Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Pharmacol ; 58(6): 1381-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11093777

ABSTRACT

It has previously been reported that the cholecystokinin analog JMV-180 behaves differently on the rat and the mouse cholecystokinin-A receptor (CCK-AR). In mice this analog acts as an agonist on low- and high-affinity sites of the CCK-AR, whereas in rats this compound acts as an agonist on high-affinity sites and as an antagonist on low-affinity sites. In an attempt to understand why the same compound behaves differently on these two CCK-A receptors, we cloned the cDNA encoding the mouse CCK-AR. We then investigated a cellular model able to mimic the effect that was observed in rats and mice. HeLa cells were transiently cotransfected with plasmids leading to expression of the rat or mouse CCK-AR in the presence of pFos-Luc as reporter plasmid; such a plasmid placed the regulatory part of the human c-Fos gene upstream from the firefly luciferase structural gene (Luc). We then observed that the two CCK-A receptors behaved differently, not only in the presence of compound JMV-180 but also in the presence of cholecystokinin or even in absence of ligand; the rat CCK-AR was 2 to 3 times more potent than the mouse CCK-AR in inducing the reporter protein, whatever the ligand studied. This result was confirmed using the same kind of experiment with the reporter plasmid p(TRE)(3)-tk-Luc. Using various mutated receptors, we investigated the role of the putative third intracellular loop. We concluded that both the primary structure of the receptor and the cellular context are in part responsible for the differential behavior of these CCK-A receptors.


Subject(s)
Gene Expression Regulation , Receptors, Cholecystokinin/physiology , Animals , COS Cells , Cloning, Molecular , DNA, Complementary/analysis , HeLa Cells , Humans , Mice , Models, Biological , Rats , Receptor, Cholecystokinin A , Receptors, Cholecystokinin/genetics , Transcriptional Activation , Transfection
2.
Eur J Pharmacol ; 403(1-2): 17-25, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10969139

ABSTRACT

alpha-amidation of a peptide (which takes place from a glycine-extended precursor) is required to produce biologically active amidated hormones, such as gastrin-releasing peptide (GRP)/Pyr-Gln-Arg-Leu-Gly-Asn-Gln-Trp-Ala-Val-Gly-His-Leu-Met-NH(2) (bombesin). It was shown that glycine-extended gastrin mediates mitogenic effects on various cell lines by interacting with a specific receptor, different from the classical CCK(1) or CCK(2) receptors. On the basis of this observation, we have extended the concept of obtaining active glycine-extended forms of others amidated peptides to produce new active analogues. In this study, we have tested the biological behaviour of a synthetic analogue of the glycine-extended bombesin (para-hydroxy-phenyl-propionyl-Gln-Trp-Ala-Val-Gly-His-Leu-Met-Gly-OH or JMV-1458) on various in vitro models. We showed that compound JMV-1458 was able to inhibit specific (3-[125I]iodotyrosyl(15)) GRP ([125I]GRP) binding in rat pancreatic acini and in Swiss 3T3 cells with K(i) values of approximately 10(-8) M. In isolated rat pancreatic acini, we found that JMV-1458 induced inositol phosphates production and amylase secretion in a dose-dependent manner. In Swiss 3T3 cells, the glycine-extended bombesin analogue dose-dependently produced [3H]thymidine incorporation. By using potent GRP/bombesin receptor antagonists, we showed that this synthetic glycine-extended bombesin analogue induces its biological activities via the classical GRP/bombesin receptor.


Subject(s)
Bombesin/analogs & derivatives , Bombesin/metabolism , Receptors, Bombesin/metabolism , 3T3 Cells , Amylases/drug effects , Amylases/metabolism , Animals , Binding, Competitive/drug effects , Bombesin/chemistry , Bombesin/pharmacology , Cell Division/drug effects , Dose-Response Relationship, Drug , Inositol Phosphates/metabolism , Iodine Radioisotopes , Male , Mice , Pancreas/cytology , Pancreas/metabolism , Rats , Rats, Wistar
3.
Br J Pharmacol ; 127(1): 259-67, 1999 May.
Article in English | MEDLINE | ID: mdl-10369481

