Revision of Cholecystokinin Type 1 Receptor from Mon, 2017-01-23 14:18

Department of Internal Medicine and the Department of Molecular Pharmacology and Expiermental Therapeutics, Mayo Clinic
miller@mayo.edu

Entry Version: 

Version 1.0, September 18, 2013

Citation: 

Desai Aditya, Miller Laurence. (2013). Cholecystokinin Type 1 Receptor
Pancreapedia: Exocrine Pancreas Knowledge Base, DOI: 10.3998/panc.2013.9
AttachmentSize
PDF icon CCK1R162.23 KB

Gene symbols: CCKAR

1.  General Information

Cholecystokinin (CCK) exerts its physiological actions through the activation of two structurally-related class A G protein-coupled receptors (GPCRs) identified as type 1 CCK receptor (CCK1R) and type 2 CCK receptor (CCK2R) (also known as CCKAR and CCKBR, respectively, related to their prominent presence in “alimentary tract” and “brain”) (14, 47). These receptors have an extracellular amino-terminal tail domain, seven hydrophobic segments representing transmembrane helices that are connected by intracellular and extracellular loops to form a helical bundle domain, and an intracellular carboxyl-terminal tail. These receptors are highly homologous to each other and share 50% overall identity, with the transmembrane segment regions reaching 70% identity (47). The focus of this article is on the type 1 receptor, CCK1R. CCK peptides of different lengths with a common carboxyl-terminal-amide sequence are produced from a single 115-residue preprohormone precursor. Mature peptides range from 58, 39, 33, to 8 residues, with each containing a sulfated tyrosine residue seven residues from the carboxyl terminus (15, 47, 62). The CCK1R requires the carboxyl-terminal CCK heptapeptide-amide that includes a sulfated tyrosine for high affinity binding and full biological potency (47).  This is in contrast to the CCK2R that only requires the carboxyl-terminal tetrapeptide-amide that is shared by CCK and gastrin, and that is not influenced by the sulfation state of the tyrosine residue. 

Figure 1. Shown is a two-dimensional representation of the sequence of the class A GPCR, CCK1R, highlighting some typical features of this receptor. It has typical heptahelical topology, with the amino-terminal tail outside the cell and the carboxyl-terminal tail inside the cell. Sites of glycosylation (Y) exist on ectodomains, while sites of phosphorylation are present intracellularly (grey ‘P’ circles). There are two disulfide bonds (connecting the cysteines) within the amino-terminal tail and linking the top of TM3 with ECL2. There is a site of palmitoylation within the carboxyl-terminal tail (two Cs anchored), helping to establish helix 8 adjacent to the inside of the plasma membrane. Also shown are typical signature sequences of the class A GPCR family (residues represented in green circles). Also illustrated is a proposed docking model for the natural CCK peptide (oval) at the CCK1R, noting key receptor residues thought to be at the interface with this ligand.

The CCK1R belongs to the class A group of GPCRs (47) having signature sequences typical of this family (Fig 1).  This includes E/DRY at the intracellular side of transmembrane segment three and NPxxY at the intracellular side of transmembrane segment seven. The cDNA encoding the CCK1R was first cloned from the rat pancreas by Wank et al. (80), and subsequently from the human gallbladder (77), with the chromosomal localization of this human receptor gene identified soon thereafter (9). The mature receptor is glycosylated and has a conserved disulfide bond between predicted extracellular loops one and two, and an additional intradomain disulfide bond within its amino terminus in the human receptor. This receptor is phosphorylated on serine and threonine residues in intracellular loop three and in the carboxyl-terminal tail in response to agonist stimulation. Relevant kinases that phosphorylate this receptor include protein kinase C and a staurosporine-insensitive G protein-coupled receptor kinase (19). A prominent function of receptor phosphorylation is to interfere with receptor coupling to G proteins, thereby desensitizating the signaling system (61). This receptor also has two cysteine residues representing sites of palmitoylation intracellularly beneath the predicted seventh transmembrane segment, which help to attach an eighth helical segment to the cytosolic face of the bilayer. The carboxyl-terminal tail has been shown to contribute determinants for ligand-induced internalization of the CCK1R (3), although phosphorylation of this region was found to not be required (61).

Agonist stimulation of CCK1R induces a conformational change in the receptor that results in receptor coupling with Gq, which leads to subsequent PLC activation and an increase in intracellular calcium levels from IP3-sensitive stores. Extensive studies have shown that the agonist-occupied Gq-coupled receptor state represents a high affinity state of this receptor (46, 87, 89). The CCK1R has also been shown to be capable of coupling with Gs upon stimulation by high concentrations of CCK, thereby also resulting in increases in cAMP (70, 84, 85, 90).  Mutation of Asn82 in the first intracellular loop has been shown to disrupt this action (84). CCK1R has also been also shown to couple with G13­ that results in activation of a RhoA pathway (44).

