Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Gen Comp Endocrinol ; 209: 74-81, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-24842717

ABSTRACT

Cholecystokinin (CCK) and gastrin are regulatory peptides in vertebrates. Their homologues are widely present in metazoan animals, in form of cionin in tunicates, neuropeptide-like protein 12 in nematodes and sulfakinin (SK) in arthropods. CCK(-like) peptides exert diverse physiological effects through binding their corresponding receptors, which are important members of the hormone-binding G-protein-coupled receptors. In this paper, CCK(-like) peptides and receptors are reviewed in a comparative way at levels of molecular structure, physiological functions and phylogeny. CCK signalling system is widely involved in the regulation of satiety, gastric acid secretion, pancreatic secretion, anxiety and memory processes in vertebrates. Its counterpart SK in arthropods is also found with similar functions on regulation of satiety and gastrointestinal motility. Co-evolution of peptide and receptor has been recognized through metazoans. The CCK(-like) receptors seem to be evolved from a common ancestor based on the phylogenetic analysis, with species-specific events in arthropods. In addition, tetraploidization has been brought up to study the evolution of receptors. There are 2 receptors in chordates and nematodes, whereas, the number of sulfakinin receptor varies in arthropods from 0 to 2. We discussed here that the presence or absence of the SK signalling system is likely to be related to feeding behaviour.


Subject(s)
Cholecystokinin/chemistry , Cholecystokinin/classification , Invertebrates/genetics , Receptors, Cholecystokinin/chemistry , Receptors, Cholecystokinin/classification , Vertebrates/genetics , Amino Acid Sequence , Animals , Biological Evolution , Cholecystokinin/genetics , Humans , Molecular Sequence Data , Phylogeny , Protein Binding/genetics , Protein Conformation , Receptors, Cholecystokinin/genetics
2.
Gen Comp Endocrinol ; 186: 1-8, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23467070

ABSTRACT

In vertebrates, the peptide cholecystokinin (CCK) is one of the most important neuroregulatory digestive hormones. CCK acts via CCK receptors that are classified into two subtypes, CCK-1 receptor (CCK-1R; formally CCK-A) and CCK-2 receptor (formally CCK-B). In particular, the CCK-1R is involved in digestion and is regulated by CCK. However, very little information is known about CCK-1R in fish. Therefore, we performed molecular cloning of CCK-1R cDNA from the digestive tract of yellowtail Seriola quinqueradiata. Phylogenetic tree analysis showed a high sequence identity between the cloned yellowtail CCK receptor cDNA and CCK-1R, which belongs to the CCK-1R cluster. Furthermore, the expression of yellowtail CCK receptor mRNA was observed in gallbladder, pyloric caeca, and intestines, similarly to CCK-1R mRNA expression in mammals, suggesting that the cloned cDNA is of CCK-1R from yellowtail. In in vivo experiments, the CCK-1R mRNA levels increased in the gallbladder and pyloric caeca after feeding, whereas in vitro, mRNA levels of CCK-1R and digestive enzymes in cultured pyloric caeca increased by the addition of CCK. These results suggest that CCK-1R plays an important role in digestion stimulated by CCK in yellowtail.


Subject(s)
Cholecystokinin/pharmacology , Perciformes/metabolism , Receptors, Cholecystokinin/metabolism , Animals , Cloning, Molecular , Gallbladder/drug effects , Gallbladder/metabolism , Intestinal Mucosa/metabolism , Intestines/drug effects , Perciformes/genetics , Phylogeny , Receptors, Cholecystokinin/chemistry , Receptors, Cholecystokinin/classification , Receptors, Cholecystokinin/genetics , Reverse Transcriptase Polymerase Chain Reaction
3.
J Biol Chem ; 280(2): 1044-50, 2005 Jan 14.
Article in English | MEDLINE | ID: mdl-15520004