ABSTRACT

The aim of this study was to analyse the antisecretory mechanism of L-365,260 in vitro in isolated rabbit gastric glands. We showed that compound L-365,260, described as a non-peptide specific competitive CCK-B receptor antagonist, was able to dose-dependently inhibit [14C]-aminopyrine accumulation induced by histamine (10(-4) M), carbachol (5x10(-5) M), 3-isobutyl-1-methyl-xanthine (IBMX) (5x10(-6) M) and forskolin (5x10(-7) M) with similar IC50 values respectively of 1.1+/-0.6x10(-7) M, 1.9+/-1.2x10(-7) M, 4.2+/-2.0x10(-7) M and 4.0+/-2.8x10(-7) M. We showed that L-365,260 acted beyond receptor activation and production of intracellular second messengers and that it had no action on the H+/K+ -ATPase. We found that L-365,260 inhibited cyclic AMP-induced [14C]-aminopyrine accumulation in digitonin-permeabilized rabbit gastric glands, suggesting that this compound acted, at least in part, as an inhibitor of the cyclic AMP-dependent protein kinase (PKA) pathway.


Subject(s)
Benzodiazepinones/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Gastric Acid/metabolism , Phenylurea Compounds/pharmacology , Receptors, Cholecystokinin/antagonists & inhibitors , Aminopyrine/metabolism , Animals , Cyclic AMP/biosynthesis , Gastric Mucosa/drug effects , Gastric Mucosa/enzymology , Gastric Mucosa/metabolism , Histamine H2 Antagonists/pharmacology , In Vitro Techniques , Inositol Phosphates/metabolism , Omeprazole/pharmacology , Proton Pump Inhibitors , Rabbits , Second Messenger Systems/drug effects
4.
Mol Pharmacol ; 52(2): 292-9, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9271352

ABSTRACT

The aim of this study was to analyze the role of cholecystokinin (CCK(B)) receptor in human lymphoblastic Jurkat T cells. We investigated the trophic effect resulting from activation of such a receptor by using the reporter gene strategy. For this purpose, we transiently transfected Jurkat T cells with the reporter plasmid p[(TRE)3-tk-Luc] and found that CCK-8 was able to dose-dependently induce luciferase expression related to activator protein-1 (AP-1) activation with a maximal response identical to that obtained with compounds known to activate AP-1 complex (quantitatively, the same level of induction was obtained with 1 nM 12-O-tetradecanoylphorbol-13-acetate, 100 microM diacylglycerol, or 4 nM epidermal growth factor). The involvement of the CCK(B) receptor in such a stimulation was demonstrated by the inhibiting effect of the selective CCK(B) receptor antagonist PD-135,158. This effect was confirmed in COS-7 cells transfected with the cDNA of CCK(B) receptor cloned from Jurkat T cells. To better understand the AP-1-dependent luciferase expression in Jurkat T cells, we tested two specific inhibitors of serine/threonine phosphatases-1 and -2A: okadaic acid and calyculin A. These compounds strongly increased the phorbol-12-myristate-13-acetate response, whereas we have not observed a contribution of phosphatase inhibitors on a CCK-8-induced luciferase activity. To confirm that CCK(B) receptors are involved in AP-1 response, we investigated the CCK-8 effect on interleukin-2 expression, a natural endogenous gene regulated by several factors, including AP-1. In Jurkat T cells activated by phorbol-12-myristate-13-acetate and phytohemagglutinin, CCK-8 induced IL-2 expression. This induction was abolished by PD-135,158. Our results indicate that CCK-8 exerts a trophic effect in Jurkat T cells through stimulation of CCK(B) receptors by modulation of expression of AP-1-regulated genes.


