of inositol trisphosphate reveals that agonists hydrolyse polyphosphoinositides instead of phosphatidylinositol. Biochem J 212: 849‐858, 1983.
 18. Berridge MJ. Inositol trisphosphate and diacylglycerol: Two interacting second messengers. Annu Rev Biochem 56: 159‐193, 1987.
 19. Bi Y , Page SL , Williams JA . Rho and Rac promote acinar morphological changes, actin reorganization, and amylase secretion. Am J Physiol Gastrointest Liver Physiol 289: G561‐G570, 2005.
 20. Bi Y , Williams JA . A role for Rho and Rac in secretagogue‐induced amylase release by pancreatic acini. Am J Physiol Cell Physiol 289: C22‐C32, 2005.
 21. Blandino‐Rosano M , Barbaresso R , Jimenez‐Palomares M , Bozadjieva N , Werneck‐de‐Castro JP , Hatanaka M , Mirmira RG , Sonenberg N , Liu M , Ruegg MA , Hall MN , Bernal‐Mizrachi E . Loss of mTORC1 signalling impairs beta‐cell homeostasis and insulin processing. Nat Commun 8: 16014, 2017.
 22. Bragado MJ , Dabrowski A , Groblewski GE , Williams JA . CCK activates p90rsk in rat pancreatic acini through protein kinase C. Am J Physiol 272: G401‐G407, 1997.
 23. Bragado MJ , Groblewski GE , Williams JA . p70s6k is activated by CCK in rat pancreatic acini. Am J Physiol 273: C101‐C109, 1997.
 24. Bragado MJ , Groblewski GE , Williams JA . Regulation of protein synthesis by cholecystokinin in rat pancreatic acini involves PHAS‐I and the p70 S6 kinase pathway. Gastroenterology 115: 733‐742, 1998.
 25. Bragado MJ , Tashiro M , Williams JA . Regulation of the initiation of pancreatic digestive enzyme protein synthesis by cholecystokinin in rat pancreas in vivo. Gastroenterology 119: 1731‐1739, 2000.
 26. Brown EJ , Albers MW , Shin TB , Ichikawa K , Keith CT , Lane WS , Schreiber SL . A mammalian protein targeted by G1‐arresting rapamycin‐receptor complex. Nature 369: 756‐758, 1994.
 27. Bruce JIE. Metabolic regulation of the PMCA: Role in cell death and survival. Cell Calcium 69: 28‐36, 2018.
 28. Bruzzone R , Regazzi R , Wollheim CB . Caerulein causes translocation of protein kinase C in rat acini without increasing cytosolic free Ca2+. Am J Physiol 255: G33‐G39, 1988.
 29. Burnham DB . Characterization of Ca2+‐activated protein phosphatase activity in exocrine pancreas. Biochem J 231: 335‐341, 1985.
 30. Busca R , Pouyssegur J , Lenormand P . ERK1 and ERK2 map kinases: Specific roles or functional redundancy? Front Cell Dev Biol 4: 53, 2016.
 31. Cantor P , Olsen O , Gertz BJ , Gjorup I , Worning H . Inhibition of cholecystokinin‐stimulated pancreaticobiliary output in man by the cholecystokinin receptor antagonist MK‐329. Scand J Gastroenterol 26: 627‐637, 1991.
 32. Carriere A , Romeo Y , Acosta‐Jaquez HA , Moreau J , Bonneil E , Thibault P , Fingar DC , Roux PP . ERK1/2 phosphorylate Raptor to promote Ras‐dependent activation of mTOR complex 1 (mTORC1). J Biol Chem 286: 567‐577, 2011.
 33. Caunt CJ , Keyse SM . Dual‐specificity MAP kinase phosphatases (MKPs): Shaping the outcome of MAP kinase signalling. FEBS J 280: 489‐504, 2013.
 34. Cawston EE , Lam PC , Harikumar KG , Dong M , Ball AM , Augustine ML , Akgun E , Portoghese PS , Orry A , Abagyan R , Sexton PM , Miller LJ . Molecular basis for binding and subtype selectivity of 1,4‐benzodiazepine antagonist ligands of the cholecystokinin receptor. J Biol Chem 287: 18618‐18635, 2012.
 35. Cendrowski J , Lobo VJ , Sendler M , Salas A , Kuhn JP , Molero X , Fukunaga R , Mayerle J , Lerch MM , Real FX . Mnk1 is a novel acinar cell‐specific kinase required for exocrine pancreatic secretion and response to pancreatitis in mice. Gut 64: 937‐947, 2015.
 36. Chandra R , Samsa LA , Vigna SR , Liddle RA . Pseudopod‐like basal cell processes in intestinal cholecystokinin cells. Cell Tissue Res 341: 289‐297, 2010.
 37. Chandrasekhar R , Yule DI , Wang L . Inositol 1,4,5‐trisphosphate receptors (InsP3R). Pancreapedia: Exocrine Pancreas Knowledge Base, DOI: 10.3998/panc.2017.07, 2017.
 38. Chang TM , Chey WY . Radioimmunoassay of cholecystokinin. Dig Dis Sci 28: 456‐468, 1983.
 39. Chen LA , Li J , Silva SR , Jackson LN , Zhou Y , Watanabe H , Ives KL , Hellmich MR , Evers BM . PKD3 is the predominant protein kinase D isoform in mouse exocrine pancreas and promotes hormone‐induced amylase secretion. J Biol Chem 284: 2459‐2471, 2009.
 40. Chey WY , Chang T‐M . Secretin, 100 years later. Journal of Gastroenterology 38: 1025‐1035, 2003.
 41. Chin D , Means AR . Calmodulin: A prototypical calcium sensor. Trends Cell Biol 10: 322‐328, 2000.
 42. Cosen‐Binker LI , Lam PP , Binker MG , Gaisano HY . Alcohol‐induced protein kinase Calpha phosphorylation of Munc18c in carbachol‐stimulated acini causes basolateral exocytosis. Gastroenterology 132: 1527‐1545, 2007.
 43. Cosen‐Binker LI , Lam PP , Binker MG , Reeve J , Pandol S , Gaisano HY . Alcohol/cholecystokinin‐evoked pancreatic acinar basolateral exocytosis is mediated by protein kinase C alpha phosphorylation of Munc18c. J Biol Chem 282: 13047‐13058, 2007.
 44. Crozier SJ , D'Alecy LG , Ernst SA , Ginsburg LE , Williams JA . Molecular mechanisms of pancreatic dysfunction induced by protein malnutrition. Gastroenterology 137: 1093‐1101, 1101 e1091‐1093, 2009.
 45. Crozier SJ , Sans MD , Guo L , D'Alecy LG , Williams JA . Activation of the mTOR signalling pathway is required for pancreatic growth in protease‐inhibitor‐fed mice. J Physiol 573: 775‐786, 2006.
 46. Crozier SJ , Sans MD , Lang CH , D'Alecy LG , Ernst SA , Williams JA . CCK‐induced pancreatic growth is not limited by mitogenic capacity in mice. Am J Physiol Gastrointest Liver Physiol 294: G1148‐1157, 2008.
 47. Crozier SJ , Sans MD , Wang JY , Lentz SI , Ernst SA , Williams JA . CCK‐independent mTORC1 activation during dietary protein‐induced exocrine pancreas growth. Am J Physiol Gastrointest Liver Physiol 299: G1154‐G1163, 2010.
 48. Cuadrado A , Nebreda AR . Mechanisms and functions of p38 MAPK signalling. Biochem J 429: 403‐417, 2010.
 49. Dabrowski A , Boguslowicz C , Dabrowska M , Tribillo I , Gabryelewicz A . Reactive oxygen species activate mitogen‐activated protein kinases in pancreatic acinar cells. Pancreas 21: 376‐384, 2000.
 50. Dabrowski A , Detjen KM , Logsdon CD , Williams JA . Stimulation of both CCK‐A and CCK‐B receptors activates MAP kinases in AR42J and receptor‐transfected CHO cells. Digestion 58: 361‐367, 1997.
 51. Dabrowski A , Grady T , Logsdon CD , Williams JA . Jun kinases are rapidly activated by cholecystokinin in rat pancreas both in vitro and in vivo. J Biol Chem 271: 5686‐5690, 1996.
 52. Dabrowski A , Groblewski GE , Schafer C , Guan KL , Williams JA . Cholecystokinin and EGF activate a MAPK cascade by different mechanisms in rat pancreatic acinar cells. Am J Physiol 273: C1472‐C1479, 1997.
 53. Dabrowski A , Tribillo I , Dabrowska MI , Wereszczynska‐Siemiatkowska U , Gabryelewicz A . Activation of mitogen‐activated protein kinases in different models of pancreatic acinar cell damage. Z Gastroenterol 38: 469‐481, 2000.
 54. Dabrowski A , VanderKuur JA , Carter‐Su C , Williams JA . Cholecystokinin stimulates formation of shc‐grb2 complex in rat pancreatic acinar cells through a protein kinase C‐dependent mechanism. J Biol Chem 271: 27125‐27129, 1996.
 55. Daniluk J , Dabrowski A . The effect of concomitant stimulation with cholecystokinin and epidermal growth factor on extracellular signal‐regulated kinase (ERK) activity in pancreatic acinar cells. J Physiol Pharmacol 58: 441‐453, 2007.
 56. Daniluk J , Liu Y , Deng D , Chu J , Huang H , Gaiser S , Cruz‐Monserrate Z , Wang H , Ji B , Logsdon CD . An NF‐kappaB pathway‐mediated positive feedback loop amplifies Ras activity to pathological levels in mice. J Clin Invest 122: 1519‐1528, 2012.
 57. Daulhac L , Kowalski‐Chauvel A , Pradayrol L , Vaysse N , Seva C . Src‐family tyrosine kinases in activation of ERK‐1 and p85/p110‐phosphatidylinositol 3‐kinase by G/CCKB receptors. J Biol Chem 274: 20657‐20663, 1999.
 58. Davis RJ . Signal transduction by the JNK group of MAP kinases. Cell 103: 239‐252, 2000.
 59. Dawson ES , Henne RM , Miller LJ , Lybrand TP . Molecular models for cholecystokinin‐A receptor. Pharmacology & Toxicology 91: 290‐296, 2002.
 60. de Lartigue G , Barbier de la Serre C , Espero E , Lee J , Raybould HE . Leptin resistance in vagal afferent neurons inhibits cholecystokinin signaling and satiation in diet induced obese rats. PLoS One 7: e32967, 2012.
 61. Desai AJ , Dong M , Langlais BT , Dueck AC , Miller LJ . Cholecystokinin responsiveness varies across the population dependent on metabolic phenotype. Am J Clin Nutr 106: 447‐456, 2017.
 62. Desai AJ , Miller LJ . Cholecystokinin type 1 receptor. Pancreapedia: Exocrine Pancreas Knowledge Base, DOI: 10.3998/panc.2013.9, 2013.
 63. Desai AJ , Miller LJ . Changes in the plasma membrane in metabolic disease: Impact of the membrane environment on G protein‐coupled receptor structure and function. Br J Pharmacol 175: 4009‐4025, 2017.
 64. Dibble CC , Manning BD . Signal integration by mTORC1 coordinates nutrient input with biosynthetic output. Nat Cell Biol 15: 555‐564, 2013.
 65. Dirice E , Walpita D , Vetere A , Meier BC , Kahraman S , Hu J , Dancik V , Burns SM , Gilbert TJ , Olson DE , Clemons PA , Kulkarni RN , Wagner BK . Inhibition of DYRK1A stimulates human beta‐cell proliferation. Diabetes 65: 1660‐1671, 2016.
 66. Dockray GJ . The action of scretin, cholecystokinin‐pancreozymin and caerulein on pancreatic secretion in the rat. J Physiol 225: 679‐692, 1972.
 67. Dockray GJ . Gastrointestinal hormones: Gastrin, cholecystokinin, somatostatin, and ghrelin. In: Physiology of the Gastrointestinal Tract (Fourth Edition). Ed. Johnson LR , Burlington: Academic Press, 2006, pp. 91‐120.
 68. Dockray GJ . Cholecystokinin and gut‐brain signalling. Regul Pept 155: 6‐10, 2009.
 69. Dormer RL , Williams JA . Secretagogue‐induced changes in subcellular Ca2+ distribution in isolated pancreatic acini. Am J Physiol 240: G130‐G140, 1981.
 70. Duan RD , Williams JA . Cholecystokinin rapidly activates mitogen‐activated protein kinase in rat pancreatic acini. Am J Physiol 267: G401‐G408, 1994.