A broad range of experimental approaches have been utilized to study the molecular basis of CCK binding to the CCK1R (refer to review (47) for details). While there has been some controversy regarding the interpretation of these studies, the most consistent pose of the bound peptide is along the extracellular surface of the membrane, with its carboxyl-terminal phenylalanine-amide adjacent to the area above transmembrane segment one (27, 49). A contrasting model has been proposed based largely on mutagenesis data in which the carboxyl terminus of the peptide dips into the bilayer within the helical bundle (29), however this interpretation is not compatible with several observations. Most recently, it has been shown that benzodiazepine ligands that occupy the intramembranous pocket proposed to be the location of the carboxyl terminus of CCK in this model are clearly allosteric ligands, able to bind in location that is distinct to that occupied by bound CCK (1, 4, 7, 18). Mutagenesis approaches including segmental deletions and site-specific modifications, as well as chimeric receptor constructs for CCK1R, have been used to provide indirect insights into residues that contribute to ligand binding and signaling (42, 49, 69). Photoaffinity labeling is another approach that has been used, where modified high affinity, biologically active CCK probes with sites of covalent attachment throughout the CCK pharmacophore have provided direct evidence for the spatial approximation between residues within the bound CCK ligand and CCK1R (11-13). Here, too, the carboxyl-terminal residue of CCK was directly shown to be spatially approximated with a residue in the receptor amino terminus, above the top of transmembrane segment one. Additionally, fluorescence-based techniques exploring the microenvironment of receptor-docked CCK fluorescent analogues have also been used. Here a fluorescence indicator was incorporated at different positions of the CCK pharmacophore, and properties such as anisotropy, fluorescence lifetime, iodide quenching, and red-edge excitation shifts for each probe were determined, providing additional insights into the molecular basis of CCK binding to CCK1R  (24-27) (refer to (10) for summary of behavior of various fluorescent probes).

Different non-natural ligands displaying high selectivity for the CCK receptor and potency have been developed (see reviews (30, 33)). A group of benzodiazepine compounds has been most extensively studied in regard to mechanism of binding to CCK1R (1, 4, 7, 18, 23). Studies incorporating receptor mutagenesis, photoaffinity labeling, and pharmacological manipulations have clearly shown that these ligands bind to a distinct allosteric site within the intramembranous helical bundle that is distinct from the orthosteric CCK peptide-binding site of CCK1R (4, 18, 22, 40). It has been shown that for the binding of the CCK1R-selective benzodiazepine-based antagonist, transmembrane segments six and seven (residues 6.51, 6.52, and 7.39 (2)*) are most critical (4). Also, recently an optimal model showing the binding of a benzodiazepine-based CCK1R agonist has been reported that demonstrates a distinct conformation of this binding pocket within the transmembrane helical bundle from that which accommodates the structurally-related antagonist. This study revealed a key role for Leu(7.39) that was predicted to interact with the isopropyl group in the N1 position of the benzodiazepine that acts as a "trigger" for biological activity, whereas the role of this residue is currently less clear for chemically distinct agonists (23).

* According to the Ballesteros and Weinstein GPCR numbering system (2) , amino acid residues predicted to reside within a transmembrane (TM) segment are assigned two numbers (N1,N2), where N1 represents the TM segment number and N2 represents sequential numbering relative to the most conserved residue in and the segment that is assigned 50.

CCK1R is also sensitive to the cholesterol composition of the membrane, in contrast to CCK2R that is not sensitive to this lipid. Membrane cholesterol depletion has been shown to reduce CCK binding affinity to CCK1R, as well as to decrease the biological response to this hormone at that receptor. Increased membrane cholesterol has been shown to be associated with an increase in CCK binding affinity; however, the biological responses to CCK under these conditions have been shown to be lower than normal as well. Some structural determinants for cholesterol sensitivity have been reported to be present within the third exon of CCK1R, which encodes most of transmembrane segment three and segment four, including one CRAC (cholesterol recognition/interaction amino acid consensus) motif and one CCM (cholesterol consensus) motif (10, 20, 21, 28, 59).

CCK elicits a variety of physiological responses via the CCK1R, including a broad variety of important functions, such as stimulation of gallbladder contraction, stimulation of pancreatic exocrine secretion, relaxation of the sphincter of Oddi, inhibition of gastric acid secretion, delay of gastric emptying, and induction of post-cibal satiety (38, 68). The CCK1R is present in various parts of the gastrointestinal tract, such as gallbladder muscularis, neurons controlling pancreatic secretion, D cells in the gastric mucosa (68), muscularis propria of gastric antrum, fundus and pylorus (63), and vagal afferent neurons (refer to review (14) for details).

The CCK1R gene is located on human chromosome 4p15.1-p15.2 and on mouse chromosome 5 (31, 65). Factors regulating levels of expression of this gene have not been extensively studied.  