Subject(s)
Gene Expression Regulation, Neoplastic , Receptors, Cholecystokinin/physiology , Sincalide/pharmacology , T-Lymphocytes/physiology , Transcription Factor AP-1/physiology , Animals , Binding, Competitive , COS Cells , Cell Division , Cell Line , Cloning, Molecular , Humans , Interleukin-2/genetics , Jurkat Cells , Ligands , Transcription, Genetic/drug effects , Transcriptional Activation
5.
Neuropeptides ; 30(5): 433-7, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8923504

ABSTRACT

We have investigated the potential role of gastrin in the regulation of cell growth in human astrocytic tumors. To this end we have used synthetic analogs of gastrin and cholecystokinin (CCK) which behave as gastrin and/or CCK antagonists, e.g. compounds JMV-97, JMV-209 and JMV-179. Their effects on astrocytic tumor cell proliferation was investigated by the use of the colorimetric MTT assay. The in vitro biological models used in the present study included the SW1088, U87 and U373 astrocytic tumor cell lines. The results demonstrated marked influence of gastrin and CCK antagonists in the regulation of astrocytic tumor growth. This suggests that gastrin and/or CCK antagonists might be tested in experimental glioblastoma.


Subject(s)
Astrocytes/pathology , Astrocytoma/pathology , Brain Neoplasms/pathology , Cholecystokinin/antagonists & inhibitors , Gastrins/antagonists & inhibitors , Gastrins/pharmacology , Glioblastoma/pathology , Peptide Fragments/pharmacology , Sincalide/analogs & derivatives , Cholecystokinin/physiology , Gastrins/physiology , Humans , Sincalide/pharmacology , Tumor Cells, Cultured
6.
Braz J Med Biol Res ; 29(10): 1253-63, 1996 Oct.
Article in English | MEDLINE | ID: mdl-9181094

ABSTRACT

The synthesis of analogs of the C-terminal tridecapeptide of gastrin is described. These pseudopeptide analogs were obtained either by replacing the C-terminal phenylalanine amide with 2-phenylethylalcohol or with 2-phenylethylamine, or by replacing the peptide bond between Trp and Leu, or between Leu and Asp with an aminomethylene (CH2NH). The ability of these compounds to stimulate gastric acid secretion in anesthetized rats and to inhibit binding of labeled CCK-8 to isolated cells from rabbit fundic mucosa was tested. [desPhe13, Leu11]-HG-12-I-beta-phenylethylester 33, [desPhe13, Leu11]-HG-12-II-beta-phenylethylester 38, [desPhe13, Leu11]-HG-12-I-beta-phenylethylamide 32, and [desPhe13, Leu11]-HG-12-II-beta-phenylethylamide 37 acted as gastrin receptor antagonists, while [Trp10-psi(CH2NH)-Leu11]-HG-13-I 31 and [Trp10-psi(CH2NH)-Leu11]-HG-13-II 36 acted as agonists. Unexpectedly, [Leu11-psi(CH2NH)-Asp12]-HG-13-I 30 and [Leu11-psi (CH2NH)-Asp12]-HG-13-II 35 were almost devoid of affinity for the gastrin receptor.


Subject(s)
Gastrins/biosynthesis , Gastric Acid/metabolism , Gastrins/chemistry , Humans , Peptide Biosynthesis , Receptors, Cholecystokinin/antagonists & inhibitors
7.
Braz. j. med. biol. res ; 29(10): 1253-63, Oct. 1996. ilus, tab
Article in English | LILACS | ID: lil-186171

ABSTRACT

The synthesis of analogs of the C-terminal tridecapeptide of gastrin in described. These pseudopeptide analogs were obtained either by replacing the C-terminal phenylalanine amide with 2-phenylethytalcohol or with 2-phenylethylamine, or by replacing the peptid bond between Trp and Leu, or between Leu and Asp with an aminomethylene (CH2NH). The ability of these compounds to stimulate gastric acid secretion in anesthetized rats and to inhibit binding of labeled CCK-8 to isolated cells from rabbit fundic mucosa was tested. [desPhe13, Leu11]-HG-12-I-beta-phenylethylester 33, [desPhe13, Leu11]-HG-12-II-beta-phenylethylester 38 [desPhe13, Leu11]-HG-12-I-beta-phenylethylamide 32, and [desPhe13, Leu11]-HG-12-II-beta-phenylethylamide 37 acted as gastrin receptor antagonists, while [Trp10-((CH2NH)-Leu11]-HG-13-I 31 and (Trp10-((CH2NH)-Leu11]-HG-13-II 36 acted as agonists. Unexpectedly, [Leu11-((CH2NH)-Asp12]-HG-13-I 30 and [Leu11-((CH2NH)-Asp12]-HG-13-II 35 were almost devoid of affinity for the gastrin receptor.