 71. Duan RD , Zheng CF , Guan KL , Williams JA . Activation of MAP kinase kinase (MEK) and Ras by cholecystokinin in rat pancreatic acini. Am J Physiol 268: G1060‐G1065, 1995.
 72. Dudley DT , Pang L , Decker SJ , Bridges AJ , Saltiel AR . A synthetic inhibitor of the mitogen‐activated protein kinase cascade. Proc Natl Acad Sci U S A 92: 7686‐7689, 1995.
 73. Dufresne M , Seva C , Fourmy D . Cholecystokinin and gastrin receptors. Physiol Rev 86: 805‐847, 2006.
 74. Eberlein GA , Eysselein VE , Hesse WH , Goebell H , Schaefer M , Reeve JR, Jr. Detection of cholecystokinin‐58 in human blood by inhibition of degradation. Am J Physiol 253: G477‐G482, 1987.
 75. Elghazi L , Blandino‐Rosano M , Alejandro E , Cras‐Meneur C , Bernal‐Mizrachi E . Role of nutrients and mTOR signaling in the regulation of pancreatic progenitors development. Mol Metab 6: 560‐573, 2017.
 76. Escos A , Risco A , Alsina‐Beauchamp D , Cuenda A . p38gamma and p38delta mitogen activated protein kinases (MAPKs), new stars in the MAPK galaxy. Front Cell Dev Biol 4: 31, 2016.
 77. Etienne‐Manneville S , Hall A . Rho GTPases in cell biology. Nature 420: 629‐635, 2002.
 78. Fan BG , Axelson J , Sternby B , Rehfeld J , Ihse I , Ekelund M . Total parenteral nutrition affects the tropic effect of cholecystokinin on the exocrine pancreas. Scand J Gastroenterol 32: 380‐386, 1997.
 79. Fingar DC , Blenis J . Target of rapamycin (TOR): An integrator of nutrient and growth factor signals and coordinator of cell growth and cell cycle progression. Oncogene 23: 3151‐3171, 2004.
 80. Fleming AK , Storz P . Protein kinase C isoforms in the normal pancreas and in pancreatic disease. Cell Signal 40: 1‐9, 2017.
 81. Fogarty KE , Kidd JF , Tuft DA , Thorn P . Mechanisms underlying InsP3‐evoked global Ca2+ signals in mouse pancreatic acinar cells. J Physiol 526 Pt 3: 515‐526, 2000.
 82. Folsch UR , Winckler K , Wormsley KG . Influence of repeated administration of cholecystokinin and secretin on the pancreas of the rat. Scand J Gastroenterol 13: 663‐671, 1978.
 83. Fukuhara S , Chikumi H , Gutkind JS . RGS‐containing RhoGEFs: The missing link between transforming G proteins and Rho? Oncogene 20: 1661‐1668, 2001.
 84. Furuichi T , Yoshikawa S , Miyawaki A , Wada K , Maeda N , Mikoshiba K . Primary structure and functional expression of the inositol 1,4,5‐trisphosphate‐binding protein P400. Nature 342: 32‐38, 1989.
 85. Futatsugi A , Nakamura T , Yamada MK , Ebisui E , Nakamura K , Uchida K , Kitaguchi T , Takahashi‐Iwanaga H , Noda T , Aruga J , Mikoshiba K . IP3 receptor types 2 and 3 mediate exocrine secretion underlying energy metabolism. Science 309: 2232‐2234, 2005.
 86. Gaestel M . MAPKAP kinases‐MKs‐two's company, three's a crowd. Nat Rev Mol Cell Biol 7: 120‐130, 2006.
 87. Gallo KA , Johnson GL . Mixed‐lineage kinase control of JNK and p38 MAPK pathways. Nat Rev Mol Cell Biol 3: 663‐672, 2002.
 88. Garcia LJ , Rosado JA , Gonzalez A , Jensen RT . Cholecystokinin‐stimulated tyrosine phosphorylation of p125FAK and paxillin is mediated by phospholipase C‐dependent and ‐independent mechanisms and requires the integrity of the actin cytoskeleton and participation of p21rho. Biochem J 327(Pt 2): 461‐472, 1997.
 89. Gardner JD , Sutliff VE , Walker MD , Jensen RT . Effects of inhibitors of cyclic nucleotide phosphodiesterase on actions of cholecystokinin, bombesin, and carbachol on pancreatic acini. Am J Physiol 245: G676‐G680, 1983.
 90. Gazzaneo MC , Orellana RA , Suryawan A , Tuckow AP , Kimball SR , Wilson FA , Nguyen HV , Torrazza RM , Fiorotto ML , Davis TA . Differential regulation of protein synthesis and mTOR signaling in skeletal muscle and visceral tissues of neonatal pigs after a meal. Pediatr Res 70: 253‐260, 2011.
 91. Gerasimenko JV , Gryshchenko O , Ferdek PE , Stapleton E , Hebert TO , Bychkova S , Peng S , Begg M , Gerasimenko OV , Petersen OH . Ca2+ release‐activated Ca2+ channel blockade as a potential tool in antipancreatitis therapy. Proc Natl Acad Sci U S A 110: 13186‐13191, 2013.
 92. Geron E , Schejter ED , Shilo BZ . Directing exocrine secretory vesicles to the apical membrane by actin cables generated by the formin mDia1. Proc Natl Acad Sci U S A 110: 10652‐10657, 2013.
 93. Goberdhan DC , Wilson C , Harris AL . Amino acid sensing by mTORC1: Intracellular transporters mark the spot. Cell Metab 23: 580‐589, 2016.
 94. Gonzalez A , Schmid A , Sternfeld L , Krause E , Salido GM , Schulz I . Cholecystokinin‐evoked Ca(2+) waves in isolated mouse pancreatic acinar cells are modulated by activation of cytosolic phospholipase A(2), phospholipase D, and protein kinase C. Biochem Biophys Res Commun 261: 726‐733, 1999.
 95. Goodyer WR , Gu X , Liu Y , Bottino R , Crabtree GR , Kim SK . Neonatal beta cell development in mice and humans is regulated by calcineurin/NFAT. Dev Cell 23: 21‐34, 2012.
 96. Grady T , Dabrowski A , Williams JA , Logsdon CD . Stress‐activated protein kinase activation is the earliest direct correlate to the induction of secretagogue‐induced pancreatitis in rats. Biochem Biophys Res Commun 227: 1‐7, 1996.
 97. Green GM , Levan VH , Liddle RA . Plasma cholecystokinin and pancreatic growth during adaptation to dietary protein. Am J Physiol 251: G70‐G74, 1986.
 98. Green GM , Lyman RL . Feedback regulation of pancreatic enzyme secretion as a mechanism for trypsin inhibitor‐induced hypersecretion in rats. Proc Soc Exp Biol Med 140: 6‐12, 1972.
 99. Gresset A , Sondek J , Harden TK . The phospholipase C isozymes and their regulation. Subcell Biochem 58: 61‐94, 2012.
 100. Groblewski GE , Grady T , Mehta N , Lambert H , Logsdon CD , Landry J , Williams JA . Cholecystokinin stimulates heat shock protein 27 phosphorylation in rat pancreas both in vivo and in vitro. Gastroenterology 112: 1354‐1361, 1997.
 101. Groblewski GE , Wagner AC , Williams JA . Cyclosporin A inhibits Ca2+/calmodulin‐dependent protein phosphatase and secretion in pancreatic acinar cells. J Biol Chem 269: 15111‐15117, 1994.
 102. Groblewski GE , Yoshida M , Bragado MJ , Ernst SA , Leykam J , Williams JA . Purification and characterization of a novel physiological substrate for calcineurin in mammalian cells. J Biol Chem 273: 22738‐22744, 1998.
 103. Guo L , Sans MD , Gurda GT , Lee SH , Ernst SA , Williams JA . Induction of early response genes in trypsin inhibitor‐induced pancreatic growth. Am J Physiol Gastrointest Liver Physiol 292: G667‐G677, 2007.
 104. Guo L , Sans MD , Hou Y , Ernst SA , Williams JA . c‐Jun/AP‐1 is required for CCK‐induced pancreatic acinar cell dedifferentiation and DNA synthesis in vitro. Am J Physiol Gastrointest Liver Physiol 302: G1381‐G1396, 2012.
 105. Gurda GT , Crozier SJ , Ji B , Ernst SA , Logsdon CD , Rothermel BA , Williams JA . Regulator of calcineurin 1 controls growth plasticity of adult pancreas. Gastroenterology 139: 609‐619, 619 e601‐606, 2010.
 106. Gurda GT , Guo L , Lee SH , Molkentin JD , Williams JA . Cholecystokinin activates pancreatic calcineurin‐NFAT signaling in vitro and in vivo. Mol Biol Cell 19: 198‐206, 2008.
 107. Haga RB , Ridley AJ . Rho GTPases: Regulation and roles in cancer cell biology. Small GTPases 7: 207‐221, 2016.
 108. Halbrook CJ , Wen HJ , Ruggeri JM , Takeuchi KK , Zhang Y , di Magliano MP , Crawford HC . Mitogen‐activated protein kinase kinase activity maintains acinar‐to‐ductal metaplasia and is required for organ regeneration in pancreatitis. Cell Mol Gastroenterol Hepatol 3: 99‐118, 2017.
 109. Hara K , Maruki Y , Long X , Yoshino K , Oshiro N , Hidayat S , Tokunaga C , Avruch J , Yonezawa K . Raptor, a binding partner of target of rapamycin (TOR), mediates TOR action. Cell 110: 177‐189, 2002.
 110. Harada E , Kanno T . Progressive enhancement in the secretory functions of the digestive system of the rat in the course of cold acclimation. J Physiol 260: 629‐645, 1976.
 111. Harikumar KG , Cawston EE , Lam PC , Patil A , Orry A , Henke BR , Abagyan R , Christopoulos A , Sexton PM , Miller LJ . Molecular basis for benzodiazepine agonist action at the type 1 cholecystokinin receptor. J Biol Chem 288: 21082‐21095, 2013.
 112. Harper AA , Raper HS . Pancreozymin, a stimulant of the secretion of pancreatic enzymes in extracts of the small intestine. J Physiol 102: 115‐125, 1943.
 113. Hashi M , Yoshizawa F , Onozuka E , Ogata M , Hara H . Adaptive changes in translation initiation activities for rat pancreatic protein synthesis with feeding of a high‐protein diet. J Nutr Biochem 16: 507‐512, 2005.
 114. Hashimoto N , Hara H . Dietary amino acids promote pancreatic protease synthesis at the translation stage in rats. J Nutr 133: 3052‐3057, 2003.
 115. Heit JJ , Apelqvist AA , Gu X , Winslow MM , Neilson JR , Crabtree GR , Kim SK . Calcineurin/NFAT signalling regulates pancreatic beta‐cell growth and function. Nature 443: 345‐349, 2006.
 116. Henin J , Maigret B , Tarek M , Escrieut C , Fourmy D , Chipot C . Probing a model of a GPCR/ligand complex in an explicit membrane environment: The human cholecystokinin‐1 receptor. Biophys J 90: 1232‐1240, 2006.
 117. Hildebrand P , Beglinger C , Gyr K , Jansen JB , Rovati LC , Zuercher M , Lamers CB , Setnikar I , Stalder GA . Effects of a cholecystokinin receptor antagonist on intestinal phase of pancreatic and biliary responses in man. J Clin Invest 85: 640‐646, 1990.
 118. Himeno S , Tarui S , Kanayama S , Kuroshima T , Shinomura Y , Hayashi C , Tateishi K , Imagawa K , Hashimura E , Hamaoka T . Plasma cholecystokinin responses after ingestion of liquid meal and intraduodenal infusion of fat, amino acids, or hydrochloric acid in man: Analysis with region specific radioimmunoassay. Am J Gastroenterol 78: 703‐707, 1983.
 119. Hocker M , Waschulewski IH , Kern HF , Domagk KA , Schwarzhoff R , Folsch UR , Schmidt WE . Cyclosporine A inhibits protein‐kinase‐C‐mediated amylase release from isolated rat pancreatic acini. Digestion 55: 380‐388, 1994.
 120. Hodge RG , Ridley AJ . Regulating Rho GTPases and their regulators. Nat Rev Mol Cell Biol 17: 496‐510, 2016.
 121. Hoffmann P , Eberlein GA , Reeve JR, Jr. , Bunte RH , Grandt D , Goebell H , Eysselein VE . Comparison of clearance and metabolism of infused cholecystokinins 8 and 58 in dogs. Gastroenterology 105: 1732‐1736, 1993.