CCK seems to be involved in pathologic states, such as irritable bowel syndrome, where CCK1R antagonists have been studied as potential treatments (67, 78). It has also been utilized extensively in experimental models of pancreatitis, where CCK hyperstimulation can cause this disorder (64). However, CCK1R antagonists have not been useful in the management of clinical pancreatitis. Reduced responsiveness of the CCK1R has also been shown to contribute to the pathogenesis of gallbladder diseases. Reduced gallbladder muscle contraction in response to CCK has been demonstrated in patients with cholesterol gallstones, as opposed to those with pigment gallstones. This defect in receptor function seems to be caused by the effect of increased membrane cholesterol in this condition (5, 6, 86, 88). A role of CCK1R in development of obesity has been proposed because of its importance in inducing satiety responses (41, 54). Polymorphisms of the CCK1R have also been associated with increased body fat content in some patients (16, 39, 72), however further studies will be necessary to validate a role for these polymorphisms in obesity. Polymorphisms in the CCK1R gene have also been described in some patients with panic disorder, Parkinson’s disease, and alcohol dependence, however this receptor is not believed to play a quantitatively important role in these problems (51, 52, 79). A rare clinical syndrome has been reported in which there is an abnormal trans-acting splicing factor that results in most of the CCK1R being misprocessed, with the third exon spliced out, and thereby yielding a non-functional receptor. This caused a profound reduction in expression levels of the receptor and was associated with obesity and premature gallstones (48).  CCK1R is shown to be present in certain types of cancers; although its role remains unclear. Immunohistochemical analyses have shown the presence of CCK1R in ductular adenocarcinoma cells from some pancreatic tumors (82). CCK1R has also been shown to be heterogeneously expressed in some ileal carcinoids (75).

2.  Specific aspects of CCK receptor function in the pancreas

The most widely recognized physiological role of CCK is stimulation of pancreatic enzyme secretion.  It has become clear, however, that the cellular basis for this may vary among species.  The early and clearly definitive studies focused on the rodent pancreatic acinar cell.  There is convincing evidence for the expression of functional CCK1R on rodent acinar cells, as demonstrated by receptor mRNA expression, CCK ligand binding assays, and in vitro and in vivo functional secretory responses to physiological concentrations of CCK (66, 83).

However, until recently, the expression and cellular distribution of CCK1R in human pancreas was less clear, and has been a subject of considerable debate. This is mainly due to the very low levels of expression of CCK1R mRNA in human pancreas compared with rodent pancreas (32, 81). Immunohistochemical studies have also failed to localize CCK1R to the human pancreas. An early report utilizing reverse-transcriptase PCR showed very low levels of expression of CCK1R mRNA in adult human pancreas, although Northern blotting approaches failed to detect the expression. The same study also reported the expression of CCK1R mRNA in human fetal (mid-trimester) and infant (50 days old) pancreas (57). A more recent study utilizing a quantitative PCR technique on several samples from human pancreas found higher levels of CCK1R mRNA (copy number of 395), but did not establish the specific cell of origin of this signal (17).

A study by Murphy et al. (56) more recently demonstrated the direct activation of isolated human pancreatic acinar cells in response to physiological concentrations of CCK, by measuring oscillatory increases in cytosolic calcium concentrations and subsequent enzyme secretion in vitro. This is the most convincing evidence supporting the hypothesis that pancreatic secretion can be mediated through direct action of CCK on pancreatic acinar cells, as well as its likely stimulation of intrapancreatic nerves (56).

In contrast, there have been many reports demonstrating the indirect action of CCK to stimulate pancreatic enzyme secretion. Studies in human and rodents have showed that cholinergic vagal activation is an important pathway for CCK to stimulate pancreatic enzyme secretion (45). Substantial evidence also supports the presence of this receptor on intrapancreatic neurons and on abdominal branches of the vagus nerve in several species (58). Indeed, CCK1R on vagal afferent fibres has been shown in vivo to mediate pancreatic enzyme secretion (45).

In addition to effects on secretion, CCK can exert trophic and proliferative effects on the pancreas mediated by the CCK1R. The essential contribution of CCK1R for pancreatic regeneration following pancreatectomy or pancreatic duct ligation and the importance of CCK for normal pancreatic growth has also been reported in rats (8, 50, 53, 60). Conversely, studies in mice and guinea pigs deficient in CCK peptide and CCK1R have demonstrated that CCK is not a required growth factor for the murine pancreas (43, 73).

In endocrine pancreas, CCK stimulates the release of insulin (34, 35), and CCK1R has been detected in human insulin- and glucagon-secreting cells (55). In human gallbladder, CCK1R expression has been directly demonstrated on smooth muscle cells, where it is responsible for mediating gallbladder contraction (76).

3.  Tools for study

 a. Molecular constructs

Mouse, rat and human wild type CCK1R cDNA clones can be purchased from GeneCopoeia (www.genecopoeia.com). Human CCK1R cDNA clone in pcDNA3.1+ vector and N-terminal HA tagged CCK1R in pcDNA3.1+ vector are available from UMR cDNA Resource Center, Missouri University of Science and Technology (www.cdna.org). Human CCK1R cDNA in lentiviral vector pReceiver-Lv105 is available from GeneCopoeia. 

b. Antibodies

Polyclonal antibodies raised against peptide epitopes within the amino-terminal and carboxyl-terminal tail regions of the CCK1R are available from many commercial sources, such as Santa Cruz Biotechnology (sc 16172, sc 16173, sc 33220), Pierce Antibodies (Thermo Scientific) (PA3-116, PA5-32692, PA5-32693, PA1-36144, PA1-31121), Novus Biologicals (NBP1-00743, NB100-2805, NB100-60552, NLS3291), Abcam (ab77269, ab28627, ab140762, ab75153, ab140805, ab115287, ab14441), Acris Antibodies (AP01210PU-N, AP02079SU-N, AP02079SU-S, AP02080SU-N, AP02080SU-S, AP02080SU-S, AP16373PU-N, AP16597PU-N, AP20083PU-N, AP20084PU-N, BP2199, EUD3801, SP4663P), LifeSpan BioSciences (LS-A3291, LS-A3293, LS-A820, LS-A822, LS-C120637, LS-C54624, LS-C22102, LS-C177096, LS-C157607, LS-C54623, LS-C151628, LS-C151629, LS-C128134, LS-C128132, LS-C128131,  LS-C89020, LS-C35919), and Merck Millipore (AB9514).