Subject(s)
Humans , Gastric Acid/metabolism , Gastrins/biosynthesis , Peptides/biosynthesis , Receptors, Cholecystokinin/antagonists & inhibitors , Gastrins/chemistry , Peptides/chemical synthesis
8.
Eur J Pharmacol ; 294(2-3): 511-9, 1995 Dec 29.
Article in English | MEDLINE | ID: mdl-8750713

ABSTRACT

In the present study we compared various CCK(B) receptor antagonists and tried to detect a difference in biological activity between the C-terminal octapeptides of cholecystokinin (CCK-8) and [Leu11]gastrin-(5-17) in isolated rabbit gastric glands. Binding experiments showed that different CCK(B)/gastrin receptor agonists bound with high affinity and that antagonists inhibited this binding in accordance with a CCK(B)/gastrin pharmacological profile. [Leu11]gastrin-(5-17), CCK-8 and cionin were found to induce [14C]aminopyrine accumulation to 25% above the basal level. Under the same experimental conditions, histamine induced a response twice as great as the response obtained with [Leu11]gastrin-(5-17) or CCK-8. [Leu11]gastrin-(5-17) (10(-7) M), CCK-8 (10(-8) M) and cionin (10(-8) M) appeared to be full agonists. CCK(B)/gastrin receptor antagonists including L-365,260 (3R-(+)-N-(2,3-dihydro-1-methyl-2-oxo-5-phenyl-1H-1,4-benzodiazepin++ +-3-yl)-N-(3-methylphenyl) urea), L-364,718 (3S-(-)-N-(2,3-dihydro-1-methyl-2-oxo-5-phenyl-1H-1,4-benzodiazepin++ +-3-yl)-1H-indole-2-carboximide) (a selective CCK(A) receptor antagonist), PD-135,158 (4([2-[[3-(1H-indol-3-yl)-2-methyl-1-oxo-2-[[[1.7.7-trimethyl-bicyclo[2. 2.1]hept-2-yl)oxy]carbonyl]amino]propyl]amino]-1-phenylethyl] amino-4-oxo-[1S-1alpha.2beta[S*(S*)]4alpha]]-butano nate N-methyl-D-glucamine) (bicyclo system 1S-endo), YM-022 ((R)-1-[2,3-dihydro-1-(2'-methylphenacyl)-2-oxo-5-phenyl-1H-1,4-++ +benzodiazepin-3-yl]-3-(3-methylphenyl)urea) and JMV-180 (Boc-Tyr(SO3H)-Nle-Gly-Trp-Nle-Asp-O-CH2-CH2-C6H5) exhibited the same profile for inhibition of [Leu11]gastrin-(5-17) or CCK-8-induced [14C]aminopyrine accumulation in rabbit gastric glands. These results suggested that [Leu11]gastrin-(5-17) and CCK-8 induced [14C]aminopyrine accumulation by the same mechanism. [Leu11]gastrin-(5-17)- or CCK-8-induced [14C]aminopyrine accumulation was inhibited by about 40% by the histamine H2 receptor blocker cimetidine. These results are consistent with there being cooperativity between [Leu11]gastrin-(5-17) (or CCK-8) and histamine in the acid secretory pathway. Similarly, the CCK(B)/gastrin receptor antagonists were tested against histamine-induced [14C]aminopyrine accumulation and surprisingly, only compound L-365,260 appeared active and even more potent than cimetidine.