 122. Hofken T , Keller N , Fleischer F , Goke B , Wagner AC . Map kinase phosphatases (MKP's) are early responsive genes during induction of cerulein hyperstimulation pancreatitis. Biochem Biophys Res Commun 276: 680‐685, 2000.
 123. Holtz BJ , Lodewyk KB , Sebolt‐Leopold JS , Ernst SA , Williams JA . ERK activation is required for CCK‐mediated pancreatic adaptive growth in mice. Am J Physiol Gastrointest Liver Physiol 307: G700‐G710, 2014.
 124. Hong JH , Park S , Shcheynikov N , Muallem S . Mechanism and synergism in epithelial fluid and electrolyte secretion. Pflugers Arch 466: 1487‐1499, 2014.
 125. Hou H , Wang F , Zhang W , Wang D , Li X , Bartlam M , Yao X , Rao Z . Structure‐functional analyses of CRHSP‐24 plasticity and dynamics in oxidative stress response. J Biol Chem 286: 9623‐9635, 2011.
 126. Huang CY , Tan TH . DUSPs, to MAP kinases and beyond. Cell Biosci 2: 24, 2012.
 127. Husain SZ , Grant WM , Gorelick FS , Nathanson MH , Shah AU . Caerulein‐induced intracellular pancreatic zymogen activation is dependent on calcineurin. Am J Physiol Gastrointest Liver Physiol 292: G1594‐G1599, 2007.
 128. Ishizuka T , Ito Y , Kajita K , Miura K , Nagao S , Nagata K , Nozawa Y . Redistribution of protein kinase C in pancreatic acinar cells stimulated with caerulein or carbachol. Biochem Biophys Res Commun 144: 551‐559, 1987.
 129. Ivy AC . A hormone mechanism for gall‐bladder contraction & evacuation. Amer J Physiology 86: 599‐613, 1928.
 130. Jaffe AB , Hall A . Rho GTPases: Biochemistry and biology. Annu Rev Cell Dev Biol 21: 247‐269, 2005.
 131. Jang Y , Soekmadji C , Mitchell JM , Thomas WG , Thorn P . Real‐time measurement of F‐actin remodelling during exocytosis using Lifeact‐EGFP transgenic animals. PLoS One 7: e39815, 2012.
 132. Jebbink MC , Jansen JB , Masclee AA , Lamers CB . Lack of effect of the specific cholecystokinin receptor antagonist loxiglumide on cholecystokinin clearance from plasma in man. Br J Clin Pharmacol 29: 770‐773, 1990.
 133. Jensen RT , Wank SA , Rowley WH , Sato S , Gardner JD . Interaction of CCK with pancreatic acinar cells. Trends Pharmacol Sci 10: 418‐423, 1989.
 134. Jewell JL , Russell RC , Guan KL . Amino acid signalling upstream of mTOR. Nat Rev Mol Cell Biol 14: 133‐139, 2013.
 135. Ji B , Bi Y , Simeone D , Mortensen RM , Logsdon CD . Human pancreatic acinar cells lack functional responses to cholecystokinin and gastrin. Gastroenterology 121: 1380‐1390, 2001.
 136. Jun I , Lee MG , Muallem S . Molecular mechanisms of pancreatic bicarbonate secretion. Pancreapedia 2017.
 137. Kanayama S , Himeno S , Kurokawa M , Shinomura Y , Kuroshima T , Okuno M , Tsuji K , Higashimoto Y , Ikei N , Hashimura E , et al. Marked prolongation in disappearance half‐time of plasma cholecystokinin‐octapeptide in patients with hepatic cirrhosis. Am J Gastroenterol 80: 557‐560, 1985.
 138. Kasai H , Augustine GJ . Cytosolic Ca2+ gradients triggering unidirectional fluid secretion from exocrine pancreas. Nature 348: 735‐738, 1990.
 139. Kasai H , Li YX , Miyashita Y . Subcellular distribution of Ca2+ release channels underlying Ca2+ waves and oscillations in exocrine pancreas. Cell 74: 669‐677, 1993.
 140. Kaufmann A , Rossler OG , Thiel G . Expression of the transcription factor Egr‐1 in pancreatic acinar cells following stimulation of cholecystokinin or Galphaq‐coupled designer receptors. Cell Physiol Biochem 33: 1411‐1425, 2014.
 141. Kidger AM , Keyse SM . The regulation of oncogenic Ras/ERK signalling by dual‐specificity mitogen activated protein kinase phosphatases (MKPs). Semin Cell Dev Biol 50: 125‐132, 2016.
 142. Kiehne K , Herzig KH , Folsch UR . CCK‐stimulated changes in pancreatic acinar morphology are mediated by rho. Digestion 65: 47‐55, 2002.
 143. Kim J , Guan KL . Amino acid signaling in TOR activation. Annu Rev Biochem 80: 1001‐1032, 2011.
 144. Kim M , Nozu F , Kusama K , Imawari M . Cholecystokinin stimulates the recruitment of the Src‐RhoA‐phosphoinositide 3‐kinase pathway by Vav‐2 downstream of G(alpha13) in pancreatic acini. Biochem Biophys Res Commun 339: 271‐276, 2006.
 145. Kim MJ , Lee KH , Min DS , Yoon SH , Hahn SJ , Kim MS , Jo YH . Distributional patterns of phospholipase C isozymes in rat pancreas. Pancreas 22: 47‐52, 2001.
 146. Kim MJ , Lee YS , Lee KH , Min DS , Yoon SH , Hahn SJ , Kim MS , Jo YH . Site‐specific localization of protein kinase C isoforms in rat pancreas. Pancreatology 1: 36‐42, 2001.
 147. Kim MS , Hong JH , Li Q , Shin DM , Abramowitz J , Birnbaumer L , Muallem S . Deletion of TRPC3 in mice reduces store‐operated Ca2+ influx and the severity of acute pancreatitis. Gastroenterology 137: 1509‐1517, 2009.
 148. Kim YS , Kim WJ , Kim HK , Hong SS . Effect of cold and hot environments on the exocrine pancreas of rats. Yonsei Med J 11: 1‐9, 1970.
 149. Klee CB , Ren H , Wang X . Regulation of the calmodulin‐stimulated protein phosphatase, calcineurin. J Biol Chem 273: 13367‐13370, 1998.
 150. Kolch W . Coordinating ERK/MAPK signalling through scaffolds and inhibitors. Nat Rev Mol Cell Biol 6: 827‐837, 2005.
 151. Kraft AS , Anderson WB . Phorbol esters increase the amount of Ca2+, phospholipid‐dependent protein kinase associated with plasma membrane. Nature 301: 621‐623, 1983.
 152. Krishna M , Narang H . The complexity of mitogen‐activated protein kinases (MAPKs) made simple. Cell Mol Life Sci 65: 3525‐3544, 2008.
 153. Lacourse KA , Swanberg LJ , Gillespie PJ , Rehfeld JF , Saunders TL , Samuelson LC . Pancreatic function in CCK‐deficient mice: Adaptation to dietary protein does not require CCK. Am J Physiol Gastrointest Liver Physiol 276: G1302‐G1309, 1999.
 154. Laplante M , Sabatini DM . mTOR signaling in growth control and disease. Cell 149: 274‐293, 2012.
 155. Lawrence MC , Bhatt HS , Watterson JM , Easom RA . Regulation of insulin gene transcription by a Ca(2+)‐responsive pathway involving calcineurin and nuclear factor of activated T cells. Mol Endocrinol 15: 1758‐1767, 2001.
 156. Le Page SL , Bi Y , Williams JA . CCK‐A receptor activates RhoA through G alpha 12/13 in NIH3T3 cells. Am J Physiol Cell Physiol 285: C1197‐C1206, 2003.
 157. Lee M , Chung S , Uhm DY , Park MK . Regulation of zymogen granule exocytosis by Ca2+, cAMP, and PKC in pancreatic acinar cells. Biochem Biophys Res Commun 334: 1241‐1247, 2005.
 158. Lee MG , Xu X , Zeng W , Diaz J , Kuo TH , Wuytack F , Racymaekers L , Muallem S . Polarized expression of Ca2+ pumps in pancreatic and salivary gland cells. Role in initiation and propagation of [Ca2+]i waves. J Biol Chem 272: 15771‐15776, 1997.
 159. Lee S , Wishart MJ , Williams JA . Identification of calcineurin regulated phosphorylation sites on CRHSP‐24. Biochem Biophys Res Commun 385: 413‐417, 2009.
 160. Leser J , Luhrs H , Beil MF , Adler G , Lutz MP . Cholecystokinin‐induced redistribution of paxillin in rat pancreatic acinar cells. Biochem Biophys Res Commun 254: 400‐405, 1999.
 161. Li C , Chen X , Williams JA . Regulation of CCK‐induced amylase release by PKC‐delta in rat pancreatic acinar cells. Am J Physiol Gastrointest Liver Physiol 287: G764‐G771, 2004.
 162. Li C , Williams JA . Regulation of CCK‐induced ERK1/2 activation by PKC epsilon in rat pancreatic acinar cells. AIMS Molecular Sci 4: 463‐477, 2017.
 163. Li H , Rao A , Hogan PG . Interaction of calcineurin with substrates and targeting proteins. Trends Cell Biol 21: 91‐103, 2011.
 164. Li W , Zhang H , Nie A , Ni Q , Li F , Ning G , Li X , Gu Y , Wang Q . mTORC1 pathway mediates beta cell compensatory proliferation in 60 % partial‐pancreatectomy mice. Endocrine 53: 117‐128, 2016.
 165. Li Y , Owyang C . Vagal afferent pathway mediates physiological action of cholecystokinin on pancreatic enzyme secretion. J Clin Invest 92: 418‐424, 1993.
 166. Liang T , Dolai S , Xie L , Winter E , Orabi AI , Karimian N , Cosen‐Binker LI , Huang YC , Thorn P , Cattral MS , Gaisano HY . Ex vivo human pancreatic slice preparations offer a valuable model for studying pancreatic exocrine biology. J Biol Chem 292: 5957‐5969, 2017.
 167. Liddle RA . Regulation of cholecystokinin secretion by intraluminal releasing factors. Am J Physiol 269: G319‐G327, 1995.
 168. Liddle RA . Cholecystokinin cells. Annu Rev Physiol 59: 221‐242, 1997.
 169. Liddle RA . Regulation of Pancreatic Secretion. In: Physiology of the Gastrointestinal Tract (Fifth Edition), edited by Johnson LR , Ghishan FK , Kaunitz JD , Merchant JL , Said HM , Wood JD . Boston: Academic Press, 2012, pp. 1425‐1460.
 170. Liddle RA , Goldfine ID , Rosen MS , Taplitz RA , Williams JA . Cholecystokinin bioactivity in human plasma. Molecular forms, responses to feeding, and relationship to gallbladder contraction. J Clin Invest 75: 1144‐1152, 1985.
 171. Liddle RA , Goldfine ID , Williams JA . Bioassay of plasma cholecystokinin in rats: Effects of food, trypsin inhibitor, and alcohol. Gastroenterology 87: 542‐549, 1984.
 172. Liener IE , Goodale RL , Deshmukh A , Satterberg TL , Ward G , DiPietro CM , Bankey PE , Borner JW . Effect of a trypsin inhibitor from soybeans (Bowman‐Birk) on the secretory activity of the human pancreas. Gastroenterology 94: 419‐427, 1988.
 173. Liou J , Kim ML , Heo WD , Jones JT , Myers JW , Ferrell JE, Jr. , Meyer T . STIM is a Ca2+ sensor essential for Ca2+‐store‐depletion‐triggered Ca2+ influx. Curr Biol 15: 1235‐1241, 2005.
 174. Litosch I . Decoding Galphaq signaling. Life Sci 152: 99‐106, 2016.
 175. Logsdon CD . Stimulation of pancreatic acinar cell growth by CCK, epidermal growth factor, and insulin in vitro. Am J Physiol 251: G487‐G494, 1986.
 176. Logsdon CD , Williams JA . Pancreatic acinar cells in monolayer culture: Direct trophic effects of caerulein in vitro. Am J Physiol 250: G440‐G447, 1986.
 177. Lur G , Haynes LP , Prior IA , Gerasimenko OV , Feske S , Petersen OH , Burgoyne RD , Tepikin AV . Ribosome‐free terminals of rough ER allow formation of STIM1 puncta and segregation of STIM1 from IP(3) receptors. Curr Biol 19: 1648‐1653, 2009.
 178. Lynch G , Kohler S , Leser J , Beil M , Garcia‐Marin LJ , Lutz MP . The tyrosine kinase Yes regulates actin structure and secretion during pancreatic acinar cell damage in rats. Pflugers Arch 447: 445‐451, 2004.
