Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Type of study
Publication year range
1.
Cell Death Differ ; 16(10): 1372-84, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19521424

ABSTRACT

We have recently reported that cytostatic concentrations of the microsomal antiestrogen-binding site (AEBS) ligands, such as PBPE (N-pyrrolidino-(phenylmethyphenoxy)-ethanamine,HCl) and tamoxifen, induced differentiation characteristics in breast cancer cells through the accumulation of post-lanosterol intermediates of cholesterol biosynthesis. We show here that exposure of MCF-7 (human breast adenocarcinoma cell line) cells to higher concentrations of AEBS ligands triggered active cell death and macroautophagy. Apoptosis was characterized by Annexin V binding, chromatin condensation, DNA laddering and disruption of the mitochondrial functions. We determined that cell death was sterol- and reactive oxygen species-dependent and was prevented by the antioxidant vitamin E. Macroautophagy was characterized by the accumulation of autophagic vacuoles, an increase in the expression of Beclin-1 and the stimulation of autophagic flux. We established that macroautophagy was sterol- and Beclin-1-dependent and was associated with cell survival rather than with cytotoxicity, as blockage of macroautophagy sensitized cells to AEBS ligands. These results show that the accumulation of sterols by AEBS ligands in MCF-7 cells induces apoptosis and macroautophagy. Collectively, these data support a therapeutic potential for selective AEBS ligands in breast cancer management and shows a mechanism that explains the induction of autophagy in MCF-7 cells by tamoxifen and other selective estrogen receptor modulators.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Apoptosis , Autophagy , Breast Neoplasms/metabolism , Cholesterol/metabolism , Estrogen Receptor Modulators/pharmacology , Ethylamines/toxicity , Pyrrolidines/toxicity , Tamoxifen/pharmacology , Binding Sites , Cell Differentiation , Female , Humans , Ligands , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured , Vitamin E/pharmacology
2.
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
3.
J Biol Chem ; 275(23): 17321-7, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10748160

ABSTRACT

Among the most conserved regions in the G-protein-coupled receptors is the (N/D)PX(2-3)Y motif of the seventh transmembrane domain (X represents any amino acid). The mutation of the Asn/Asp residue of this motif in different G-protein-coupled receptors was shown to affect the activation of either adenylyl cyclase or phospholipase C. We have mutated the Asn residue (Asn-391) of the NPXXY motif in the CCKBR to Ala and determined the effects of the mutation on binding, signaling, and G-proteins coupling after expression of the mutated receptor in COS cells. The mutated receptor displayed similar expression levels and high affinity CCK binding compared with the wild type CCKBR. However, unlike the wild type CCKBR, the mutated receptor was completely unable to mediate activation of either phospholipase C and protein kinase C-dependent and -independent mitogen-activated protein kinase pathways, indicating an essential role of Asn-391 in CCKBR signaling. Coimmunoprecipitation experiments allowed us to show that the inactive mutant retains an intact capacity to form stable complexes with G(q)alpha subunits in response to CCK. These results indicate that the formation of high affinity CCK-receptor-G(q) protein complexes is not sufficient to activate G(q) and suggest that Asn-391 is specifically involved in G(q) proteins activation.


Subject(s)
Asparagine , GTP-Binding Proteins/metabolism , Receptors, Cholecystokinin/chemistry , Receptors, Cholecystokinin/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Aspartic Acid , Binding Sites , COS Cells , Cholecystokinin/pharmacology , Conserved Sequence , GTP-Binding Protein alpha Subunits, Gq-G11 , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Inositol Phosphates/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis, Site-Directed , Peptide Fragments/pharmacology , Protein Kinase C/metabolism , Rats , Receptor, Cholecystokinin B , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection , Type C Phospholipases/metabolism
4.
Protein Sci ; 8(11): 2347-54, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10595537