Subject(s)
Gastric Acid/metabolism , Gastric Mucosa/drug effects , Gastrins/pharmacology , Sincalide/pharmacology , Amino Acid Sequence , Aminopyrine/metabolism , Animals , Gastric Mucosa/metabolism , Histamine/pharmacology , In Vitro Techniques , Molecular Sequence Data , Rabbits , Receptors, Cholecystokinin/drug effects , Receptors, Cholecystokinin/metabolism
9.
Eur J Pharmacol ; 290(2): 61-7, 1995 Jul 18.
Article in English | MEDLINE | ID: mdl-8575534

ABSTRACT

We have shown that gastrin and cholecystokinin octapeptide (CCK-8) are differently coupled to G protein (GTP-binding protein) through type B cholecystokinin receptors in guinea-pig brain membranes and Jurkat cells. Indeed, the gastrin-13 binding affinity is strongly reduced by stable guanyl nucleotides, whereas CCK-8 binding is only slightly affected. In order to determine the structural requirements regulating such coupling, we have synthesized several gastrin and cholecystokinin fragments (sulphated or unsulphated) elongated at the N-terminus of the common C-terminal tetrapeptide. We investigated their interaction with CCKB receptors in guinea pig brain membranes and Jurkat cells and their involvement in the G protein coupling. Their apparent binding affinities to CCKB receptors were measured by inhibition of [125I]Bolton Hunter-CCK-8 (3-[125I]iodo-4-hydroxyphenyl)propionyl-CCK-8) binding in the presence or absence of GTP gamma S (guanosine 5'-O-(3-thio)triphosphate) or aluminum tetrafluoride (AlF4-). Activation of the G proteins by GTP gamma S or AlF4- led to a decrease in binding affinity for the gastrin related peptides, the common CCK-gastrin C-terminal forms, the cholecystokinin hexapeptide and the unsulphated cholecystokinin heptapeptide. Sulphated CCK-7, CCK-8, and cionin apparent binding affinities were not affected. These finding indicated that the sulphated tyrosine in position 7 in CCK (as counted from the C-terminus), provides the cholecystokinin selectivity for the CCKB receptor compared to gastrin. The results are discussed with the aim to better clarify the physiological relevance of gastrin and cholecystokinin toward CCKB receptors and their related intracellular events.


Subject(s)
Gastrins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Receptors, Cholecystokinin/metabolism , Sincalide/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Cell Line , Cell Membrane/metabolism , Guinea Pigs , Humans , Male , Molecular Sequence Data , T-Lymphocytes/metabolism
10.
Int J Pept Protein Res ; 44(4): 348-56, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7875937

ABSTRACT

In the course of our study concerning gastrin and cholecystokinin (CCK) receptors, we have synthesized and characterized a new labeled gastrin ligand, 125I-BH-[Leu15]-gastrin-(5-17) [(3-[125I]iodo-4-hydroxyphenyl)-propionyl-[Leu15]-gastrin-(5-17)]. Binding of 125I-BH-[Leu15]-gastrin-(5-17) to isolated canine fundic mucosal cells was specific, saturable and of high affinity. 125I-BH-[Leu15]-gastrin- (5-17) and 125I-BH-CCK-8[(3-[125I]iodo-4-hydroxyphenyl)-propionyl-CCK-8] interact with isolated canine fundic mucosal cells with small differences in maximal binding capacities and affinities, 3800 +/- 900 binding sites/cell (Kd = 0.52 +/- 0.23 nM) and 6200 +/- 1100 binding sites/cell (Kd = 0.31 +/- 0.18 nM), respectively. The relative order of potencies for gastrin and CCK analogs in displacing 125I-BH-[Leu15]-gastrin-(5-17) binding correlated well with those obtained using 125I-BH-CCK-8. Selective CCK/gastrin antagonists L-364,718 (MK-329) and L-365,260 also inhibited 125I-BH-[Leu15]-gastrin-(5-17) binding. These results indicate that 125I-BH-[Leu15]-gastrin-(5-17) binds to gastrin receptors in isolated canine fundic mucosal cells. We have also characterized 125I-BH-[Leu15]-gastrin-(5-17) binding to the human Jurkat lymphoblastic cell line (Jurkat cells) known to express the CCK-B/gastrin receptor. Saturation experiments have shown that both 125I-BH-[Leu15]-gastrin-(5-17) and 125I-BH-CCK-8 interact with a single class of high-affinity binding sites in the Jurkat cell line.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Gastric Mucosa/metabolism , Gastrins/chemical synthesis , Gastrins/metabolism , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Phenylurea Compounds , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Benzodiazepinones/pharmacology , Calcium/metabolism , Cell Line , Cholecystokinin/antagonists & inhibitors , Devazepide , Dogs , Gastric Fundus/metabolism , Gastric Mucosa/drug effects , Gastrins/pharmacology , Humans , Iodine Radioisotopes , Kinetics , Molecular Sequence Data , Peptide Fragments/pharmacology , Receptors, Cholecystokinin/antagonists & inhibitors , Sincalide/pharmacology , T-Lymphocytes/drug effects , Temperature
11.
Eur J Pharmacol ; 267(3): 297-305, 1994 May 17.
Article in English | MEDLINE | ID: mdl-8088368