 179. Machado‐de Domenech E , Soling HD . Effects of stimulation of muscarinic and of beta‐catecholamine receptors on the intracellular distribution of protein kinase C in guinea pig exocrine glands. Biochem J 242: 749‐754, 1987.
 180. Malo A , Kruger B , Goke B , Kubisch CH . 4‐Phenylbutyric acid reduces endoplasmic reticulum stress, trypsin activation, and acinar cell apoptosis while increasing secretion in rat pancreatic acini. Pancreas 42: 92‐101, 2013.
 181. Malo A , Kruger B , Seyhun E , Schafer C , Hoffmann RT , Goke B , Kubisch CH . Tauroursodeoxycholic acid reduces endoplasmic reticulum stress, trypsin activation, and acinar cell apoptosis while increasing secretion in rat pancreatic acini. Am J Physiol Gastrointest Liver Physiol 299: G877‐G886, 2010.
 182. Marino CR , Leach SD , Schaefer JF , Miller LJ , Gorelick FS . Characterization of cAMP‐dependent protein kinase activation by CCK in rat pancreas. FEBS Lett 316: 48‐52, 1993.
 183. Matozaki T , Goke B , Tsunoda Y , Rodriguez M , Martinez J , Williams JA . Two functionally distinct cholecystokinin receptors show different modes of action on Ca2+ mobilization and phospholipid hydrolysis in isolated rat pancreatic acini. Studies using a new cholecystokinin analog, JMV‐180. J Biol Chem 265: 6247‐6254, 1990.
 184. Matozaki T , Williams JA . Multiple sources of 1,2‐diacylglycerol in isolated rat pancreatic acini stimulated by cholecystokinin. Involvement of phosphatidylinositol bisphosphate and phosphatidylcholine hydrolysis. J Biol Chem 264: 14729‐14734, 1989.
 185. McCormack JG , Halestrap AP , Denton RM . Role of calcium ions in regulation of mammalian intramitochondrial metabolism. Physiol Rev 70: 391‐425, 1990.
 186. McLaughlin CL , Baile CA , Peikin SR . Hyperphagia during lactation: Satiety response to CCK and growth of the pancreas. Am J Physiol 244: E61‐E65, 1983.
 187. Means AR . Regulatory cascades involving calmodulin‐dependent protein kinases. Mol Endocrinol 14: 4‐13, 2000.
 188. Meloche S , Pouyssegur J . The ERK1/2 mitogen‐activated protein kinase pathway as a master regulator of the G1‐ to S‐phase transition. Oncogene 26: 3227‐3239, 2007.
 189. Merritt JE , Rubin RP . Pancreatic amylase secretion and cytoplasmic free calcium. Effects of ionomycin, phorbol dibutyrate and diacylglycerols alone and in combination. Biochem J 230: 151‐159, 1985.
 190. Mignen O , Thompson JL , Yule DI , Shuttleworth TJ . Agonist activation of arachidonate‐regulated Ca2+‐selective (ARC) channels in murine parotid and pancreatic acinar cells. J Physiol 564: 791‐801, 2005.
 191. Miller LJ , Gao F . Structural basis of cholecystokinin receptor binding and regulation. Pharmacol Ther 119: 83‐95, 2008.
 192. Miller WE , Lefkowitz RJ . Expanding roles for beta‐arrestins as scaffolds and adapters in GPCR signaling and trafficking. Curr Opin Cell Biol 13: 139‐145, 2001.
 193. Minami T. Calcineurin‐NFAT activation and DSCR‐1 auto‐inhibitory loop: How is homoeostasis regulated? J Biochem 155: 217‐226, 2014.
 194. Mishra V , Cline R , Noel P , Karlsson J , Baty CJ , Orlichenko L , Patel K , Trivedi RN , Husain SZ , Acharya C , Durgampudi C , Stolz DB , Navina S , Singh VP . Src dependent pancreatic acinar injury can be initiated independent of an increase in cytosolic calcium. PLoS One 8: e66471, 2013.
 195. Miyasaka K , Shinozaki H , Jimi A , Funakoshi A . Amylase secretion from dispersed human pancreatic acini: Neither cholecystokinin a nor cholecystokinin B receptors mediate amylase secretion in vitro. Pancreas 25: 161‐165, 2002.
 196. Mochly‐Rosen D. Localization of protein kinases by anchoring proteins: A theme in signal transduction. Science 268: 247‐251, 1995.
 197. Morisset J , Aliaga JC , Calvo EL , Bourassa J , Rivard N . Expression and modulation of p42/p44 MAPKs and cell cycle regulatory proteins in rat pancreas regeneration. Am J Physiol Gastrointest Liver Physiol 277: G953‐G959, 1999.
 198. Morisset J , Guan D , Jurkowska G , Rivard N , Green GM . Endogenous cholecystokinin, the major factor responsible for dietary protein‐induced pancreatic growth. Pancreas 7: 522‐529, 1992.
 199. Morisset JA , Webster PD . Effects of fasting and feeding on protein synthesis by the rat pancreas. J Clin Invest 51: 1‐8, 1972.
 200. Muili KA , Jin S , Orabi AI , Eisses JF , Javed TA , Le T , Bottino R , Jayaraman T , Husain SZ . Pancreatic acinar cell nuclear factor kappaB activation because of bile acid exposure is dependent on calcineurin. J Biol Chem 288: 21065‐21073, 2013.
 201. Murali A , Rajalingam K . Small Rho GTPases in the control of cell shape and mobility. Cell Mol Life Sci 71: 1703‐1721, 2014.
 202. Murphy JA , Criddle DN , Sherwood M , Chvanov M , Mukherjee R , McLaughlin E , Booth D , Gerasimenko JV , Raraty MG , Ghaneh P , Neoptolemos JP , Gerasimenko OV , Tepikin AV , Green GM , Reeve JR, Jr. , Petersen OH , Sutton R . Direct activation of cytosolic Ca2+ signaling and enzyme secretion by cholecystokinin in human pancreatic acinar cells. Gastroenterology 135: 632‐641, 2008.
 203. Nakamura Y , Fukami K . Regulation and physiological functions of mammalian phospholipase C. J Biochem 161: 315‐321, 2017.
 204. Nathanson MH , Fallon MB , Padfield PJ , Maranto AR . Localization of the type 3 inositol 1,4,5‐trisphosphate receptor in the Ca2+ wave trigger zone of pancreatic acinar cells. J Biol Chem 269: 4693‐4696, 1994.
 205. Nathanson MH , Padfield PJ , O'Sullivan AJ , Burgstahler AD , Jamieson JD . Mechanism of Ca2+ wave propagation in pancreatic acinar cells. J Biol Chem 267: 18118‐18121, 1992.
 206. Nemoto T , Kojima T , Oshima A , Bito H , Kasai H . Stabilization of exocytosis by dynamic F‐actin coating of zymogen granules in pancreatic acini. J Biol Chem 279: 37544‐37550, 2004.
 207. Newton AC. Regulation of the ABC kinases by phosphorylation: Protein kinase C as a paradigm. Biochem J 370: 361‐371, 2003.
 208. Nicke B , Tseng MJ , Fenrich M , Logsdon CD . Adenovirus‐mediated gene transfer of RasN17 inhibits specific CCK actions on pancreatic acinar cells. Am J Physiol Gastrointest Liver Physiol 276: G499‐G506, 1999.
 209. Niederau C , Liddle RA , Williams JA , Grendell JH . Pancreatic growth: Interaction of exogenous cholecystokinin, a protease inhibitor, and a cholecystokinin receptor antagonist in mice. Gut 28(Suppl): 63‐69, 1987.
 210. Nishizuka Y. Intracellular signaling by hydrolysis of phospholipids and activation of protein kinase C. Science 258: 607‐614, 1992.
 211. Nobes CD , Hall A . Rho, rac, and cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia. Cell 81: 53‐62, 1995.
 212. Noguchi M , Adachi H , Gardner JD , Jensen RT . Calcium‐activated, phospholipid‐dependent protein kinase in pancreatic acinar cells. Am J Physiol Gastrointest Liver Physiol 248: G692‐G701, 1985.
 213. Nozu F , Owyang C , Tsunoda Y . Involvement of phosphoinositide 3‐kinase and its association with pp60src in cholecystokinin‐stimulated pancreatic acinar cells. Eur J Cell Biol 79: 803‐809, 2000.
 214. Nozu F , Tsunoda Y , Ibitayo AI , Bitar KN , Owyang C . Involvement of RhoA and its interaction with protein kinase C and Src in CCK‐stimulated pancreatic acini. Am J Physiol Gastrointest Liver Physiol 276: G915‐G923, 1999.
 215. Nuche‐Berenguer B , Jensen RT . Gastrointestinal hormones/neurotransmitters and growth factors can activate P21 activated kinase 2 in pancreatic acinar cells by novel mechanisms. Biochim Biophys Acta 1853: 2371‐2382, 2015.
 216. Nuche‐Berenguer B , Moreno P , Jensen RT . Elucidation of the roles of the Src kinases in pancreatic acinar cell signaling. J Cell Biochem 116: 22‐36, 2015.
 217. Nuche‐Berenguer B , Ramos‐Alvarez I , Jensen RT . Src kinases play a novel dual role in acute pancreatitis affecting severity but no role in stimulated enzyme secretion. Am J Physiol Gastrointest Liver Physiol 310: G1015‐G1027, 2016.
 218. Osipchuk YV , Wakui M , Yule DI , Gallacher DV , Petersen OH . Cytoplasmic Ca2+ oscillations evoked by receptor stimulation, G‐protein activation, internal application of inositol trisphosphate or Ca2+: Simultaneous microfluorimetry and Ca2+ dependent Cl‐ current recording in single pancreatic acinar cells. EMBO J 9: 697‐704, 1990.
 219. Owyang C , Logsdon CD . New insights into neurohormonal regulation of pancreatic secretion. Gastroenterology 127: 957‐969, 2004.
 220. Pace A , Garcia‐Marin LJ , Tapia JA , Bragado MJ , Jensen RT . Phosphospecific site tyrosine phosphorylation of p125FAK and proline‐rich kinase 2 is differentially regulated by cholecystokinin receptor type A activation in pancreatic acini. J Biol Chem 278: 19008‐19016, 2003.
 221. Pace A , Tapia JA , Garcia‐Marin LJ , Jensen RT . The Src family kinase, Lyn, is activated in pancreatic acinar cells by gastrointestinal hormones/neurotransmitters and growth factors which stimulate its association with numerous other signaling molecules. Biochim Biophys Acta 1763: 356‐365, 2006.
 222. Pandol SJ , Schoeffield MS . 1,2‐Diacylglycerol, protein kinase C, and pancreatic enzyme secretion. J Biol Chem 261: 4438‐4444, 1986.
 223. Park AM , Kudo M , Hagiwara S , Tabuchi M , Watanabe T , Munakata H , Sakurai T . p38MAPK suppresses chronic pancreatitis by regulating HSP27 and BAD expression. Free Radic Biol Med 52: 2284‐2291, 2012.
 224. Parker EM , Zaman MM , Freedman SD . GP2, a GPI‐anchored protein in the apical plasma membrane of the pancreatic acinar cell, co‐immunoprecipitates with src kinases and caveolin. Pancreas 21: 219‐225, 2000.
 225. Patel R , Atherton P , Wackerhage H , Singh J . Signaling proteins associated with diabetic‐induced exocrine pancreatic insufficiency in rats. Ann N Y Acad Sci 1084: 490‐502, 2006.
 226. Peikin SR , Rottman AJ , Batzri S , Gardner JD . Kinetics of amylase release by dispersed acini prepared from guinea pig pancreas. Am J Physiol 235: E743‐E749, 1978.
 227. Peiris H , Raghupathi R , Jessup CF , Zanin MP , Mohanasundaram D , Mackenzie KD , Chataway T , Clarke JN , Brealey J , Coates PT , Pritchard MA , Keating DJ . Increased expression of the glucose‐responsive gene, RCAN1, causes hypoinsulinemia, beta‐cell dysfunction, and diabetes. Endocrinology 153: 5212‐5221, 2012.