c. Antagonists

Several peptide and non-peptidyl antagonists of the CCK1R have been developed for the treatment of a variety of gastrointestinal disorders. These compounds have been successfully used for in vitro and in vivo studies. The CCK1R antagonists that are commercially available include the following: devazepide (L-364,718) (pIC50 9.7, Tocris Bioscience, cat no. 2304; Sigma-Aldrich, cat no. D3821; Santa Cruz Biotechnology. cat no. sc-203562), SR 27897 (pIC50 8.3, Tocris Bioscience, cat no. 2190), lorglumide (pIC50 6.7-8.2, Sigma Aldrich, cat no. L109), loxiglumide (pIC50 6.5, Sigma Aldrich, cat no. SML0130), and CR 1409 (pIC50 7.86, Phoenix Pharmaceuticals, cat no. 069-08).

d. Transgenic mice

A specific strain of rat known as OLETF rats (Otsuka Long Evans Tokushima Fatty) which naturally lacks CCK1R expression was characterized at the Tokushima Research Institute (Otsuka Pharmaceutical, Tokushima, Japan) (36, 37, 74). A CCK1R-/- mouse lacking exon 3 which encodes for a portion of the third transmembrane segment and the second intracellular loop including the “ERY” motif was developed by Kopin et al. (41). This animal has been shown to exhibit reduced inhibition of food intake, but normal body weight. Suzuki et al. have generated a CCK1R-/- mouse lacking exon 2 (71), which exhibits decreased biliary and pancreatic secretion.

Acknowledgements: This work was supported by a grant from the National Institutes of Health (DK032878) and by the Mayo Clinic. 