ABSTRACT

In the course of our study concerning gastrin and cholecystokinin (CCK) receptors, we synthesized and characterized a labelled gastrin ligand, [125I]BH[Leu15]gastrin-(5-17) (3-(3-[125I]iodo-4-hydroxyphenyl)propionyl[Leu15]gastrin-(5-17)). On isolated canine fundic mucosal cells and human Jurkat lymphoblastic cell line, known to express CCKB/gastrin receptors, the binding experiments performed indicate that [125I]BH[Leu15]gastrin-(5-17) provides a convenient biologically active ligand for cholecystokinin/gastrin receptor studies. We showed in this study that, on guinea-pig brain membranes known to possess CCKB and CCKA receptors, [125I]BH[Leu15]gastrin-(5-17) binds to a single class of high-affinity binding sites in a saturable and specific manner. [125I]BH[Leu15]gastrin-(5-17) interacts with guinea-pig brain membranes with a maximal binding capacity that is about three-fold lower than that of [125I]BHCCK8 (CCK8, the C-terminal octapeptide of cholecystokinin). The apparent affinities of CCK analogues and selective CCK receptor antagonists L-365,260 and MK-329 for the sites labelled by both probes were in accordance with a CCKB-like profile. Association-dissociation kinetics of [125I]BH[Leu15]gastrin-(5-17) and [125I]BHCCK8 were performed and compared. They showed that [125I]BHCCK8 equilibrated more slowly than [125I]BH[Leu15]gastrin-(5-17). The effects of pH, monovalent and divalent cations on binding of both probes were investigated. The results obtained did not indicate strong differences between [125I]BH[Leu15]gastrin-(5-17) and [125I]BHCCK8 binding. Binding experiments in the presence of stable analogues of GTP showed a different behavior between [125I]BH[Leu15]gastrin-(5-17) and [125I]BHCCK8. GTP gamma S strongly decreased [125I]BH[Leu15]gastrin-(5-17) binding whereas it weakly affected [125I]BHCCK8 binding.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Brain/metabolism , GTP-Binding Proteins/metabolism , Gastrins/metabolism , Peptide Fragments/metabolism , Phenylurea Compounds , Sincalide/metabolism , Animals , Benzodiazepinones/metabolism , Binding Sites , Devazepide , Gastrins/chemistry , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Guinea Pigs , In Vitro Techniques , Male
13.
Peptides ; 13(3): 519-25, 1992.
Article in English | MEDLINE | ID: mdl-1523164

ABSTRACT

Binding to gastrin receptors and gastric acid secretion experiments were performed with gastrin derivatives modified at the C-terminal tetrapeptide amide from HG-13 sequence. 1. When the ultimate phenylalanine amide was replaced by a phenethylester or a phenetylamide moiety, the resulting compound bound to gastrin receptors (Kd approximately 10 nM) and exhibited antagonist activity on gastrin-induced acid secretion in the anesthetized rat. 2. Changing the peptide bond between Trp and Leu residues to a -omega(CH2-NH)- bond resulted in a compound which also bound to gastrin receptors (Kd approximately 10 nM) but presented agonist activity on acid secretion in the rat. In contrast, when the peptide bond between Leu and Asp residues was replaced by a -omega(CH2-NH)- bond, the resulting compound was devoid of any affinity for gastrin receptor (Kd greater than 10(-6) M) and of any biological activity. 3. The HG-13 derivatives were synthesized in sulfated and unsulfated forms: O-sulfation of the HG-13 tyrosine residue did not change its intrinsic in vivo activity but enhanced its affinity for gastrin receptors (Kd approximately 0.3 nM). On the contrary, O-sulfation of the various chemically modified HG-13 had no significant effect in either in vitro or in vivo experiments. 4. Finally, no significant difference between binding on parietal (F3) and nonparietal (F1) cells was observed, in agreement with the presence of a gastrin-type receptor in these two cell populations.