 228. Petersen OH , Tepikin AV . Polarized calcium signaling in exocrine gland cells. Annu Rev Physiol 70: 273‐299, 2008.
 229. Pfeiffer CJ , Chernenko GA , Kohli Y , Barrowman JA . Trophic effects of cholecystokinin octapeptide on the pancreas of the Syrian hamster. Can J Physiol Pharmacol 60: 358‐362, 1982.
 230. Piiper A , Elez R , You SJ , Kronenberger B , Loitsch S , Roche S , Zeuzem S . Cholecystokinin stimulates extracellular signal‐regulated kinase through activation of the epidermal growth factor receptor, Yes, and protein kinase C. Signal amplification at the level of Raf by activation of protein kinase Cepsilon. J Biol Chem 278: 7065‐7072, 2003.
 231. Piiper A , Gebhardt R , Kronenberger B , Giannini CD , Elez R , Zeuzem S . Pertussis toxin inhibits cholecystokinin‐ and epidermal growth factor‐induced mitogen‐activated protein kinase activation by disinhibition of the cAMP signaling pathway and inhibition of c‐Raf‐1. Mol Pharmacol 58: 608‐613, 2000.
 232. Piiper A , Stryjek‐Kaminska D , Klengel R , Zeuzem S . CCK, carbachol, and bombesin activate distinct PLC‐beta isoenzymes via Gq/11 in rat pancreatic acinar membranes. Am J Physiol Gastrointest Liver Physiol 272: G135‐G140, 1997.
 233. Pozo‐Guisado E , Campbell DG , Deak M , Alvarez‐Barrientos A , Morrice NA , Alvarez IS , Alessi DR , Martin‐Romero FJ . Phosphorylation of STIM1 at ERK1/2 target sites modulates store‐operated calcium entry. J Cell Sci 123: 3084‐3093, 2010.
 234. Proud CG . Control of the translational machinery by amino acids. Am J Clin Nutr 99: 231S‐236S, 2014.
 235. Raman M , Chen W , Cobb MH . Differential regulation and properties of MAPKs. Oncogene 26: 3100‐3112, 2007.
 236. Ramnath RD , Sun J , Adhikari S , Bhatia M . Effect of mitogen‐activated protein kinases on chemokine synthesis induced by substance P in mouse pancreatic acinar cells. J Cell Mol Med 11: 1326‐1341, 2007.
 237. Ramnath RD , Sun J , Bhatia M . Involvement of SRC family kinases in substance P‐induced chemokine production in mouse pancreatic acinar cells and its significance in acute pancreatitis. J Pharmacol Exp Ther 329: 418‐428, 2009.
 238. Ramos JW. The regulation of extracellular signal‐regulated kinase (ERK) in mammalian cells. Int J Biochem Cell Biol 40: 2707‐2719, 2008.
 239. Rao A , Luo C , Hogan PG . Transcription factors of the NFAT family: Regulation and function. Annu Rev Immunol 15: 707‐747, 1997.
 240. Redondo PC , Lajas AI , Salido GM , Gonzalez A , Rosado JA , Pariente JA . Evidence for secretion‐like coupling involving pp60src in the activation and maintenance of store‐mediated Ca2+ entry in mouse pancreatic acinar cells. Biochem J 370: 255‐263, 2003.
 241. Reeve JR, Jr. , Green GM , Chew P , Eysselein VE , Keire DA . CCK‐58 is the only detectable endocrine form of cholecystokinin in rat. Am J Physiol Gastrointest Liver Physiol 285: G255‐G265, 2003.
 242. Rehfeld JF . Accurate measurement of cholecystokinin in plasma. Clin Chem 44: 991‐1001, 1998.
 243. Rehfeld JF . Cholecystokinin‐from local gut hormone to ubiquitous messenger. Front Endocrinol (Lausanne) 8: 47, 2017.
 244. Rehfeld JF . Four basic characteristics of the gastrin‐cholecystokinin system. Am J Physiol Gastrointest Liver Physiol 240: G255‐G266, 1981.
 245. Rehfeld JF , Sun G , Christensen T , Hillingso JG . The predominant cholecystokinin in human plasma and intestine is cholecystokinin‐33. J Clin Endocrinol Metab 86: 251‐258, 2001.
 246. Renckens BA , van Emst‐de Vries SE , de Pont JJ , Bonting SL . Rat pancreas adenylate cyclase: VII. Effect of extracellular calcium on pancreozymin‐induced cyclic AMP formation. Biochim Biophys Acta 630: 511‐518, 1980.
 247. Rey O , Reeve JR, Jr. , Zhukova E , Sinnett‐Smith J , Rozengurt E . G protein‐coupled receptor‐mediated phosphorylation of the activation loop of protein kinase D: Dependence on plasma membrane translocation and protein kinase Cepsilon. J Biol Chem 279: 34361‐34372, 2004.
 248. Rindler MJ , Xu CF , Gumper I , Smith NN , Neubert TA . Proteomic analysis of pancreatic zymogen granules: Identification of new granule proteins. J Proteome Res 6: 2978‐2992, 2007.
 249. Rodriguez‐Martin E , Boyano‐Adanez MC , Bodega G , Martin M , Hernandez C , Quin Y , Vadillo M , Arilla‐Ferreiro E . Redistribution of protein kinase C isoforms in rat pancreatic acini during lactation and weaning. FEBS Lett 445: 356‐360, 1999.
 250. Rosado JA , Salido GM , Jensen RT , Garcia LJ . Are tyrosine phosphorylation of p125(FAK) and paxillin or the small GTP binding protein, rho, needed for CCK‐stimulated pancreatic amylase secretion? Biochim Biophys Acta 1404: 412‐426, 1998.
 251. Roskoski R, Jr. ERK1/2 MAP kinases: Structure, function, and regulation. Pharmacol Res 66: 105‐143, 2012.
 252. Rossman KL , Der CJ , Sondek J . GEF means go: Turning on RHO GTPases with guanine nucleotide‐exchange factors. Nat Rev Mol Cell Biol 6: 167‐180, 2005.
 253. Rousseau A , Bertolotti A . An evolutionarily conserved pathway controls proteasome homeostasis. Nature 536: 184‐189, 2016.
 254. Rozengurt E. Protein kinase D signaling: Multiple biological functions in health and disease. Physiology (Bethesda) 26: 23‐33, 2011.
 255. Rubin RP , Godfrey PP , Chapman DA , Putney JW, Jr. Secretagogue‐induced formation of inositol phosphates in rat exocrine pancreas. Implications for a messenger role for inositol trisphosphate. Biochem J 219: 655‐659, 1984.
 256. Rubin RP , Hundley TR , Adolf MA . Regulation of diacylglycerol levels in carbachol‐stimulated pancreatic acinar cells: Relationship to the breakdown of phosphatidylcholine and metabolism to phosphatidic acid. Biochim Biophys Acta 1133: 127‐132, 1992.
 257. Rusnak F , Mertz P . Calcineurin: Form and function. Physiol Rev 80: 1483‐1521, 2000.
 258. Sabatini DM , Erdjument‐Bromage H , Lui M , Tempst P , Snyder SH . RAFT1: A mammalian protein that binds to FKBP12 in a rapamycin‐dependent fashion and is homologous to yeast TORs. Cell 78: 35‐43, 1994.
 259. Sabbatini ME . Cyclic nucleotides as mediators of acinar and ductal function. Pancreapedia: Exocrine Pancreas Knowledge Base, DOI: 10.3998/panc.2016.2, 2016.
 260. Sabbatini ME , Bi Y , Ji B , Ernst SA , Williams JA . CCK activates RhoA and Rac1 differentially through Galpha13 and Galphaq in mouse pancreatic acini. Am J Physiol Cell Physiol 298: C592‐C601, 2010.
 261. Sabbatini ME , Chen X , Ernst SA , Williams JA . Rap1 activation plays a regulatory role in pancreatic amylase secretion. J Biol Chem 283: 23884‐23894, 2008.
 262. Sabbatini ME , D'Alecy L , Lentz SI , Tang T , Williams JA . Adenylyl cyclase 6 mediates the action of cyclic AMP‐dependent secretagogues in mouse pancreatic exocrine cells via protein kinase A pathway activation. J Physiol 591: 3693‐3707, 2013.
 263. Sabbatini ME , Williams JA . Cholecystokinin‐mediated RhoGDI phosphorylation via PKCalpha promotes both RhoA and Rac1 signaling. PLoS One 8: e66029, 2013.
 264. Sakamoto C , Goldfine ID , Roach E , Williams JA . Localization of saturable CCK binding sites in rat pancreatic islets by light and electron microscope autoradiography. Diabetes 34: 390‐394, 1985.
 265. Samuel I , Zaheer A , Fisher RA . In vitro evidence for role of ERK, p38, and JNK in exocrine pancreatic cytokine production. J Gastrointest Surg 10: 1376‐1383, 2006.
 266. Sancak Y , Bar‐Peled L , Zoncu R , Markhard AL , Nada S , Sabatini DM . Ragulator‐Rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids. Cell 141: 290‐303, 2010.
 267. Sancho V , Nuche‐Berenguer B , Jensen RT . The Src kinase Yes is activated in pancreatic acinar cells by gastrointestinal hormones/neurotransmitters, but not pancreatic growth factors, which stimulate its association with numerous other signaling molecules. Biochim Biophys Acta 1823: 1285‐1294, 2012.
 268. Sans M , Amin R , Vogel N , D'Alecy L , Kahn R , Williams J . Specific deletion of insulin receptors on pancreatic acinar cells defines the insulin‐acinar axis: Implications for pancreatic insufficiency in diabetes. Gastroenterology 140: S‐156, 2011.
 269. Sans MD , Lee SH , D'Alecy LG , Williams JA . Feeding activates protein synthesis in mouse pancreas at the translational level without increase in mRNA. Am J Physiol Gastrointest Liver Physiol 287: G667‐G675, 2004.
 270. Sans MD , Sabbatini ME , Ernst SA , D'Alecy LG , Nishijima I , Williams JA . Secretin is not necessary for exocrine pancreatic development and growth in mice. Am J Physiol Gastrointest Liver Physiol 301: G791‐G798, 2011.
 271. Sans MD , Tashiro M , Vogel NL , Kimball SR , D'Alecy LG , Williams JA . Leucine activates pancreatic translational machinery in rats and mice through mTOR independently of CCK and insulin. J Nutr 136: 1792‐1799, 2006.
 272. Sans MD , Williams JA . Calcineurin is required for translational control of protein synthesis in rat pancreatic acini. Am J Physiol Cell Physiol 287: C310‐C319, 2004.
 273. Sans MD , Williams JA . The mTOR signaling pathway and regulation of pancreatic function. Pancreapedia 2017.
 274. Sans MD , Xie Q , Williams JA . Regulation of translation elongation and phosphorylation of eEF2 in rat pancreatic acini. Biochem Biophys Res Commun 319: 144‐151, 2004.
 275. Sato N , Suzuki S , Kanai S , Ohta M , Jimi A , Noda T , Takiguchi S , Funakoshi A , Miyasaka K . Different effects of oral administration of synthetic trypsin inhibitor on the pancreas between cholecystokinin‐A receptor gene knockout mice and wild type mice. Jpn J Pharmacol 89: 290‐295, 2002.
 276. Satoh A , Gukovskaya AS , Nieto JM , Cheng JH , Gukovsky I , Reeve JR, Jr. , Shimosegawa T , Pandol SJ . PKC‐delta and ‐epsilon regulate NF‐kappaB activation induced by cholecystokinin and TNF‐alpha in pancreatic acinar cells. Am J Physiol Gastrointest Liver Physiol 287: G582‐G591, 2004.
 277. Satoh K , Narita T , Katsumata‐Kato O , Sugiya H , Seo Y . Involvement of myristoylated alanine‐rich C kinase substrate phosphorylation and translocation in cholecystokinin‐induced amylase release in rat pancreatic acini. Am J Physiol Gastrointest Liver Physiol 310: G399‐G409, 2016.
 278. Saxton RA , Sabatini DM . mTOR signaling in growth, metabolism, and disease. Cell 169: 361‐371, 2017.