4.  Refrences

  1. Aquino CJ, Armour DR, Berman JM, Birkemo LS, Carr RA, Croom DK, Dezube M, Dougherty RW, Jr., Ervin GN, Grizzle MK, Head JE, Hirst GC, James MK, Johnson MF, Miller LJ, Queen KL, Rimele TJ, Smith DN, and Sugg EE. Discovery of 1,5-benzodiazepines with peripheral cholecystokinin (CCK-A) receptor agonist activity. 1. Optimization of the agonist "trigger". J Med Chem 39:562-569, 1996. PMID: 8558528
  2. Ballesteros JA, and Weinstein H. Analysis and refinement of criteria for predicting the structure and relative orientations of transmembranal helical domains. Biophys J 62:107-109, 1992. PMID: 1600090
  3. Cawston EE, Harikumar KG, and Miller LJ. Ligand-induced internalization of the type 1 cholecystokinin receptor independent of recognized signaling activity. Am J Physiol Cell Physiol 302:C615-627, 2012. PMID: 22049215
  4. Cawston EE, Lam PC, Harikumar KG, Dong M, Ball AM, Augustine ML, Akgun E, Portoghese PS, Orry A, Abagyan R, Sexton PM, and 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. PMID: 22467877
  5. Chen Q, Amaral J, Biancani P, and Behar J. Excess membrane cholesterol alters human gallbladder muscle contractility and membrane fluidity. Gastroenterology 116:678-685, 1999. PMID: 10029627
  6. Chen Q, Amaral J, Oh S, Biancani P, and Behar J. Gallbladder relaxation in patients with pigment and cholesterol stones. Gastroenterology 113:930-937, 1997. PMID: 9287986
  7. Darrow JW, Hadac EM, Miller LJ, and Sugg EE. Structurally similar small molecule photoaffinity CCK-A agonists and antagonists as novel tools for directly probing 7TM receptors-ligand interactions. Bioorg Med Chem Lett 8:3127-3132, 1998. PMID: 9873689
  8. De la Mano AM, Sevillano S, Manso MA, and de Dios I. Effect of long-term CCK blockade on the pancreatic acinar cell renewal in rats with acute pancreatitis. Peptides 24:535-541, 2003. PMID: 12860197
  9. De Weerth A, Pisegna JR, Huppi K, and Wank SA. Molecular cloning, functional expression and chromosomal localization of the human cholecystokinin type A receptor. Biochem Biophys Res Commun 194:811-818, 1993. PMID: 8343165
  10. Desai AJ, and Miller LJ. Sensitivity of cholecystokinin receptors to membrane cholesterol content. Front Endocrinol (Lausanne) 3:123, 2012. PMID: 23087674
  11.  Ding XQ, Dolu V, Hadac EM, Holicky EL, Pinon DI, Lybrand TP, and Miller LJ. Refinement of the structure of the ligand-occupied cholecystokinin receptor using a photolabile amino-terminal probe. J Biol Chem 276:4236-4244, 2001. PMID: 11050076
  12. Dong M, Ding XQ, Thomas SE, Gao F, Lam PC, Abagyan R, and Miller LJ. Role of lysine187 within the second extracellular loop of the type A cholecystokinin receptor in agonist-induced activation. Use of complementary charge-reversal mutagenesis to define a functionally important interdomain interaction. Biochemistry 46:4522-4531, 2007. PMID: 17381074
  13. Dong M, Liu G, Pinon DI, and Miller LJ. Differential docking of high-affinity peptide ligands to type A and B cholecystokinin receptors demonstrated by photoaffinity labeling. Biochemistry 44:6693-6700, 2005. PMID: 15850403
  14. Dufresne M, Seva C, and Fourmy D. Cholecystokinin and gastrin receptors. Physiol Rev 86:805-847, 2006. PMID: 16816139
  15. Eysselein VE, Eberlein GA, Hesse WH, Schaeffer M, Grandt D, Williams R, Goebell H, and Reeve JR, Jr. Molecular variants of cholecystokinin after endogenous stimulation in humans: a time study. Am J Physiol 258:G951-957, 1990. PMID: 2360639
  16. Funakoshi A, Miyasaka K, Matsumoto H, Yamamori S, Takiguchi S, Kataoka K, Takata Y, Matsusue K, Kono A, and Shimokata H. Gene structure of human cholecystokinin (CCK) type-A receptor: body fat content is related to CCK type-A receptor gene promoter polymorphism. FEBS Lett 466:264-266, 2000. PMID: 10682840
  17. Galindo J, Jones N, Powell GL, Hollingsworth SJ, and Shankley N. Advanced qRT-PCR technology allows detection of the cholecystokinin 1 receptor (CCK1R) expression in human pancreas. Pancreas 31:325-331, 2005. PMID: 16258365
  18. Gao F, Sexton PM, Christopoulos A, and Miller LJ. Benzodiazepine ligands can act as allosteric modulators of the Type 1 cholecystokinin receptor. Bioorg Med Chem Lett 18:4401-4404, 2008. PMID: 18621527
  19. Gates LK, Ulrich CD, and Miller LJ. Multiple kinases phosphorylate the pancreatic cholecystokinin receptor in an agonist-dependent manner. Am J Physiol 264:G840-847, 1993. PMID: 8498511
  20. Gimpl G, Burger K, and Fahrenholz F. Cholesterol as modulator of receptor function. Biochemistry 36:10959-10974, 1997. PMID: 9283088
  21. Gimpl G, Wiegand V, Burger K, and Fahrenholz F. Cholesterol and steroid hormones: modulators of oxytocin receptor function. Prog Brain Res 139:43-55, 2002. PMID: 12436925
  22. Hadac EM, Dawson ES, Darrow JW, Sugg EE, Lybrand TP, and Miller LJ. Novel benzodiazepine photoaffinity probe stereoselectively labels a site deep within the membrane-spanning domain of the cholecystokinin receptor. J Med Chem 49:850-863, 2006. PMID: 16451051