Subject(s)
Gastrins/pharmacology , Parietal Cells, Gastric/metabolism , Receptors, Cholecystokinin/metabolism , Amino Acid Sequence , Animals , Dose-Response Relationship, Drug , Gastric Acid/metabolism , Gastrins/chemistry , Humans , Molecular Sequence Data , Parietal Cells, Gastric/drug effects , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Rabbits , Sincalide/metabolism , Structure-Activity Relationship
14.
Am J Physiol ; 260(2 Pt 1): G182-8, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1996638

ABSTRACT

Previous studies have demonstrated that gastrin and the COOH-terminal octapeptide of cholecystokinin (CCK-8) stimulated in vitro acid secretion from isolated rabbit gastric parietal cells. Both peptides bind to receptor sites located on these cells and induce an increase in phosphoinositide turnover and an uptake of [14C]aminopyrine ([14C]AP) with the same efficacy and potency. In the present study, we used the 3-(benzoylamino)-benzodiazepine analogue L365,260 and the 3-(acylamino)-benzodiazepine analogue L364,718 to investigate what type of receptor (gastrin type or CCK-A type) is involved in the regulation of the H+ secretory activity of the rabbit parietal cell. Neither L365,260 nor L364,718 alone caused stimulation of [3H]inositol phosphates ([3H]InsP) production. Each analogue inhibited 125I-labeled gastrin or 125I-CCK-8 binding to parietal cells and gastrin- or CCK-8-induced [3H]InsP production and [14C]AP accumulation. In all cases, L365,260 was approximately 70-100 times more potent than L364,718 (IC50 approximately 2-4 nM for L365,260 and approximately 0.2-0.4 microM for L364,718). Nevertheless, each antagonist displayed the same potency to inhibit the effects of gastrin or CCK-8. These results demonstrate that gastrin and CCK-8 interact with the same "gastrin-type" receptor on parietal cells. Moreover, L365,260 behaves as a competitive antagonist of the action of gastrin on parietal cells. Gastrin induces a rise in the levels of inositol 1,4,5-trisphosphate [Ins(1,4,5)P3] and inositol 1,3,4,5-tetrakisphosphate [Ins(1,3,4,5)P4] within the first seconds after parietal cell stimulation. The fact that L365,260 (10 nM) totally suppressed the gastrin-induced formation of Ins(1,4,5)P3 and Ins(1,3,4,5)P4 suggests the involvement of these isomers in the mediation of acid secretion through gastrin receptor activation.


Subject(s)
Benzodiazepinones/pharmacology , Cholecystokinin/antagonists & inhibitors , Gastrins/pharmacology , Inositol Phosphates/metabolism , Parietal Cells, Gastric/metabolism , Phenylurea Compounds , Receptors, Cholecystokinin/metabolism , Sincalide/pharmacology , Aminopyrine/metabolism , Animals , Biological Transport/drug effects , Chromatography, High Pressure Liquid , Devazepide , Gastrins/metabolism , In Vitro Techniques , Inositol Phosphates/isolation & purification , Kinetics , Rabbits , Receptors, Cholecystokinin/drug effects
15.
Biochemistry ; 28(18): 7182-8, 1989 Sep 05.
Article in English | MEDLINE | ID: mdl-2819061

ABSTRACT

Synthesis, conformation, and biological properties of the Ala10 analogue of des-Trp1,Nle12-minigastrin are reported. Replacement of the Gly residue in the original sequence with Ala remarkably changes the conformational preference of the hormone in trifluoroethanol. CD and NMR results indicate that the conformational change is mainly located in the C-terminal portion of the molecule, with probable extension of the N-terminal alpha-helix throughout the entire sequence. The structural modification causes a 10-fold decrease in the biological potency of the hormone, which is about as active as the C-terminal tetrapeptide amide. These findings support our previous hypothesis that the optimal bioactive conformation of the native hormone is U-shaped, with mutual interactions among the two end segments.