 279. Schafer C , Clapp P , Welsh MJ , Benndorf R , Williams JA . HSP27 expression regulates CCK‐induced changes of the actin cytoskeleton in CHO‐CCK‐A cells. Am J Physiol 277: C1032‐C1043, 1999.
 280. Schafer C , Ross SE , Bragado MJ , Groblewski GE , Ernst SA , Williams JA . A role for the p38 mitogen‐activated protein kinase/Hsp 27 pathway in cholecystokinin‐induced changes in the actin cytoskeleton in rat pancreatic acini. J Biol Chem 273: 24173‐24180, 1998.
 281. Schafer C , Williams JA . Stress kinases and heat shock proteins in the pancreas: Possible roles in normal function and disease. J Gastroenterol 35: 1‐9, 2000.
 282. Schmelzle T , Hall MN . TOR, a central controller of cell growth. Cell 103: 253‐262, 2000.
 283. Schwarzendrube J , Niederau M , Luthen R , Niederau C . Effects of cholecystokinin‐receptor blockade on pancreatic and biliary function in healthy volunteers. Gastroenterology 100: 1683‐1690, 1991.
 284. Sebolt‐Leopold JS , Bridges AJ . Road to PD0325901 and beyond: The MEK Inhibitor Quest. In: Kinase Inhibitor Drugs. Hoboken, NJ: John Wiley & Sons, Inc., 2009, pp. 203‐227.
 285. Sengupta S , Peterson TR , Sabatini DM . Regulation of the mTOR complex 1 pathway by nutrients, growth factors, and stress. Mol Cell 40: 310‐322, 2010.
 286. Sewell WA , Young JA . Secretion of electrolytes by the pancreas of the anaestetized rat. J Physiol 252: 379‐396, 1975.
 287. Shin DM , Luo X , Wilkie TM , Miller LJ , Peck AB , Humphreys‐Beher MG , Muallem S . Polarized expression of G protein‐coupled receptors and an all‐or‐none discharge of Ca2+ pools at initiation sites of [Ca2+]i waves in polarized exocrine cells. J Biol Chem 276: 44146‐44156, 2001.
 288. Simeone DM , Zhang L , Graziano K , Nicke B , Pham T , Schaefer C , Logsdon CD . Smad4 mediates activation of mitogen‐activated protein kinases by TGF‐beta in pancreatic acinar cells. Am J Physiol Cell Physiol 281: C311‐C319, 2001.
 289. Sinagoga KL , Stone WJ , Schiesser JV , Schweitzer JI , Sampson L , Zheng Y , Wells JM . Distinct roles for the mTOR pathway in postnatal morphogenesis, maturation and function of pancreatic islets. Development 144: 2402‐2414, 2017.
 290. Singh VP . Src: Regulation and function in the exocrine pancreas. Pancreapedia: Exocrine Pancreas Knowledge Base, DOI: 10.3998/panc.2012.5, 2012.
 291. Singh VP , McNiven MA . Src‐mediated cortactin phosphorylation regulates actin localization and injurious blebbing in acinar cells. Mol Biol Cell 19: 2339‐2347, 2008.
 292. Sjodin L , Gardner JD . Effect of cholecystokinin variant (CCK39) on dispersed acinar cells from guinea pig pancreas. Gastroenterology 73: 1015‐1018, 1977.
 293. Smith GP , Gibbs J . Satiating effect of cholecystokinin. Ann N Y Acad Sci 713: 236‐241, 1994.
 294. Smrcka AV , Sternweis PC . Regulation of purified subtypes of phosphatidylinositol‐specific phospholipase C beta by G protein alpha and beta gamma subunits. J Biol Chem 268: 9667‐9674, 1993.
 295. Solomon TE , Petersen H , Elashoff J , Grossman MI . Interaction of caerulein and secretin on pancreatic size and composition in rat. Am J Physiol 235: E714‐E719, 1978.
 296. Solomon TE , Vanier M , Morisset J . Cell site and time course of DNA synthesis in pancreas after caerulein and secretin. Am J Physiol Gastrointest Liver Physiol 245: G99‐G105, 1983.
 297. Soudah HC , Lu Y , Hasler WL , Owyang C . Cholecystokinin at physiological levels evokes pancreatic enzyme secretion via a cholinergic pathway. Am J Physiol Gastrointest Liver Physiol 263: G102‐G107, 1992.
 298. Stauffer PL , Zhao H , Luby‐Phelps K , Moss RL , Star RA , Muallem S . Gap junction communication modulates [Ca2+]i oscillations and enzyme secretion in pancreatic acini. J Biol Chem 268: 19769‐19775, 1993.
 299. Straub SV , Giovannucci DR , Yule DI . Calcium wave propagation in pancreatic acinar cells: Functional interaction of inositol 1,4,5‐trisphosphate receptors, ryanodine receptors, and mitochondria. J Gen Physiol 116: 547‐560, 2000.
 300. Streb H , Irvine RF , Berridge MJ , Schulz I . Release of Ca2+ from a nonmitochondrial intracellular store in pancreatic acinar cells by inositol‐1,4,5‐trisphosphate. Nature 306: 67‐69, 1983.
 301. Sung CK , Hootman SR , Stuenkel EL , Kuroiwa C , Williams JA . Downregulation of protein kinase C in guinea pig pancreatic acini: Effects on secretion. Am J Physiol Gastrointest Liver Physiol 254: G242‐G248, 1988.
 302. Sung CK , Williams JA . Insulin and ribosomal protein S6 kinase in rat pancreatic acini. Diabetes 38: 544‐549, 1989.
 303. Sung CK , Williams JA . Cholecystokinin stimulates a specific ribosomal S6 kinase in rat pancreatic acini. Pancreas 5: 668‐676, 1990.
 304. Szalmay G , Varga G , Kajiyama F , Yang XS , Lang TF , Case RM , Steward MC . Bicarbonate and fluid secretion evoked by cholecystokinin, bombesin and acetylcholine in isolated guinea‐pig pancreatic ducts. J Physiol 535: 795‐807, 2001.
 305. Takiguchi S , Suzuki S , Sato Y , Kanai S , Miyasaka K , Jimi A , Shinozaki H , Takata Y , Funakoshi A , Kono A , Minowa O , Kobayashi T , Noda T . Role of CCK‐A receptor for pancreatic function in mice: A study in CCK‐A receptor knockout mice. Pancreas 24: 276‐283, 2002.
 306. Tapia JA , Ferris HA , Jensen RT , Garcia LJ . Cholecystokinin activates PYK2/CAKbeta by a phospholipase C‐dependent mechanism and its association with the mitogen‐activated protein kinase signaling pathway in pancreatic acinar cells. J Biol Chem 274: 31261‐31271, 1999.
 307. Tapia JA , Garcia‐Marin LJ , Jensen RT . Cholecystokinin‐stimulated protein kinase C‐delta kinase activation, tyrosine phosphorylation, and translocation are mediated by Src tyrosine kinases in pancreatic acinar cells. J Biol Chem 278: 35220‐35230, 2003.
 308. Tashiro M , Dabrowski A , Guo L , Sans MD , Williams JA . Calcineurin‐dependent and calcineurin‐independent signal transduction pathways activated as part of pancreatic growth. Pancreas 32: 314‐320, 2006.
 309. Tashiro M , Samuelson LC , Liddle RA , Williams JA . Calcineurin mediates pancreatic growth in protease inhibitor‐treated mice. Am J Physiol Gastrointest Liver Physiol 286: G784‐G790, 2004.
 310. Taylor SJ , Chae HZ , Rhee SG , Exton JH . Activation of the beta 1 isozyme of phospholipase C by alpha subunits of the Gq class of G proteins. Nature 350: 516‐518, 1991.
 311. Tcherkezian J , Lamarche‐Vane N . Current knowledge of the large RhoGAP family of proteins. Biol Cell 99: 67‐86, 2007.
 312. Tepikin AV , Voronina SG , Gallacher DV , Petersen OH . Pulsatile Ca2+ extrusion from single pancreatic acinar cells during receptor‐activated cytosolic Ca2+ spiking. J Biol Chem 267: 14073‐14076, 1992.
 313. Thompson JL , Shuttleworth TJ . Molecular basis of activation of the arachidonate‐regulated Ca2+ (ARC) channel, a store‐independent Orai channel, by plasma membrane STIM1. J Physiol 591: 3507‐3523, 2013.
 314. Thorn P , Lawrie AM , Smith PM , Gallacher DV , Petersen OH . Local and global cytosolic Ca2+ oscillations in exocrine cells evoked by agonists and inositol trisphosphate. Cell 74: 661‐668, 1993.
 315. Thorn P , Petersen OH . Calcium oscillations in pancreatic acinar cells, evoked by the cholecystokinin analogue JMV‐180, depend on functional inositol 1,4,5‐trisphosphate receptors. J Biol Chem 268: 23219‐23221, 1993.
 316. Thrower EC , Osgood S , Shugrue CA , Kolodecik TR , Chaudhuri AM , Reeve JR, Jr. , Pandol SJ , Gorelick FS . The novel protein kinase C isoforms ‐delta and ‐epsilon modulate caerulein‐induced zymogen activation in pancreatic acinar cells. Am J Physiol Gastrointest Liver Physiol 294: G1344‐G1353, 2008.
 317. Thrower EC , Wang J , Cheriyan S , Lugea A , Kolodecik TR , Yuan J , Reeve JR, Jr. , Gorelick FS , Pandol SJ . Protein kinase C delta‐mediated processes in cholecystokinin‐8‐stimulated pancreatic acini. Pancreas 38: 930‐935, 2009.
 318. Thrower EC , Yuan J , Usmani A , Liu Y , Jones C , Minervini SN , Alexandre M , Pandol SJ , Guha S . A novel protein kinase D inhibitor attenuates early events of experimental pancreatitis in isolated rat acini. Am J Physiol Gastrointest Liver Physiol 300: G120‐G129, 2011.
 319. Tietz AB , Malo A , Diebold J , Kotlyarov A , Herbst A , Kolligs FT , Brandt‐Nedelev B , Halangk W , Gaestel M , Goke B , Schafer C . Gene deletion of MK2 inhibits TNF‐alpha and IL‐6 and protects against cerulein‐induced pancreatitis. Am J Physiol Gastrointest Liver Physiol 290: G1298‐G1306, 2006.
 320. Toescu EC , Lawrie AM , Petersen OH , Gallacher DV . Spatial and temporal distribution of agonist‐evoked cytoplasmic Ca2+ signals in exocrine acinar cells analysed by digital image microscopy. EMBO J 11: 1623‐1629, 1992.
 321. Torrazza RM , Suryawan A , Gazzaneo MC , Orellana RA , Frank JW , Nguyen HV , Fiorotto ML , El‐Kadi S , Davis TA . Leucine supplementation of a low‐protein meal increases skeletal muscle and visceral tissue protein synthesis in neonatal pigs by stimulating mTOR‐dependent translation initiation. J Nutr 140: 2145‐2152, 2010.
 322. Tsunoda Y , Stuenkel EL , Williams JA . Characterization of sustained [Ca2+]i increase in pancreatic acinar cells and its relation to amylase secretion. Am J Physiol Gastrointest Liver Physiol 259: G792‐G801, 1990.
 323. Tsunoda Y , Stuenkel EL , Williams JA . Oscillatory mode of calcium signaling in rat pancreatic acinar cells. Am J Physiol Cell Physiol 258: C147‐C155, 1990.
 324. Tsunoda Y , Yoshida H , Africa L , Steil GJ , Owyang C . Src kinase pathways in extracellular Ca(2+)‐dependent pancreatic enzyme secretion. Biochem Biophys Res Commun 227: 876‐884, 1996.
 325. Uchida T , Iwashita N , Ohara‐Imaizumi M , Ogihara T , Nagai S , Choi JB , Tamura Y , Tada N , Kawamori R , Nakayama KI , Nagamatsu S , Watada H . Protein kinase Cdelta plays a non‐redundant role in insulin secretion in pancreatic beta cells. J Biol Chem 282: 2707‐2716, 2007.
 326. Unal EB , Uhlitz F , Bluthgen N . A compendium of ERK targets. FEBS Lett 591: 2607‐2615, 2017.
 327. Urano F , Wang X , Bertolotti A , Zhang Y , Chung P , Harding HP , Ron D . Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 287: 664‐666, 2000.