  23. Harikumar KG, Cawston EE, Lam PC, Patil A, Orry A, Henke BR, Abagyan R, Christopoulos A, Sexton PM, and Miller LJ. Molecular basis for benzodiazepine agonist action at the type 1 cholecystokinin receptor. J Biol Chem 288:21082-21095, 2013. PMID: 23754289
  24. Harikumar KG, Clain J, Pinon DI, Dong M, and Miller LJ. Distinct molecular mechanisms for agonist peptide binding to types A and B cholecystokinin receptors demonstrated using fluorescence spectroscopy. J Biol Chem 280:1044-1050, 2005. PMID: 15520004
  25. Harikumar KG, and Miller LJ. Fluorescence resonance energy transfer analysis of the antagonist- and partial agonist-occupied states of the cholecystokinin receptor. J Biol Chem 280:18631-18635, 2005. PMID: 15757907
  26. Harikumar KG, Pinon DI, and Miller LJ. Fluorescent indicators distributed throughout the pharmacophore of cholecystokinin provide insights into distinct modes of binding and activation of type A and B cholecystokinin receptors. J Biol Chem 281:27072-27080, 2006. PMID: 16857665
  27. Harikumar KG, Pinon DI, Wessels WS, Dawson ES, Lybrand TP, Prendergast FG, and Miller LJ. Measurement of intermolecular distances for the natural agonist Peptide docked at the cholecystokinin receptor expressed in situ using fluorescence resonance energy transfer. Mol Pharmacol 65:28-35, 2004. PMID: 14722234
  28. Harikumar KG, Puri V, Singh RD, Hanada K, Pagano RE, and Miller LJ. Differential effects of modification of membrane cholesterol and sphingolipids on the conformation, function, and trafficking of the G protein-coupled cholecystokinin receptor. J Biol Chem 280:2176-2185, 2005. PMID: 15537636
  29. Henin J, Maigret B, Tarek M, Escrieut C, Fourmy D, and 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. PMID: 16326901
  30. Herranz R. Cholecystokinin antagonists: pharmacological and therapeutic potential. Med Res Rev 23:559-605, 2003. PMID: 12789687
  31. Huppi K, Siwarski D, Pisegna JR, and Wank S. Chromosomal localization of the gastric and brain receptors for cholecystokinin (CCKAR and CCKBR) in human and mouse. Genomics 25:727-729, 1995. PMID: 7759110
  32. Ji B, Bi Y, Simeone D, Mortensen RM, and Logsdon CD. Human pancreatic acinar cells lack functional responses to cholecystokinin and gastrin. Gastroenterology 121:1380-1390, 2001. PMID: 11729117
  33. Kalindjian SB, and McDonald IM. Strategies for the design of non-peptide CCK2 receptor agonist and antagonist ligand. Curr Top Med Chem 7:1195-1204, 2007. PMID: 17584141
  34. Karlsson S, and Ahren B. CCKA receptor antagonism inhibits mechanisms underlying CCK-8-stimulated insulin release in isolated rat islets. Eur J Pharmacol 202:253-257, 1991. PMID: 1666367
  35. Karlsson S, and Ahren B. Effects of three different cholecystokinin receptor antagonists on basal and stimulated insulin and glucagon secretion in mice. Acta Physiol Scand 135:271-278, 1989. PMID: 2648765
  36. Kawano K, Hirashima T, Mori S, Saitoh Y, Kurosumi M, and Natori T. New inbred strain of Long-Evans Tokushima lean rats with IDDM without lymphopenia. Diabetes 40:1375-1381, 1991. PMID: 1682194
  37. Kawano K, Hirashima T, Mori S, Saitoh Y, Kurosumi M, and Natori T. Spontaneous long-term hyperglycemic rat with diabetic complications. Otsuka Long-Evans Tokushima Fatty (OLETF) strain. Diabetes 41:1422-1428, 1992. PMID: 1397718
  38. Kerstens PJ, Lamers CB, Jansen JB, de Jong AJ, Hessels M, and Hafkenscheid JC. Physiological plasma concentrations of cholecystokinin stimulate pancreatic enzyme secretion and gallbladder contraction in man. Life Sci 36:565-569, 1985. PMID: 3968978
  39. Koda M, Ando F, Niino N, Shimokata H, Miyasaka K, and Funakoshi A. Association of cholecystokinin 1 receptor and beta3-adrenergic receptor polymorphisms with midlife weight gain. Obes Res 12:1212-1216, 2004. PMID: 15340101
  40. Kopin AS, Beinborn M, Lee YM, McBride EW, and Quinn SM. The CCK-B/gastrin receptor. Identification of amino acids that determine nonpeptide antagonist affinity. Ann N Y Acad Sci 713:67-78, 1994. PMID: 8185216
  41. Kopin AS, Mathes WF, McBride EW, Nguyen M, Al-Haider W, Schmitz F, Bonner-Weir S, Kanarek R, and Beinborn M. The cholecystokinin-A receptor mediates inhibition of food intake yet is not essential for the maintenance of body weight. J Clin Invest 103:383-391, 1999. PMID: 9927499
  42. Kopin AS, McBride EW, Quinn SM, Kolakowski LF, Jr., and Beinborn M. The role of the cholecystokinin-B/gastrin receptor transmembrane domains in determining affinity for subtype-selective ligands. J Biol Chem 270:5019-5023, 1995. PMID: 7890609
  43. Lacourse KA, Swanberg LJ, Gillespie PJ, Rehfeld JF, Saunders TL, and Samuelson LC. Pancreatic function in CCK-deficient mice: adaptation to dietary protein does not require CCK. Am J Physiol 276:G1302-1309, 1999. PMID: 10330022
  44. Le Page SL, Bi Y, and Williams JA. CCK-A receptor activates RhoA through G alpha 12/13 in NIH3T3 cells. Am J Physiol Cell Physiol 285:C1197-1206, 2003. PMID: 12853286