ABSTRACT

Fluorescence spectroscopy provides a direct method for evaluating the environment of a fluorescent ligand bound to its receptor. We utilized this methodology to determine the environment of Alexa within a cholecystokinin (CCK)-like probe (Alexa488-Gly-[(Nle(28,31))CCK-26-33]; CCK-8 probe) bound to the type A CCK receptor (Harikumar, K. G., Pinon, D. L., Wessels, W. S., Prendergast, F. G., and Miller, L. J. (2002) J. Biol. Chem. 277, 18552-18560). Here, we study this probe at the type B CCK receptor and develop another probe with its fluorophore closer to the carboxyl-terminal pharmacophore of type B receptor ligands (Alexa488-Trp-Nle-Asp-Phe-NH2; CCK-4 probe). Both probes bound to type B CCK receptors in a saturable and specific manner and represented full agonists. Similar to the type A receptor, at the type B receptor these probes exhibited shorter lifetimes and lower anisotropy when the receptor was in the active conformation than when it was shifted to its inactive, G protein-uncoupled state using guanosine 5'-[beta,gamma-imido]-triphosphate trisodium salt. Absolute values for lifetime and anisotropy were lower for the CCK-8 probe bound to the type B receptor than for this probe bound to the type A receptor, and Alexa fluorescence was more easily quenched by iodide at the type B receptor. This represents the first direct evidence that, despite having identical affinities for binding and potencies for activating type A and B receptors, CCK is docked via distinct mechanisms, with the amino terminus more exposed to the aqueous milieu when bound to the type B CCK receptor than to the type A CCK receptor. Of interest, despite this difference in binding, activation of both receptors results in analogous direction of movement of the fluorescent indicator probes.


Subject(s)
Peptides/metabolism , Peptides/pharmacology , Receptors, Cholecystokinin/agonists , Receptors, Cholecystokinin/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cholecystokinin/chemistry , Cholecystokinin/metabolism , Cricetinae , Fluorescence Polarization , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Fluorescent Dyes/pharmacology , Guanylyl Imidodiphosphate/pharmacology , Humans , Iodides/pharmacology , Ligands , Molecular Probes/chemistry , Molecular Probes/metabolism , Molecular Probes/pharmacology , Peptides/chemistry , Protein Binding/drug effects , Receptors, Cholecystokinin/classification , Receptors, Cholecystokinin/genetics , Spectrometry, Fluorescence
4.
Regul Pept ; 111(1-3): 103-9, 2003 Mar 28.
Article in English | MEDLINE | ID: mdl-12609756

ABSTRACT

This study was undertaken to confirm the presence of CCK receptor subtypes in calf pancreas and establish their cellular localization. Using specific antibodies against CCKA and CCKB receptors, somatostatin, glucagon and insulin, we were able to confirm by Western blot the presence of both CCK receptor protein subtypes in the calf pancreas as a 80-85-kDa CCKA receptor and 40-45-kDa CCKB receptor. By immunofluorescence, the CCKB receptor colocalizes with the islets' somatostatin delta cells, confirming what was previously shown in other species, as well as on ductal cells. We could not reproduce in the calf its colocalization with glucagon alpha cells as observed in human and rat. Any specific localization of CCKA receptors with our multiple antibodies failed. Our observation that the CCKB receptor subtype is specifically localized on pancreatic delta cells as well as on ductal cells lets us support the hypothesis that in this species, CCK could be involved in somatostatin metabolism as well as hydrelatic secretion; its effect on enzyme secretion would be indirect.


Subject(s)
Cattle/metabolism , Pancreas/metabolism , Receptors, Cholecystokinin/metabolism , Animals , Blotting, Western , Fluorescent Antibody Technique , Glucagon/isolation & purification , Immunohistochemistry , Insulin/isolation & purification , Islets of Langerhans/metabolism , Keratins/isolation & purification , Male , Pancreas/cytology , Receptors, Cholecystokinin/classification , Somatostatin/isolation & purification
5.
Scand J Gastroenterol ; 36(7): 738-43, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11444473