 328. Valentijn JA , Valentijn K , Pastore LM , Jamieson JD . Actin coating of secretory granules during regulated exocytosis correlates with the release of rab3D. Proc Natl Acad Sci U S A 97: 1091‐1095, 2000.
 329. Vander Haar E , Lee SI , Bandhakavi S , Griffin TJ , Kim DH . Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40. Nat Cell Biol 9: 316‐323, 2007.
 330. Vigna SR , Thorndyke MC , Williams JA . Evidence for a common evolutionary origin of brain and pancreas cholecystokinin receptors. Proc Natl Acad Sci U S A 83: 4355‐4359, 1986.
 331. von Kriegsheim A , Baiocchi D , Birtwistle M , Sumpton D , Bienvenut W , Morrice N , Yamada K , Lamond A , Kalna G , Orton R , Gilbert D , Kolch W . Cell fate decisions are specified by the dynamic ERK interactome. Nat Cell Biol 11: 1458‐1464, 2009.
 332. von Manteuffel SR , Dennis PB , Pullen N , Gingras AC , Sonenberg N , Thomas G . The insulin‐induced signalling pathway leading to S6 and initiation factor 4E binding protein 1 phosphorylation bifurcates at a rapamycin‐sensitive point immediately upstream of p70s6k. Mol Cell Biol 17: 5426‐5436, 1997.
 333. Voronina S , Sukhomlin T , Johnson PR , Erdemli G , Petersen OH , Tepikin A . Correlation of NADH and Ca2+ signals in mouse pancreatic acinar cells. J Physiol 539: 41‐52, 2002.
 334. Voronina S , Tepikin A . Mitochondrial calcium in the life and death of exocrine secretory cells. Cell Calcium 52: 86‐92, 2012.
 335. Voronina SG , Barrow SL , Simpson AW , Gerasimenko OV , da Silva Xavier G , Rutter GA , Petersen OH , Tepikin AV . Dynamic changes in cytosolic and mitochondrial ATP levels in pancreatic acinar cells. Gastroenterology 138: 1976‐1987, 2010.
 336. Wang S , Lukinius A , Zhou Y , Stalberg P , Gobl A , Oberg K , Skogseid B . Subcellular distribution of phospholipase C isoforms in rodent pancreas and gastric mucosa. Endocrinology 141: 2589‐2593, 2000.
 337. Wank SA . G protein‐coupled receptors in gastrointestinal physiology. I. CCK receptors: An exemplary family. Am J Physiol Gastrointest Liver Physiol 274: G607‐G613, 1998.
 338. Waschulewski IH , Hall DV , Kern HF , Edwardson JM . Effects of the immunosuppressants cyclosporin A and FK 506 on exocytosis in the rat exocrine pancreas in vitro. Br J Pharmacol 108: 892‐900, 1993.
 339. Watanabe H , Saito H , Rychahou PG , Uchida T , Evers BM . Aging is associated with decreased pancreatic acinar cell regeneration and phosphatidylinositol 3‐kinase/Akt activation. Gastroenterology 128: 1391‐1404, 2005.
 340. Weston CR , Davis RJ . The JNK signal transduction pathway. Curr Opin Genet Dev 12: 14‐21, 2002.
 341. Whitmarsh AJ , Cavanagh J , Tournier C , Yasuda J , Davis RJ . A mammalian scaffold complex that selectively mediates MAP kinase activation. Science 281: 1671‐1674, 1998.
 342. Williams JA . Regulation of normal and adaptive pancreatic growth. Pancreapedia: Exocrine Pancreas Knowledge Base, DOI: 10.3998/panc.2017.2, 2017.
 343. Williams JA . Isolation of rodent pancreatic acinar cells and acini by collagenase digestion. Pancreapedia; Exocrine Pancreas Knowledge Base, DOI: 10.3998/panc.2010.18, 2010.
 344. Williams JA , Chen X , Sabbatini ME . Small G proteins as key regulators of pancreatic digestive enzyme secretion. Am J Physiol Endocrinol Metab 296: E405‐E414, 2009.
 345. Williams JA , Holtz BJ . ERK activation and its role in pancreatic acinar cell function. Pancreapedia: Exocrine Pancreas Knowledge Base, DOI: 10,3998/panc.2017.6, 2017.
 346. Williams JA , Korc M , Dormer RL . Action of secretagogues on a new preparation of functionally intact, isolated pancreatic acini. Am J Physiol 235: 517‐524, 1978.
 347. Williams JA , Sans MD , Tashiro M , Schafer C , Bragado MJ , Dabrowski A . Cholecystokinin activates a variety of intracellular signal transduction mechanisms in rodent pancreatic acinar cells. Pharmacol Toxicol 91: 297‐303, 2002.
 348. Williams JA , Yule DI . Stimulus‐secretion coupling in pancreatic acinar cells In: Physiology of the Gastrointestinal Tract (Fifth Edition), edited by Johnson LR , Ghishan FK , Merchant JL , Said HM , and Wood JD . Boston: Academic Press, 2012, pp. 1361‐1398.
 349. Willoughby EA , Perkins GR , Collins MK , Whitmarsh AJ . The JNK‐interacting protein‐1 scaffold protein targets MAPK phosphatase‐7 to dephosphorylate JNK. J Biol Chem 278: 10731‐10736, 2003.
 350. Wishart MJ , Groblewski G , Goke BJ , Wagner AC , Williams JA . Secretagogue regulation of pancreatic acinar cell protein phosphorylation shown by large‐scale 2D‐PAGE. Am J Physiol Gastrointest Liver Physiol 267: G676‐G686, 1994.
 351. Wooten MW , Wrenn RW . Phorbol ester induces intracellular translocation of phospholipid/Ca2+‐dependent protein kinase and stimulates amylase secretion in isolated pancreatic acini. FEBS Lett 171: 183‐186, 1984.
 352. Wooten MW , Wrenn RW . Redistribution of phospholipid/calcium‐dependent protein kinase and altered phosphorylation of its soluble and particulate substrate proteins in phorbol ester‐treated rat pancreatic acini. Cancer Res 45: 3912‐3917, 1985.
 353. Wu‐Zhang AX , Newton AC . Protein kinase C pharmacology: Refining the toolbox. Biochem J 452: 195‐209, 2013.
 354. Wu H , Peisley A , Graef IA , Crabtree GR . NFAT signaling and the invention of vertebrates. Trends Cell Biol 17: 251‐260, 2007.
 355. Wu L , Cai B , Zheng S , Liu X , Cai H , Li H . Effect of emodin on endoplasmic reticulum stress in rats with severe acute pancreatitis. Inflammation 36: 1020‐1029, 2013.
 356. Wu SV , Yang M , Avedian D , Birnbaumer M , Walsh JH . Single amino acid substitution of serine82 to asparagine in first intracellular loop of human cholecystokinin (CCK)‐B receptor confers full cyclic AMP responses to CCK and gastrin. Mol Pharmacol 55: 795‐803, 1999.
 357. Wu V , Yang M , McRoberts JA , Ren J , Seensalu R , Zeng N , Dagrag M , Birnbaumer M , Walsh JH . First intracellular loop of the human cholecystokinin‐A receptor is essential for cyclic AMP signaling in transfected HEK‐293 cells. J Biol Chem 272: 9037‐9042, 1997.
 358. Yamamoto M , Otani M , Jia DM , Fukumitsu K , Yoshikawa H , Akiyama T , Otsuki M . Differential mechanism and site of action of CCK on the pancreatic secretion and growth in rats. Am J Physiol Gastrointest Liver Physiol 285: G681‐G687, 2003.
 359. Yamanishi R , Kotera J , Fushiki T , Soneda T , Iwanaga T , Sugimoto E . Characteristic and localization of the monitor peptide receptor. Biosci Biotechnol Biochem 57: 1153‐1156, 1993.
 360. Yamasaki M , Thomas JM , Churchill GC , Garnham C , Lewis AM , Cancela JM , Patel S , Galione A . Role of NAADP and cADPR in the induction and maintenance of agonist‐evoked Ca2+ spiking in mouse pancreatic acinar cells. Curr Biol 15: 874‐878, 2005.
 361. Yang J , Waldron RT , Su HY , Moro A , Chang HH , Eibl G , Ferreri K , Kandeel FR , Lugea A , Li L , Pandol SJ . Insulin promotes proliferation and fibrosing responses in activated pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol 311: G675‐G687, 2016.
 362. Yao Z , Seger R . The ERK signaling cascade–views from different subcellular compartments. Biofactors 35: 407‐416, 2009.
 363. Yoon S , Seger R . The extracellular signal‐regulated kinase: Multiple substrates regulate diverse cellular functions. Growth Factors 24: 21‐44, 2006.
 364. You CH , Rominger JM , Chey WY . Potentiation effect of cholecystokinin‐octapeptide on pancreatic bicarbonate secretion stimulated by a physiologic dose of secretin in humans. Gastroenterology 85: 40‐45, 1983.
 365. Yuan HX , Xiong Y , Guan KL . Nutrient sensing, metabolism, and cell growth control. Mol Cell 49: 379‐387, 2013.
 366. Yuan J , Lugea A , Zheng L , Gukovsky I , Edderkaoui M , Rozengurt E , Pandol SJ . Protein kinase D1 mediates NF‐kappaB activation induced by cholecystokinin and cholinergic signaling in pancreatic acinar cells. Am J Physiol Gastrointest Liver Physiol 295: G1190‐G1201, 2008.
 367. Yuan J , Tan T , Geng M , Tan G , Chheda C , Pandol SJ . Novel small molecule inhibitors of protein kinase D suppress NF‐kappaB activation and attenuate the severity of rat cerulein pancreatitis. Front Physiol 8: 1014, 2017.
 368. Yule DI . Ca2+ signaling in pancreatic acinar cells. Pancreapedia: Exocrine Pancreas Knowledge Base, DOI: 10.3998/panc.2015.24, 2015.
 369. Yule DI , Baker CW , Williams JA . Calcium signaling in rat pancreatic acinar cells: A role for Galphaq, Galpha11, and Galpha14. Am J Physiol Gastrointest Liver Physiol 276: G271‐G279, 1999.
 370. Yule DI , Ernst SA , Ohnishi H , Wojcikiewicz RJ . Evidence that zymogen granules are not a physiologically relevant calcium pool. Defining the distribution of inositol 1,4,5‐trisphosphate receptors in pancreatic acinar cells. J Biol Chem 272: 9093‐9098, 1997.
 371. Yule DI , Lawrie AM , Gallacher DV . Acetylcholine and cholecystokinin induce different patterns of oscillating calcium signals in pancreatic acinar cells. Cell Calcium 12: 145‐151, 1991.
 372. Yule DI , Stuenkel E , Williams JA . Intercellular calcium waves in rat pancreatic acini: Mechanism of transmission. Am J Physiol Cell Physiol 271: C1285‐C1294, 1996.
 373. Yule DI , Tseng MJ , Williams JA , Logdson CD . A cloned CCK‐A receptor transduces multiple signals in response to full and partial agonists. Am J Physiol Gastrointest Liver Physiol 265: G999‐G1004, 1993.
 374. Yutsudo Y , Kido Y , Okabayashi Y , Matsumoto M , Ogawa W , Ohba M , Kuroki T , Kasuga M . Protein kinase Calpha is implicated in cholecystokinin‐induced activation of 70‐kd S6 kinase in AR42J cells. Pancreas 30: 50‐53, 2005.
 375. Zarbin MA , Wamsley JK , Innis RB , Kuhar MJ . Cholecystokinin receptors: Presence and axonal flow in the rat vagus nerve. Life Sci 29: 697‐705, 1981.
 376. Zhang X , Wen J , Aletta JM , Rubin RP . Inhibition of expression of PKC‐alpha by antisense mRNA is associated with diminished cell growth and inhibition of amylase secretion by AR4‐2J cells. Exp Cell Res 233: 225‐231, 1997.
 377. Zoncu R , Efeyan A , Sabatini DM . mTOR: From growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol 12: 21‐35, 2011.
 378. Zucker KA , Adrian TE , Bilchik AJ , Modlin IM . Effects of the CCK receptor antagonist L364,718 on pancreatic growth in adult and developing animals. Am J Physiol Gastrointest Liver Physiol 257: G511‐G516, 1989.