  45. Li Y, Hao Y, and Owyang C. High-affinity CCK-A receptors on the vagus nerve mediate CCK-stimulated pancreatic secretion in rats. Am J Physiol 273:G679-685, 1997. PMID: 9316472
  46. Matozaki T, Goke B, Tsunoda Y, Rodriguez M, Martinez J, and 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. PMID: 1690723
  47. Miller LJ, and Gao F. Structural basis of cholecystokinin receptor binding and regulation. Pharmacol Ther 119:83-95, 2008. PMID: 18558433
  48. Miller LJ, Holicky EL, Ulrich CD, and Wieben ED. Abnormal processing of the human cholecystokinin receptor gene in association with gallstones and obesity. Gastroenterology 109:1375-1380, 1995. PMID: 7557108
  49. Miller LJ, and Lybrand TP. Molecular basis of agonist binding to the type A cholecystokinin receptor. Pharmacol Toxicol 91:282-285, 2002. PMID: 12688369
  50. Miyasaka K, Ohta M, Tateishi K, Jimi A, and Funakoshi A. Role of cholecystokinin-A (CCK-A) receptor in pancreatic regeneration after pancreatic duct occlusion: a study in rats lacking CCK-A receptor gene expression. Pancreas 16:114-123, 1998. PMID: 9510132
  51. Miyasaka K, Yoshida Y, Matsushita S, Higuchi S, Maruyama K, Niino N, Ando F, Shimokata H, Ohta S, and Funakoshi A. Association of cholecystokinin-A receptor gene polymorphism with alcohol dependence in a Japanese population. Alcohol Alcohol 39:25-28, 2004. PMID: 14691070
  52. Miyasaka K, Yoshida Y, Matsushita S, Higuchi S, Shirakawa O, Shimokata H, and Funakoshi A. Association of cholecystokinin-A receptor gene polymorphisms and panic disorder in Japanese. Am J Med Genet B Neuropsychiatr Genet 127B:78-80, 2004. PMID: 15108185
  53. Moralejo DH, Ogino T, Kose H, Yamada T, and Matsumoto K. Genetic verification of the role of CCK-AR in pancreatic proliferation and blood glucose and insulin regulation using a congenic rat carrying CCK-AR null allele. Res Commun Mol Pathol Pharmacol 109:259-274, 2001. PMID: 12889510
  54. Moran TH, Katz LF, Plata-Salaman CR, and Schwartz GJ. Disordered food intake and obesity in rats lacking cholecystokinin A receptors. Am J Physiol 274:R618-625, 1998. PMID: 9530226
  55. Morisset J, Julien S, and Laine J. Localization of cholecystokinin receptor subtypes in the endocine pancreas. J Histochem Cytochem 51:1501-1513, 2003. PMID: 14566022
  56. 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, and Sutton R. Direct activation of cytosolic Ca2+ signaling and enzyme secretion by cholecystokinin in human pancreatic acinar cells. Gastroenterology 135:632-641, 2008. PMID: 18555802
  57. Nishimori I, Kamakura M, Fujikawa-Adachi K, Nojima M, Onishi S, Hollingsworth MA, and Harris A. Cholecystokinin A and B receptor mRNA expression in human pancreas. Pancreas 19:109-113, 1999. PMID: 10438155
  58. Owyang C, and Logsdon CD. New insights into neurohormonal regulation of pancreatic secretion. Gastroenterology 127:957-969, 2004. PMID: 15362050
  59. Potter RM, Harikumar KG, Wu SV, and Miller LJ. Differential sensitivity of types 1 and 2 cholecystokinin receptors to membrane cholesterol. J Lipid Res 53:137-148, 2012. PMID: 22021636
  60. Povoski SP, Zhou W, Longnecker DS, Jensen RT, Mantey SA, and Bell RH, Jr. Stimulation of in vivo pancreatic growth in the rat is mediated specifically by way of cholecystokinin-A receptors. Gastroenterology 107:1135-1146, 1994. PMID: 7523219
  61. Rao RV, Roettger BF, Hadac EM, and Miller LJ. Roles of cholecystokinin receptor phosphorylation in agonist-stimulated desensitization of pancreatic acinar cells and receptor-bearing Chinese hamster ovary cholecystokinin receptor cells. Mol Pharmacol 51:185-192, 1997. PMID: 9203622
  62. Rehfeld JF, Sun G, Christensen T, and Hillingso JG. The predominant cholecystokinin in human plasma and intestine is cholecystokinin-33. J Clin Endocrinol Metab 86:251-258, 2001. PMID: 11232009
  63. Reubi JC, Waser B, Laderach U, Stettler C, Friess H, Halter F, and Schmassmann A. Localization of cholecystokinin A and cholecystokinin B-gastrin receptors in the human stomach. Gastroenterology 112:1197-1205, 1997. PMID: 9098003
  64. Saluja AK, Saluja M, Printz H, Zavertnik A, Sengupta A, and Steer ML. Experimental pancreatitis is mediated by low-affinity cholecystokinin receptors that inhibit digestive enzyme secretion. Proc Natl Acad Sci U S A 86:8968-8971, 1989. PMID: 2479032
  65. Samuelson LC, Isakoff MS, and Lacourse KA. Localization of the murine cholecystokinin A and B receptor genes. Mamm Genome 6:242-246, 1995. PMID: 7613026
  66. Sankaran H, Goldfine ID, Deveney CW, Wong KY, and Williams JA. Binding of cholecystokinin to high affinity receptors on isolated rat pancreatic acini. J Biol Chem 255:1849-1853, 1980. PMID: 6243650
  67. Scarpignato C, and Pelosini I. Management of irritable bowel syndrome: novel approaches to the pharmacology of gut motility. Can J Gastroenterol 13 Suppl A:50A-65A, 1999. PMID: 10202210