ABSTRACT

BACKGROUND: It has been assumed that gastrin stimulates the growth of pancreatic cancer in an autocrine way through co-expression of gastrin and the cholecystokinin-B receptor (CCK-BR). However, pancreatic cancer cell lines established directly from patients have revealed a great heterogeneity in cell proliferation when exposed to CCK, gastrin and their receptor antagonists. The aim of this study was therefore to examine co-expression of CCK-A and CCK-B receptor (CCK-AR and CCK-BR), and gastrin mRNA as well as the secretion of CCK and gastrin peptides in these cell lines. METHODS: Fourteen cell lines were established from primary pancreatic cancers or their metastases. Total RNA was isolated from the cell lines and reverse-transcribed into single-stranded cDNA. A PCR technique based on Taq polymerase-antibody interaction and CCK-AR, CCK-BR and gastrin-specific primers, followed by Southern blot analysis, were the methods used. The incubation mediums were analysed for the presence of secreted CCK/proCCK and gastrin/progastrin peptides by specific radioimmunoassays (RIA). RESULTS: By means of nested Reverse-Transcribed Polymerase Chain Reaction (nested RT-PCR), combined with Southem blot analysis of the PCR amplified products, CCK-AR and gastrin mRNA co-expression was detected in cell lines LPC-6p and LPC-10m, whereas CCK-BR and gastrin mRNA could be detected in cell lines LPC-8p and LPC-12m. A low level of secreted CCK peptides was detected in cell line LPC-6p, which also expressed CCK-AR mRNA. In no other cases were CCK or gastrin peptides detected in the cell culture mediums. CONCLUSION: The lack of CCK-BR and gastrin mRNA co-expression, and not detectable levels of secreted CCK and gastrin in culture media, does not lend support to the hypothesis that concomitant gene-expression of CCK receptors and gastrin or CCK are essential to maintaining pancreatic cancer cell proliferation.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Cholecystokinin/genetics , Gastrins/physiology , Gene Expression Regulation, Neoplastic/physiology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA, Messenger/physiology , Receptors, Cholecystokinin/genetics , Aged , Biopsy , Blotting, Southern , Cell Division , Cholecystokinin/analysis , Cholecystokinin/antagonists & inhibitors , Cholecystokinin/classification , Gastrins/analysis , Gastrins/antagonists & inhibitors , Humans , Middle Aged , Neoplasm Staging , RNA, Messenger/analysis , Radioimmunoassay , Receptors, Cholecystokinin/analysis , Receptors, Cholecystokinin/antagonists & inhibitors , Receptors, Cholecystokinin/classification , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
7.
Can J Physiol Pharmacol ; 77(7): 490-504, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10535709

ABSTRACT

We performed anatomical and physiological studies to determine the site and actions of sulfated cholecystokinin octapeptide (CCK8-S) on breathing. Peptide locations were determined by combined immunodetection of CCK8-S- containing synaptic varicosities and retrograde labeling of medullary neurons projecting to the ventral respiratory group. Retrogradely labeled neurons and CCK8-S immunolabeled varicosities overlapped within the nuclei of the solitary tract, ventral respiratory group, and the Kolliker-Fuse nucleus. Additional CCK8-S immunoreactive terminals were located in the rostroventrolateral medullary reticular nucleus, lateral paragigantocellular reticular nucleus, and the caudal pontine reticular nucleus. The respiratory effects of CCK8-S, which binds to CCK(A) and CCK(B) receptors, were examined by intravenous injection in adult rats and by bath application in the in vitro neonatal rat brainstem - spinal cord preparation. CCK8-S produced an increase in the mean amplitude of diaphragmatic electromyogram (EMG) of 28 +/- 35% (SD) and a decrease in mean respiratory interval of 13 +/- 4% in vivo. In vitro, CCK8-S significantly increased inspiratory duration and decreased respiratory interval, primarily by shortening expiratory duration. CCK8-unsulfated, a specific agonist for CCK(B) receptors, did not produce these effects. CCK8-S effects in the in vitro preparation were partially blocked by the CCK receptor antagonist lorglumide (final bath concentration 600 nM). These results suggest that CCK8-S modulates the respiratory rhythm via CCK(A) receptors within one or more medullary or pontine respiratory groups in both neonatal and adult rats.


Subject(s)
Medulla Oblongata/drug effects , Pons/drug effects , Respiration/drug effects , Sincalide/pharmacology , Animals , Dose-Response Relationship, Drug , Female , Male , Medulla Oblongata/chemistry , Pons/chemistry , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Receptors, Cholecystokinin/classification , Receptors, Cholecystokinin/drug effects , Receptors, Cholecystokinin/physiology , Sincalide/analysis , Sulfates/pharmacology
8.
Peptides ; 20(7): 885-98, 1999.
Article in English | MEDLINE | ID: mdl-10477091