 

Teaching Material

J. A. Williams. Cholecystokinin (CCK) Regulation of Pancreatic Acinar Cells: Physiological Actions and Signal Transduction Mechanisms. Compr Physiol 9: 2019, 535-564.

Didactic Synopsis

Major Teaching Points:

  • Cholecystokinin (CCK) is a peptide hormone and paracrine regulator produced in unique intestinal enteroendocrine cells and specific neurons in the brain.
  • The major targets for CCK regulation in the gastrointestinal tract are the pancreatic acinar cell which is stimulated to secrete, gallbladder smooth muscle which contracts and gastric smooth muscle which is relaxed to inhibit gastric emptying.
  • The actions of CCK on pancreatic acinar cells are mediated by CCK type1 receptors, heterotrimeric G proteins and signal transduction pathways.
  • Acinar cell digestive enzyme secretion is mediated by the traditional second messenger pathways centered on Ca2+, cAMP, and diacylglycerol – PKC.
  • Distinct pathways regulating pancreatic adaptive growth and gene expression are mediated by mTOR, the three MAPK pathways centered on ERK, JNK, and p38MAPK as well as the calcineurin – NFAT.
  • Small G proteins Rho and Rac are activated by CCK and affect both secretory and nonsecretory processes.
  • Other signaling proteins that can be activated by CCK include PKD, Src, and FAK.

Didactic Legends

The figures—in a freely downloadable PowerPoint format—can be found on the Images tab along with the formal legends published in the article. The following legends to the same figures are written to be useful for teaching.

Figure 1 Teaching Points. The primary linear amino acid sequence (without most individual amino acids) is shown for the CCK1 Receptor as it is organized through the plasma membrane in two-dimensions.

Figure 2 Teaching Points. This diagram illustrates how the CCK1R can initiate multiple signaling pathways by activating different heterotrimeric G proteins including three different alpha subunits and possibly the beta/gamma subunits.

Figure 3 Teaching Points. A record of changes in intracellular Ca2+ concentrations recorded from a single cell by fura 2 flourescence when a pancreatic acinar cell in vitro is stimulated with a physiological concentration of CCK. Note that the transient increases in Ca2+ termed oscillations are maintained for 10 min in the absence of external calcium.

Figure 4 Teaching Points. This figure shows intracellular Ca2+ concentrations in cells of a pancreatic acini as a digital image with color coding to represent the concentration of Ca2+. At any one point in an acini the calcium concentration shows oscillations as shown in panel A. Panel B shows a rise in calcium concentration in a region of one cell that spreads to an adjacent cell through gap junctions and on around the acini. After a resting basal period a new cycle is initiated.

Figure 5 Teaching Points. Summary diagram of intracellular Ca2+ signaling in the pancreatic acinar cell and the protein and lipid molecules and the intracellular organells that are involved. The diagram can be used to trace the intracellular pathway from receptors to elevation of intracellular free Ca2+ and zymogen granule exocytosis. Details are given in the regular figure legend.

Figure 6 Teaching Points. This diagram illustrates the diacylglycerol – Protein kinase C signaling pathway leading to the activation of specific PKC isoforms.

Figure 7 Teaching Points. This diagram shows the three main mitogen activated protein kinase pathways activated by the CCK receptor. The mechanism for how each pathway is activated is only understood for the ERK pathway as shown in Fig. 8. The bottom of each pathway in Figure 7 shows proteins activated by that particular MAPK.

Figure 8 Teaching Points. This diagram shows mechanisms for CCK and epidermal growth factor (EGF) to activate the ERK pathway in pancreatic acinar cells.

Figure 9 Teaching Points. This diagram shows how CCK and insulin activate the mammalian target of rapamycin complex 1 pathway (mTORC1) in acinar cells. Activation of TORC1 in model cells involves translocation to the lysosomal surface where other proteins can activate it. The bottom of the drawing shows proteins and fucnctions regulated by TORC1 most of which has been shown to occur in acinar cells.

Figure 10 Teaching Points. This diagram shows how the calcineurin – NFAT pathway is activated by CCK and acetylcholine (Ach) in acinar cells. It also shows how Rho regulates the actin cytoskeleton and gene expression.

Figure 11 Teaching Points. This diagram shows how the small G protein Rho is activated by CCK and Ach in acinar cells. It also shows how Rho regulates the actin cytoskeleton and gene expression.

Figure 12 Teaching Points. This diagram shows how receptors for GI hormones regulate Adenylyl cyclase and the formation of cAMP. While the primary activators are secretin and VIP whose receptors activate Gαs, the CCK1R can also activate to a limited degree. Somatostatin inhibits adenylyl cyclase through Gαi. The figure also shows how cAMP activates PKA and EPAC1 and thereby effects exocytosis and chloride (fluid) secretion. Details are given in the regular figure legends.

 


Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

John A. Williams. Cholecystokinin (CCK) Regulation of Pancreatic Acinar Cells: Physiological Actions and Signal Transduction Mechanisms. Compr Physiol 2019, 9: 535-564. doi: 10.1002/cphy.c180014