ABSTRACT

The knowledge of the binding sites of G protein-coupled cholecystokinin receptors represents important insights that may serve to understand their activation processes and to design or optimize ligands. Our aim was to identify the amino acid of the cholecystokinin-A receptor (CCK-AR) binding site in an interaction with the sulfate of CCK, which is crucial for CCK binding and activity. A three-dimensional model of the [CCK-AR-CCK] complex was built. In this model, Arg197 was the best candidate residue for a ionic interaction with the sulfate of CCK. Arg197 was exchanged for a methionine by site-directed mutagenesis. Wild-type and mutated CCK-AR were transiently expressed in COS-7 cells for pharmacological and functional analysis. The mutated receptor on Arg197 did not bind the agonist radioligand 125I-BH-[Thr, Nle]-CCK-9; however, it bound the nonpeptide antagonist [3H]-SR27,897 as the wild-type receptor. The mutant was approximately 1,470- and 3,200-fold less potent than the wild-type CCK-AR to activate G proteins and to induce inositol phosphate production, respectively. This is consistent with the 500-fold lower potency and 800-fold lower affinity of nonsulfated CCK relative to sulfated CCK on the wild-type receptor. These data, together with those showing that the mutated receptor failed to discriminate nonsulfated and sulfated CCK while it retained other pharmacological features of the CCK-AR, strongly support an interaction between Arg197 of the CCK-AR binding site and the sulfate of CCK. In addition, the mutated CCK-AR resembled the low affinity state of the wild-type CCK-AR, suggesting that Arg197-sulfate interaction regulates conformational changes of the CCK-AR that are required for its physiological activation.


Subject(s)
Arginine , Cholecystokinin/chemistry , Cholecystokinin/metabolism , Receptors, Cholecystokinin/chemistry , Receptors, Cholecystokinin/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , COS Cells , Computer Simulation , Humans , Indoleacetic Acids/pharmacokinetics , Inositol Phosphates/metabolism , Iodine Radioisotopes , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Radioligand Assay , Receptor, Cholecystokinin A , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thiazoles/pharmacokinetics , Transfection , Tritium
5.
J Biol Chem ; 274(33): 23191-7, 1999 Aug 13.
Article in English | MEDLINE | ID: mdl-10438490

ABSTRACT

Recently, we reported that the mutation of His(207) to Phe located in the second extracellular loop of the cholecystokinin B receptor strongly affected cholecystokinin (CCK) binding (Silvente-Poirot, S., Escrieut, C., and Wank, S. A. (1998) Mol. Pharmacol. 54, 364-371). To characterize the functional group in CCK that interacts with His(207), we first substituted His(207) to Ala. This mutation decreased the affinity and the potency of CCK to produce total inositol phosphates 302-fold and 456-fold without affecting the expression of the mutant receptor. The screening of L-alanine-modified CCK peptides to bind and activate the wild type and mutant receptors allowed the identification of the interaction of the C-terminal Asp(8) of CCK with His(207). The H207A-CCKBR mutant, unlike the wild type receptor, was insensitive to substitution of Asp(8) of CCK to other amino acid residues. This interaction was further confirmed by mutating His(207) to Asp. The affinity of CCK for the H207D-CCKBR mutant was 100-fold lower than for the H207A-CCKBR mutant, consistent with an electrostatic repulsion between the negative charges of the two interacting aspartic acids. Peptides with neutral amino acids in position eight of CCK reversed this effect and displayed a gain of affinity for the H207D mutant compared with CCK. To date, this is the first report concerning the identification of a direct contact point between the CCKB receptor and CCK.