ABSTRACT

In addition to its fundamental role in stimulating gastric acid secretion, the peptide hormone gastrin induces growth-promoting effects on diversity of target cells. Various mechanisms, including endocrine, paracrine, and autocrine, have been proposed for gastrin's growth-promoting actions. The mitogenic effects of gastrin are mediated by specific cell surface receptors activated after gastrin binding. The functionally defined receptors for gastrin include cholecystokinin A (CCKA) receptor, which is discriminating for sulfated CCK8; cholecystokinin B (CCKB)/gastrin receptor, which binds gastrin17 sulfated, and nonsulfated CCK8 with nearly equal affinities; cholecystokinin C (CCKC), which is a low-affinity gastrin binding protein; and novel, high-affinity receptors selective for amidated gastrin, processing intermediates of gastrin, or both. The signaling pathways mediating gastrin's stimulation of the CCKB/gastrin receptor have been progressively outlined, and the pathways mediating other receptors have been slowly emerging. Engagement of the gastrin receptor initiates various biochemical and molecular events, including recruitment and activation of tyrosine kinases, activation of the phospholipase C signaling pathway leading to phosphoinositide breakdown, intracellular calcium mobilization and protein kinase C stimulation, activation of the mitogen-activated protein kinase pathway, and induction of early response genes. Current emphasis is on understanding the functional significance of processing intermediate forms of gastrin, and the receptor subtypes and pathways that promote the trophic/mitogenic effects of the different molecular forms of gastrin.


Subject(s)
Gastrins/pharmacology , Gastrins/physiology , Growth Substances/pharmacology , Growth Substances/physiology , Animals , Cell Division/drug effects , Cell Division/physiology , Humans , Mitogen-Activated Protein Kinases/physiology , Protein-Tyrosine Kinases/physiology , Receptors, Cholecystokinin/classification , Receptors, Cholecystokinin/drug effects , Receptors, Cholecystokinin/physiology , Signal Transduction , Transcription, Genetic , Type C Phospholipases/physiology
9.
Gen Pharmacol ; 31(4): 519-24, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9792209

ABSTRACT

1. The peptide hormone and neurotransmitter, cholecystokinin, is widely distributed throughout the gastrointestinal tract and central nervous system and mediates a diverse number of biological functions. 2. Two receptor subtypes, CCK-A and CCK-B, have been identified by both pharmacological characterization and molecular cloning. The CCK-A receptor is the predominant peripheral CCK receptor subtype and the CCK-B receptor is the predominant central CCK receptor. In addition, there are discrete populations of CCK-A receptors in the brain and CCK-B receptors are present in gastric mucosa. 3. Subtype selective antagonists have been developed which discriminate between the two receptor subtypes. One of the major chemical classes has exploited a benzodiazepine template present in asperlicin which was initially discovered in a natural product screen for CCK receptor antagonists. 4. The structurally related benzodiazepines L-365,260, L-740,093, and YM022 are selective antagonists of the CCK-B receptor subtype. Their in vitro pharmacological profiles have been characterized using the human CCK-B receptor expressed in CHO cells. 5. L-365,260 behaves in a manner consistent with that of a competitive antagonist and both L-740,093 and YM022 behave as insurmountable CCK-B receptor antagonists in vitro.


Subject(s)
Receptors, Cholecystokinin/antagonists & inhibitors , Animals , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Benzodiazepinones/chemistry , Benzodiazepinones/pharmacology , Cells, Cultured , Humans , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacology , Receptor, Cholecystokinin A , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/classification , Receptors, Cholecystokinin/physiology
10.
Regul Pept ; 78(1-3): 95-103, 1998 Nov 30.
Article in English | MEDLINE | ID: mdl-9879752

ABSTRACT

Cholecystokinin (CCK) stimulates insulin secretion. It is, however, not established whether CCK receptors are expressed in insulin-producing cells. We therefore investigated, by in situ hybridization, whether CCK-A or CCK-B receptor mRNA could be detected in normal rat pancreatic islets and in the rat insulinoma cell line, RINm5F. The CCK-A, but not the CCK-B, receptor transcript was detected in both islets and RINm5F cells. Islet CCK-A receptors were mostly confined to the center of the islets corresponding to the distribution of the B cells. In RINm5F cells, insulin release was not significantly affected by cholecystokinin (CCK)-8-S (10(-13) to 10(-7) M), which is in contrast to the insulinotropic effect of CCK-8-S in normal rat islets. Similarly, in FURA-2AM-loaded cells, CCK-8-S (10(-11) to 10(-7) M) was without effect on the intracellular Ca2+ concentration ([Ca2+]ic) in RINm5F cells, whereas CCK-8-S (10(-7) M) markedly increased [Ca2+]ic (by 366+/-2 nM (P < 0.001) in normal rat islet cells. We conclude that the CCK-A, but not the CCK-B, receptor subtype is expressed in both normal rat islets and in the rat insulinoma-derived cell line RINmS5F. There is, however, a functional difference between normal islets and the RINm5F cells with respect to effects of CCK-8-S on insulin release and [Ca2+]ic.