Subject(s)
Gastrins , Alanine , Animals , Chemical Phenomena , Chemistry , Circular Dichroism , Gastric Acid/metabolism , Gastrins/analysis , Gastrins/biosynthesis , Gastrins/metabolism , Gastrins/pharmacology , Humans , Magnetic Resonance Spectroscopy , Male , Protein Binding , Protein Conformation , Rabbits , Rats , Temperature
16.
Biochim Biophys Acta ; 1010(3): 357-62, 1989 Mar 06.
Article in English | MEDLINE | ID: mdl-2920184

ABSTRACT

The differentiation between gastrin (HG) and cholecystokinin (CCK) receptors in gastric mucosa was examined on isolated parietal (F3) and non-parietal (F1) cells from rabbit fundic mucosa separated by elutriation. Direct binding assays on enriched cell populations were performed using 125I-labeled HG-17, 125I-labeled CCK-8 and 125I-labeled CCK-39 as probes. (1) On F1 cells, the dissociation constants (Kd) for the two labeled CCKs were nearly the same (62 pM for CCK-8 and 74 pM for CCK-39) but the binding capacity for CCK-8 was 2-times higher than for CCK-39. HG-17 also bound to this cell population, but its Kd value as about 2-times higher (110 pM) than that of CCK. The presence of two distinct classes of sites on F1 cells can be suggested from competition studies: one more specific for CCK, which bound CCK-8 and CCK-39 with the same affinity, and another class more specific for gastrin, which bound CCK-8 and HG-17 with the same affinity and CCK-39 with a low affinity. (2) On F3 cells, CCK-8 and HG-17 bound with similar affinities (Kd values 81 pM for CCK-8 and 87 pM for HG-17), but CCK-39 did not specifically bind to this cell population. The presence of a binding site more specific for HG than for CCK on F3 cells was confirmed by competition studies in which CCK-33 competed for binding with labeled HG-17 and labeled CCK-8 with a 50-times lower affinity than the other peptides.


Subject(s)
Gastric Mucosa/analysis , Receptors, Cholecystokinin/analysis , Animals , Binding Sites , Binding, Competitive , In Vitro Techniques , Rabbits
17.
C R Seances Soc Biol Fil ; 182(3): 335-43, 1988.
Article in French | MEDLINE | ID: mdl-2852529

ABSTRACT

The diterpene, forskolin, direct activator of the catalytic subunit of the adenylate cyclase from various tissues, also stimulates gastric acid secretion: in vitro, with an isolated parietal cell preparation, forskolin dose-dependently stimulated acid secretion (EC50: 1 microM) (measured by accumulation in the acidic spaces of the weak base [14C]-aminopyrine) and the maximal acid secretory value at 0.1 mM was 4 times higher than that obtained with histamine. Forskolin dramatically increased the production of intracellular cyclic-AMP at a level 4 times higher than that obtained with histamine at the same concentration. In vivo, gastric acid secretion of the rat is dose-dependently increased. The doses required to get a significant response (100 nmol/kg) were 1,000 times higher than those required for gastrin and 100 times lower than those for histamine, but the same maximal value was obtained. Cimetidine did not significantly modified this response. These results demonstrate that, both in vitro and in vivo, forskolin is a potent stimulant for gastric acid secretion.


Subject(s)
Colforsin/pharmacology , Gastric Acid/metabolism , Aminopyridines/metabolism , Animals , Cyclic AMP/analysis , Dose-Response Relationship, Drug , Gastrins/pharmacology , Histamine/pharmacology , Parietal Cells, Gastric/metabolism , Rabbits , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...