Subject(s)
Aspartic Acid/metabolism , Cholecystokinin/metabolism , Histidine/metabolism , Receptors, Cholecystokinin/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Cholecystokinin/chemistry , Mutagenesis, Site-Directed , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/chemistry , Receptors, Cholecystokinin/genetics , Structure-Activity Relationship
6.
Mol Pharmacol ; 54(2): 364-71, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9687578

ABSTRACT

The cholecystokinin (CCK) receptor types A and B (CCKAR and CCKBR) are G protein-coupled receptors with approximately 50% amino acid identity; both have high affinity for the sulfated CCK octapeptide (CCK-8), whereas only the CCKBR has high affinity for gastrin. Previously, we identified five amino acids in the second extracellular loop (ECL) of the CCKBR that were essential for gastrin selectivity. Subsequent mutagenesis of one of these five amino acids (H207F) resulted in the loss of radiolabeled CCK-8 binding. CCK-8 stimulated total inositol phosphate accumulation in COS-1 cells transiently expressing the CCKBR-H207F with full efficacy and a 3044-fold reduced potency, which suggests that the loss of radioligand binding was caused by a loss in affinity. Alanine scanning mutagenesis was performed on the amino terminus near the top of transmembrane domain I (TMI) and on ECL1, two extracellular domains implicated in ligand binding by previous mutagenesis studies. 125I-Bolton-Hunter-CCK-8 binding to mutant receptors transiently expressed in COS-1 identified one nonconserved amino acid, R57A, at the top of TMI that caused a 21-fold reduction in CCK-8 affinity and four conserved amino acids, N115A, L116A, F120A and F122A, in the ECL1 that caused a 15.6-, 6-, 440-, and 8-fold reduction in affinity or efficacy. Alanine substitution of the equivalent amino acids in the CCKAR corresponding to each of the five amino acids in ECL1 and ECL2 affecting CCK-8 affinity for the CCKBR revealed only two mutations, L103A and F107A, that decreased CCK-8 affinity (68- and 2885-fold, respectively). These data suggest that CCK-8 interacts at multiple contact points in the extracellular domains of CCK receptors and that the CCKAR and CCKBR have distinct binding sites despite their shared high affinity for CCK-8.


Subject(s)
Receptors, Cholecystokinin/agonists , Sincalide/pharmacology , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Molecular Sequence Data , Protein Conformation , Receptors, Cholecystokinin/metabolism , Sincalide/metabolism
7.
J Biol Chem ; 273(23): 14380-6, 1998 Jun 05.
Article in English | MEDLINE | ID: mdl-9603948

ABSTRACT

Sulfation of the tyrosine at the seventh position from the C terminus of cholecystokinin (CCK) is crucial for CCK binding to the CCK-A receptor. Using three-dimensional modeling, we identified methionine 195 of the CCK-A receptor as a putative amino acid in interaction with the aromatic ring of the sulfated tyrosine of CCK. We analyzed the role played by the two partners of this interaction. The exchange of Met-195 for a leucine caused a minor decrease (2. 8-fold) on the affinity of the high affinity sites for sulfated CCK-9, a strong drop (73%) of their number, and a 30-fold decrease on the affinity of the low and very low affinity sites for sulfated CCK-9, with no change in their number. The mutation also caused a 54-fold decrease of the potency of the receptor to induce inositol phosphates production. The high affinity sites of the wild-type CCK-A receptor were highly selective (800-fold) toward sulfated versus nonsulfated CCK, whereas low and very low affinity sites were poorly selective (10- and 18-fold). In addition, the M195L mutant bound, and responded to, sulfated CCK analogues with decreased affinities and potencies, whereas it bound and responded to nonsulfated CCK identically to the wild-type receptor. Thus, Met-195 interacts with the aromatic ring of the sulfated tyrosine to correctly position the sulfated group of CCK in the binding site of the receptor. This interaction is essential for CCK-dependent transition of the CCK-A receptor to a high affinity state. Our data should represent an important step toward the identification of the residue(s) of the receptor in interaction with the sulfate moiety of CCK and the understanding of the molecular mechanisms that govern CCK-A receptor activation.