Subject(s)
Insulinoma/metabolism , Islets of Langerhans/metabolism , Receptors, Cholecystokinin/metabolism , Animals , Calcium/metabolism , Carbachol/pharmacology , Fura-2/analogs & derivatives , Fura-2/metabolism , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Immunohistochemistry , In Situ Hybridization , Insulin/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Male , Pancreas/cytology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cholecystokinin/classification , Sincalide/pharmacology , Tumor Cells, Cultured
11.
Behav Neurosci ; 111(6): 1375-82, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9438806

ABSTRACT

The aim of this study was to investigate the effect of selective cholecystokinin (CCK) antagonists on the development of a preference for the mother by newly born lambs. At birth lambs received an injection of the CCK-A antagonist devazepide (0.01 or 0.1 mg/kg), the CCK-B antagonist PD135158 (0.01 or 0.1 mg/kg), or saline for the controls (1 ml/kg). No major side effects were observed in the first 4 postnatal hours except that lambs receiving PD135158 displayed more exploratory behavior towards the maternal body than lambs from the other groups. When tested in a 2-choice test situation at 24 hr of age, lambs treated with PD135158 or saline spent significantly more time near their dams than near the alien ewes, whereas lambs treated with devazepide did not show any discrimination. The effect of devazepide persisted at 48 hr of age. The use of a CCK-A antagonist, but not a CCK-B antagonist, was concluded to prevent the formation of a preferential relationship between the lamb and its mother, most probably by impairing neonatal learning.


Subject(s)
Animals, Newborn/physiology , Behavior, Animal , Imprinting, Psychological/physiology , Object Attachment , Receptors, Cholecystokinin/physiology , Sheep/physiology , Animals , Animals, Newborn/psychology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Benzodiazepinones/pharmacology , Choice Behavior/drug effects , Devazepide , Discrimination Learning/drug effects , Discrimination Learning/physiology , Exploratory Behavior/drug effects , Hormone Antagonists/pharmacology , Imprinting, Psychological/drug effects , Indoles/pharmacology , Meglumine/analogs & derivatives , Meglumine/pharmacology , Receptors, Cholecystokinin/antagonists & inhibitors , Receptors, Cholecystokinin/classification , Receptors, Cholecystokinin/drug effects , Sheep/psychology , Social Behavior
13.
Peptides ; 17(3): 483-8, 1996.
Article in English | MEDLINE | ID: mdl-8735976

ABSTRACT

Administration of a relatively large IP dose of sulfated cholecystokinin (26-33) (CCK-8; 1.0 mumol/kg) consistently induced moderate taste aversion conditioning (TAC) using a 20-min, one-bottle test in Long-Evans rats. Because CCK-8 has affinity for both CCKA and CCKB receptor subtypes, we wanted to determine the subtype involved in CCK-8-induced TAC. Pretreatment with the selective CCKA antagonist MK-329 (L-364, 718 or devazepide), at doses of 0.1, 1.0, or 10.0 mumol/kg, markedly antagonized (> 70%) CCK-8-induced TAC. Pretreatment with the selective CCKB antagonist L-365,260, at doses of 0.1 or 1.0 mumol/kg, partially antagonized (approximately 50%) CCK-8-induced TAC, although the highest dose of L-365,260. 10.0 mumol/kg, did not. These partial antagonistic effects of L-365,260 on CCK-8-induced TAC were replicated in our second study. In our third study, we observed that another CCKB antagonist, the dipeptoid CI-988, also partially antagonized CCK-8-induced TAC at a dose of 0.1, but not 1.0 or 10.0, mumol/kg. In our final study, pretreatments with a single dose (i.e., 10.0, but not 0.1 or 1.0, mumol/kg) of either MK-329 or L-365,260 were also shown to partially antagonize the formation of moderate TAC induced by treatment with LiCl at 708 mumol/kg. Marked antagonism of LiCl-induced TAC was also observed following pretreatment with the known anxiolytic chlordiazepoxide HCl at 7.4 mumol/kg. Considering the existing data on the induction of TAC by various CCK analogues, we consider an action of CCK-8 on peripheral CCKA, but not CCKB, receptors necessary for the induction of TAC. Our results of partial antagonism of CCK-8- and LiCl-induced TAC by L-365,260, CI-988, or MK-329 suggest, but do not prove, that both CCKA and CCKB mechanisms may be operative during TAC. Because the CCK antagonists affected TAC like chlordiazepoxide, blockade of CCKA and CCKB mechanisms may produce a mild anxiolytic effect.