  68. Schmitz F, Goke MN, Otte JM, Schrader H, Reimann B, Kruse ML, Siegel EG, Peters J, Herzig KH, Folsch UR, and Schmidt WE. Cellular expression of CCK-A and CCK-B/gastrin receptors in human gastric mucosa. Regul Pept 102:101-110, 2001. PMID: 11730982
  69. Silvente-Poirot S, Escrieut C, and Wank SA. Role of the extracellular domains of the cholecystokinin receptor in agonist binding. Mol Pharmacol 54:364-371, 1998. PMID: 9687578
  70. Sjodin L, and Gardner JD. Effect of cholecystokinin variant (CCK39) on dispersed acinar cells from guinea pig pancreas. Gastroenterology 73:1015-1018, 1977. PMID: 198332
  71. Suzuki S, Takiguchi S, Sato N, Kanai S, Kawanami T, Yoshida Y, Miyasaka K, Takata Y, Funakoshi A, and Noda T. Importance of CCK-A receptor for gallbladder contraction and pancreatic secretion: a study in CCK-A receptor knockout mice. Jpn J Physiol 51:585-590, 2001. PMID: 11734079
  72. Takata Y, Takeda S, Kawanami T, Takiguchi S, Yoshida Y, Miyasaka K, and Funakoshi A. Promoter analysis of human cholecystokinin type-A receptor gene. J Gastroenterol 37:815-820, 2002. PMID: 12424565
  73. Takiguchi S, Suzuki S, Sato Y, Kanai S, Miyasaka K, Jimi A, Shinozaki H, Takata Y, Funakoshi A, Kono A, Minowa O, Kobayashi T, and 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. PMID: 11893936
  74. Takiguchi S, Takata Y, Funakoshi A, Miyasaka K, Kataoka K, Fujimura Y, Goto T, and Kono A. Disrupted cholecystokinin type-A receptor (CCKAR) gene in OLETF rats. Gene 197:169-175, 1997. PMID: 9332364
  75. Tang C, Biemond I, and Lamers CB. Expression of peptide receptors in human endocrine tumours of the pancreas. Gut 40:267-271, 1997. PMID: 9071943
  76. Tokunaga Y, Cox KL, Coleman R, Concepcion W, Nakazato P, and Esquivel CO. Characterization of cholecystokinin receptors on the human gallbladder. Surgery 113:155-162, 1993. PMID: 7679224
  77. Ulrich CD, Ferber I, Holicky E, Hadac E, Buell G, and Miller LJ. Molecular cloning and functional expression of the human gallbladder cholecystokinin A receptor. Biochem Biophys Res Commun 193:204-211, 1993. PMID: 8503909
  78. Ursini A, Capelli AM, Carr RA, Cassara P, Corsi M, Curcuruto O, Curotto G, Dal Cin M, Davalli S, Donati D, Feriani A, Finch H, Finizia G, Gaviraghi G, Marien M, Pentassuglia G, Polinelli S, Ratti E, Reggiani AM, Tarzia G, Tedesco G, Tranquillini ME, Trist DG, and Van Amsterdam FT. Synthesis and SAR of new 5-phenyl-3-ureido-1,5-benzodiazepines as cholecystokinin-B receptor antagonists. J Med Chem 43:3596-3613, 2000. PMID: 11020274
  79. Wang J, Si YM, Liu ZL, and Yu L. Cholecystokinin, cholecystokinin-A receptor and cholecystokinin-B receptor gene polymorphisms in Parkinson's disease. Pharmacogenetics 13:365-369, 2003. PMID: 12777967
  80. Wank SA, Harkins R, Jensen RT, Shapira H, de Weerth A, and Slattery T. Purification, molecular cloning, and functional expression of the cholecystokinin receptor from rat pancreas. Proc Natl Acad Sci U S A 89:3125-3129, 1992. PMID: 1313582
  81. Wank SA, Pisegna JR, and de Weerth A. Cholecystokinin receptor family. Molecular cloning, structure, and functional expression in rat, guinea pig, and human. Ann N Y Acad Sci 713:49-66, 1994. PMID: 8185215
  82. Weinberg DS, Ruggeri B, Barber MT, Biswas S, Miknyocki S, and Waldman SA. Cholecystokinin A and B receptors are differentially expressed in normal pancreas and pancreatic adenocarcinoma. J Clin Invest 100:597-603, 1997. PMID: 9239407
  83. Williams JA, Korc M, and Dormer RL. Action of secretagogues on a new preparation of functionally intact, isolated pancreatic acini. Am J Physiol 235:517-524, 1978. PMID: 215042
  84. Wu SV, Yang M, Avedian D, Birnbaumer M, and 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. PMID: 10220557
  85. Wu V, Yang M, McRoberts JA, Ren J, Seensalu R, Zeng N, Dagrag M, Birnbaumer M, and 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. PMID: 9083028
  86. Xiao ZL, Chen Q, Amaral J, Biancani P, Jensen RT, and Behar J. CCK receptor dysfunction in muscle membranes from human gallbladders with cholesterol stones. Am J Physiol 276:G1401-1407, 1999. PMID: 10362643
  87. Xu X, Croy JT, Zeng W, Zhao L, Davignon I, Popov S, Yu K, Jiang H, Offermanns S, Muallem S, and Wilkie TM. Promiscuous coupling of receptors to Gq class alpha subunits and effector proteins in pancreatic and submandibular gland cells. J Biol Chem 273:27275-27279, 1998. PMID: 9765251
  88. Yu P, Chen Q, Harnett KM, Amaral J, Biancani P, and Behar J. Direct G protein activation reverses impaired CCK signaling in human gallbladders with cholesterol stones. Am J Physiol 269:G659-665, 1995. PMID: 7491956
  89. Yule DI, Baker CW, and Williams JA. Calcium signaling in rat pancreatic acinar cells: a role for Galphaq, Galpha11, and Galpha14. Am J Physiol 276:G271-279, 1999. PMID: 9887004
  90. Yule DI, Tseng MJ, Williams JA, and Logdson CD. A cloned CCK-A receptor transduces multiple signals in response to full and partial agonists. Am J Physiol 265:G999-1004, 1993. PMID: 8238528