Subject(s)
Cholecystokinin/metabolism , Methionine/metabolism , Receptors, Cholecystokinin/chemistry , Tyrosine/analogs & derivatives , Animals , Binding Sites/physiology , Binding, Competitive , COS Cells , Inositol Phosphates/analysis , Ligands , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed/genetics , Protein Binding/physiology , Receptor, Cholecystokinin A , Transfection/genetics , Tyrosine/metabolism
8.
J Biol Chem ; 272(29): 18179-84, 1997 Jul 18.
Article in English | MEDLINE | ID: mdl-9218453

ABSTRACT

Internalization of a variety of different heptahelical G protein-coupled receptors has been shown to be influenced by a number of different structural determinants of the receptors, including the carboxyl terminus. To investigate the role of the carboxyl terminus of cholecystokinin (CCK) receptors in receptor internalization, the rat wild type (WT) CCK-A receptor (WT CCKAR) and the rat WT CCK-B receptor (WT CCKBR) were truncated after amino acid residue 399 (CCKAR Tr399) and 408 (CCKBR Tr408), thereby deleting the carboxyl-terminal 45 and 44 residues, respectively. All WT and mutant CCK receptors were stably expressed in NIH/3T3 cells. Internalization of the CCKAR Tr399 was not significantly different from the WT CCKAR. In contrast, internalization of the CCKBR Tr408 was decreased to 26% compared with the WT CCKBR internalization of 92%. The mutation of all 10 serine and threonine residues (as potential phosphorylation sites) in the carboxyl terminus of the CCKBR to alanines (mutant CCKBR DeltaS/T) could account for the majority of this effect (39% internalization). All mutant receptors displayed similar ligand binding characteristics, G protein coupling, and signal transduction as their respective WT receptors, indicating that the carboxyl termini are not necessary for these processes. Thus, internalization of the CCKBR, unlike that of the CCKAR, depends on the carboxyl terminus of the receptor. These results suggest that, despite the high degree of homology between CCKAR and CCKBR, the structural determinants that mediate the interaction with the endocytic pathway reside in different regions of the receptors.


Subject(s)
Receptors, Cholecystokinin/chemistry , Receptors, Cholecystokinin/metabolism , Sincalide/pharmacology , 3T3 Cells , Amino Acid Sequence , Animals , Calcium/metabolism , Fluorescent Dyes , Iodine Radioisotopes , Kinetics , Ligands , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Secondary , Radioligand Assay , Rats , Receptor, Cholecystokinin A , Receptor, Cholecystokinin B , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Rhodamines , Sequence Deletion , Sincalide/metabolism , Substrate Specificity , Transfection
9.
J Biol Chem ; 271(25): 14698-706, 1996 Jun 21.
Article in English | MEDLINE | ID: mdl-8663021

ABSTRACT

The two known receptors mediating the actions of cholecystokinin (CCK) and gastrin, CCK type A (CCKAR) and CCK type B (CCKBR) receptors, are G protein-coupled receptors having approximately 50% amino acid homology. Both the CCKAR and CCKBR have high affinity for sulfated CCK peptides, while only the CCKBR has high affinity for gastrin peptides. To determine the structural basis for the selectivity of the CCKBR for gastrin, we first constructed a series of CCKB/AR chimeras in which restriction endonuclease-defined segments of the CCKBR were replaced with the corresponding segments of the CCKAR. Chimeras transiently expressed in COS-1 cells were screened for the selective loss of gastrin affinity according to the displacement of 125I-labeled Bolton-Hunter-CCK-8 binding by gastrin-17-I and CCK-8. The sequence spanning from transmembrane domain III (TM III) to TM V was the only segment that resulted in the selective loss of gastrin affinity. This segment could account for 100 of the expected 300-fold lower affinity of gastrin-17-I observed for the control CCKAR compared to the control CCKBR. Using site-directed mutagenesis in this segment of the CCKBR, we identified a sequence of 5 amino acids in the second extracellular loop responsible for this 100-fold selective loss in gastrin affinity. 125I-labeled Bolton-Hunter-CCK-8 binding displacement by L365,260 (a CCKBR selective antagonist) was unaffected by the changes in these 5 amino acids. These results present for the first time the identification of the amino acid sequence of the CCKBR conferring the majority of the selectivity for gastrin.