Subject(s)
Conditioning, Psychological/drug effects , Lithium Chloride/pharmacology , Phenylurea Compounds , Receptors, Cholecystokinin/metabolism , Sincalide/pharmacology , Taste/drug effects , Animals , Benzodiazepinones/pharmacology , Chlordiazepoxide/pharmacology , Devazepide , Dose-Response Relationship, Drug , Hormone Antagonists/pharmacology , Indoles/pharmacology , Male , Meglumine/analogs & derivatives , Meglumine/pharmacology , Rats , Rats, Inbred Strains , Receptor, Cholecystokinin A , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/antagonists & inhibitors , Receptors, Cholecystokinin/classification
14.
Eur J Neurosci ; 7(12): 2364-9, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-8845940

ABSTRACT

The effect of iontophoretically applied cholecystokinin (CCK) on neurons of the neostriatum was studied in rats anaesthetized with urethane. The most frequently observed effect of the sulphated octapeptide (CCK-8S) on striatal neurons was excitation. Spontaneously active neurons responded more often to CCK-8S than quiescent cells. Silent, primarily non-responsive neurons could often be stimulated with CCK-8S using glutamate to induce an ongoing discharge. Thus, 45.8% of the 177 neurons studied changed their discharge rate by more than 30%. Certain CCK receptor antagonists could prevent the effect of CCK-8S, fully or at least partly, in the majority of CCK-responsive neurons. The data suggest that cholecystokinin modulates the firing of active neostriatal neurons via the CCKA or the CCKB receptor type. Furthermore, we compared neuronal responses to glutamate with those recorded during concomitant administration of CCK-8S in order to study the interaction of both transmitters, which may be colocalized in striatal afferents. CCK-8S mainly enhanced the excitatory effect of glutamate on striatal neurons, but in several neurons the response to glutamate was reduced. The CCKB receptor antagonist could prevent CCK-8S from increasing the glutamate-induced activation.


Subject(s)
Neostriatum/drug effects , Receptors, Cholecystokinin/drug effects , Sincalide/pharmacology , Animals , Drug Interactions , Glutamic Acid/pharmacology , Male , Membrane Potentials/drug effects , Neurons/drug effects , Rats , Rats, Wistar , Receptors, Cholecystokinin/classification
16.
Am J Physiol ; 268(4 Pt 1): G605-12, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7733286

ABSTRACT

Functional studies suggest that guinea pig chief cells have both cholecystokinin-A (CCK-A) and CCK-B receptors (CCK-A-R and CCK-B-R, respectively). However, all efforts to directly characterize the specific CCK-A-R using binding have been unsuccessful. Recent studies describe specific CCK-A-R agonists such as A-71378 ([desamino-Nle28,31-(N-methyl)Asp32]CCK heptapeptide]. In the present study, [D-Tyr-Gly]A-71378 was synthesized, which has > 300-fold selectivity for CCK-A-R and can be iodinated. [D-Tyr-Gly]A-71378 was equipotent to A-71378 in stimulating pepsinogen release from purified guinea pig chief cells. Binding of 125I-labeled [D-Tyr-Gly]A-71378 was saturable and specific. Potencies for inhibiting binding were as follows: [D-Tyr-Gly]A-71378 = A-71378 = 4x CCK octapeptide (CCK-8) > 1,000x des(SO4)-CCK-8, gastrin. In contrast, for 125I-gastrin binding they were CCK-8 > gastrin-17-I > des(SO4)-CCK-8 >> A-71378 or [D-Tyr-Gly]A-71378. Binding of [D-Tyr-Gly]A-71378 was best fitted by a two-site model. In contrast, 125I-gastrin binding was fitted with a single-site model. For inhibiting binding of 125I-[D-Tyr-Gly]A-71378, the CCK antagonists had relative affinities of L-364,718 >> L-365,260, and the reverse was true with 125I-gastrin. Correlation of binding with changes in biological activity suggested low-affinity CCK-A-R were mediating these changes. These results demonstrate directly for the first time that guinea pig chief cells possess CCK-A-R and CCK-B-R. The pharmacology of these CCK-A-R resembles those on other tissues. This novel, highly selective CCK-A ligand should be useful because it will identify CCK-A-R when they make up as little as 0.2% of the total CCK receptor number.


Subject(s)
Gastric Mucosa/metabolism , Phenylurea Compounds , Receptors, Cholecystokinin/metabolism , Animals , Benzodiazepinones/pharmacology , Devazepide , Gastrins/antagonists & inhibitors , Guinea Pigs , Ligands , Male , Oligopeptides/metabolism , Pancreas/metabolism , Pepsinogens/metabolism , Receptors, Cholecystokinin/classification , Sincalide/pharmacology , Stomach/cytology
17.
Neuroreport ; 5(18): 2565-8, 1994 Dec 20.
Article in English | MEDLINE | ID: mdl-7696605

ABSTRACT

The ability of the selective cholecystokinin-B (CCK-B) receptor antagonist Cl 988 to reverse tolerance to morphine's antinociceptive effect was investigated with the hot-plate test in Sprague-Dawley rats. Tolerance was induced by subcutaneous (s.c.) injection of 10 mg kg-1 morphine twice daily for four days. On the fifth day rats were administered CI 988 (10 mg kg-1) or saline plus 5 mg kg-1 morphine s.c. Significant antinociception was observed in the group that received the CCK-B antagonist plus morphine, whereas the saline plus morphine group exhibited total tolerance. These results suggest that upregulation of the endogenous CCK system during repeated morphine administration may have an important role in the development of opiate tolerance.


Subject(s)
Indoles/pharmacology , Meglumine/analogs & derivatives , Morphine/pharmacology , Receptors, Cholecystokinin/antagonists & inhibitors , Analgesia , Animals , Dose-Response Relationship, Drug , Drug Synergism , Drug Tolerance , Male , Meglumine/pharmacology , Nociceptors/drug effects , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Receptors, Cholecystokinin/classification , Time Factors
18.
Gastroenterology ; 107(4): 1135-46, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7523219

ABSTRACT

BACKGROUND/AIMS: Cholecystokinin (CCK) and gastrin stimulate growth of rodent pancreas in vivo. However, it remains unclear whether these growth effects are mediated specifically by CCK-A receptors, CCK-B receptors, or both. To clarify this issue, the present study examined the effect of highly selective and biologically active CCK agonists on pancreatic growth. METHODS: Rats were subcutaneously injected with either (1) CCK-8, a nonselective CCK agonist (2.50 micrograms/kg body wt); (2) A-71623, a selective CCK-A agonist, tert-butyl-oxycarbonyl-Trp-Lys (epsilon-N-2-methylphenylaminocarbonyl)-Asp-(N-methyl)-Phe-NH2 (1.84 micrograms/kg body wt); (3) SNF-8815; a selective CCK-B agonist, [(2R,3S)-beta-MePhe28, N-MeNle31]CCK26-33 (2.40 micrograms/kg body wt); or (4) saline (control) for 21 days. Rats were killed, and pancreatic weight, protein content, RNA content, DNA content, protein-DNA ratio, RNA-DNA ratio, pancreatic area per nucleus, and number of mitoses per 10,000 acinar cells were determined. RESULTS: Nonselective CCK agonist significantly increased pancreatic weight, protein, RNA, and DNA contents, and number of mitoses per 10,000 acinar cells. Likewise, selective CCK-A agonist significantly increased pancreatic weight, protein, RNA, and DNA contents, protein-DNA ratio, RNA-DNA ratio, pancreatic area per nucleus, and number of mitoses per 10,000 acinar cells. In contrast, selective and biologically active CCK-B agonist had no effect. CONCLUSION: These findings indicate that pancreatic growth is mediated specifically by CCK-A receptors in the rat in vivo.