Subject(s)
Gastrins/metabolism , Phenylurea Compounds , Protein Structure, Secondary , Receptors, Cholecystokinin/chemistry , Receptors, Cholecystokinin/metabolism , Amino Acid Sequence , Animals , Benzodiazepinones/pharmacology , Binding Sites , Binding, Competitive , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Gastrins/pharmacology , Humans , Indicators and Reagents , Kinetics , Models, Structural , Molecular Sequence Data , Mutagenesis, Site-Directed , Point Mutation , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/agonists , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Sincalide/analogs & derivatives , Sincalide/metabolism , Succinimides/metabolism , Transfection
11.
Eur J Biochem ; 212(2): 529-38, 1993 Mar 01.
Article in English | MEDLINE | ID: mdl-8444190

ABSTRACT

The full peptide antagonist of the pancreatic cholecystokinin (CCK) receptor, JMV 179, [Boc-Tyr(SO3H)-Ahx-Gly-dTrp-Ahx-Asp phenylethyl ester, where Tyr(SO3H) = sulfated tyrosine, Ahx = 6-aminohexanoic acid] was modified at its N-terminus by incorporation of p-hydroxyphenyl propionate (Bolton-Hunter reagent, BH) and was subsequently radioiodinated. After HPLC purification, 125I-BH-JMV-179, a CCK antagonist radioligand of high specific activity (2000 Ci/mmol) was obtained. 125I-BH-JMV-179 bound to a single population of sites on rat pancreatic plasma membranes, (Kd = 3.9 nM, Bmax = 40 pmol/mg protein). Binding was dependent on time, temperature, and protein concentration, and was fully reversible. JMV 179 radioligand detected four times as many sites as an agonist radioligand [C. Hadjiivanova, M. Dufresne, S. Poirot, P. Sozzani, N. Vaysse, L. Moroder and D. Fourmy (1992) Eur. J. Biochem. 204, 273-279]. Agonists and antagonists of the A- and B-subtype CCK/gastrin receptors inhibited 125I-BH-JMV-179 binding with an order of potency compatible with the A-subtype CCK receptor pharmacology. Moreover, the sulfate group on the tyrosine residue of the CCK peptides appeared to be of much less importance for antagonist affinity than for agonist affinity. Inhibition of 125I-BH-JMV-179 binding by agonists (except JMV 180), demonstrated the presence of two affinity classes of binding sites. The population of sites having an apparent high affinity for CCK represented 30 pmol/mg protein and threefold the number of high-affinity sites previously identified by an agonist radioligand. In presence of non-hydrolyzable GTP, all the sites bound CCK agonists with a low affinity. Moreover, saturation analysis of JMV 179 radioligand binding in the presence of CCK indicated that CCK interacted competitively with all JMV 179 sites and demonstrated binding of JMV 179 radioligand to two distinct affinity classes of sites. In the presence of GTP[S] a single affinity class of sites for JMV 179 radioligand was found as in the control experiments without CCK. This study, with the first CCK peptide antagonist radioligand, demonstrates that CCK receptors exist in two interconvertible affinity states regulated by guanine-nucleotide-binding regulatory protein(s) in rat pancreatic plasma membranes. JMV 179 radioligand does not induce receptor coupling but distinguishes the two affinity states of the CCK receptors. JMV 179 reveals the existence of populations of high-affinity and low-affinity sites for CCK which had not previously been detected by agonist radioligand binding, thus suggesting heterogeneity of CCK receptor sites in membranes.


Subject(s)
Pancreas/chemistry , Receptors, Cholecystokinin/antagonists & inhibitors , Receptors, Cholecystokinin/metabolism , Sincalide/analogs & derivatives , Amino Acid Sequence , Animals , Cholecystokinin/antagonists & inhibitors , Cholecystokinin/pharmacology , In Vitro Techniques , Molecular Sequence Data , Rats , Receptors, Cholecystokinin/analysis , Sincalide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...