Subject(s)
Pancreas/growth & development , Receptors, Cholecystokinin/physiology , Animals , Binding, Competitive , Cholecystokinin/agonists , Cholecystokinin/analogs & derivatives , Cholecystokinin/pharmacology , DNA/metabolism , Gastrins/pharmacology , Male , Mitosis , Pancreas/cytology , Pancreas/metabolism , Peptide Fragments/pharmacology , Proteins/metabolism , RNA/metabolism , Rats , Rats, Inbred Lew , Receptor, Cholecystokinin A , Receptors, Cholecystokinin/classification , Receptors, Cholecystokinin/metabolism , Tetragastrin/analogs & derivatives , Tetragastrin/pharmacology
19.
Gastroenterology ; 107(4): 1019-30, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7926455

ABSTRACT

BACKGROUND/AIMS: Cholecystokinin (CCK) is a neuropeptide that exerts numerous effects in the gut. To determine the sites of action of CCK, the distribution and properties of CCK receptor subtypes were studied. METHODS: CCK receptors were localized by autoradiographic analysis of 125I-CCK binding to frozen sections of the canine upper gastrointestinal tract. RESULTS: In the cardiac and fundic stomach, CCK-B/gastrin receptors were found in the mucosa and in a subpopulation of neuronal elements in the circular muscle. The antrum expressed CCK-B/gastrin receptors in a few neurons in the circular muscle and in the entire myenteric plexus; no receptors were observed in the antral mucosa or esophagus. The duodenum showed a high concentration of CCK-B/gastrin receptors in the myenteric plexus. The cardiac and fundic basal mucosae expressed CCK-A receptors. Two nonpeptide CCK receptor antagonists were unable to differentiate between the receptor subtypes. CONCLUSIONS: The differential expression of CCK receptor subtypes in the gastric mucosa provides a morphological basis for the separate regulatory roles of CCK and gastrin in gastric function. CCK-B/gastrin receptor expression in a subset of neurons in gastric circular muscle suggests a novel site of action for CCK and/or gastrin.


Subject(s)
Digestive System/metabolism , Receptors, Cholecystokinin/metabolism , Animals , Autoradiography , Binding Sites , Cholecystokinin/metabolism , Densitometry , Digestive System/innervation , Dogs , Gastric Mucosa/metabolism , Myenteric Plexus/cytology , Myenteric Plexus/metabolism , Neurons/metabolism , Radioligand Assay , Receptors, Cholecystokinin/classification , Tissue Distribution
20.
Gastroenterology ; 107(4): 1059-66, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7926458

ABSTRACT

BACKGROUND/AIMS: Transient lower esophageal sphincter relaxations have been found to be involved in gastroesophageal reflux. The purpose of this study was to determine whether cholecystokinin (CCK) and nitric oxide are involved in the occurrence of these relaxations. METHODS: Pharyngeal, esophageal, lower esophageal sphincter, and gastric pressures were monitored in five dogs through a cervical esophagostomy. Gastric distentions with air, at a constant pressure of 1.56 kPa, were performed for 30-minute sessions using a barostat. RESULTS: During gastric distention at 1.56 kPa, transient relaxations occurred at a mean rate of 7.2 +/- 0.6 every 30 minutes. CCK-8 infused intravenously (0.1-1 microgram.kg-1.h-1) dose dependently increased the occurrence of relaxations while it was reduced by the CCK-A receptor antagonist devazepide but not the CCK-B antagonist L365260, both administered intravenously in a dose range of 0.1-100 micrograms/kg. The two antagonists administered intracerebroventricularly (1 microgram/kg) did not modify the occurrence of relaxations. Both devazepide and L365260 (10 micrograms/kg) reduced the CCK-induced relaxations, but devazepide was more potent. The nitric oxide synthase inhibitor NG-nitro-L-arginine-methyl ester (20 mg/kg) reduced the number of relaxations during gastric distention in the presence or absence of CCK infusion. This effect was reversed by L-arginine but not D-arginine (200 mg/kg). CONCLUSIONS: CCK is involved in the occurrence of transient lower esophageal sphincter relaxations through peripheral CCK-A receptors and an L-arginine nitric oxide pathway.


Subject(s)
Cholecystokinin/physiology , Esophagogastric Junction/physiology , Muscle Relaxation , Nitric Oxide/physiology , Stomach/physiology , Air , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Dogs , NG-Nitroarginine Methyl Ester , Nitric Oxide/antagonists & inhibitors , Pressure , Receptors, Cholecystokinin/antagonists & inhibitors , Receptors, Cholecystokinin/classification , Sincalide/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...