Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 342(2): 318-26, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22547573

ABSTRACT

The melanocortin 1 receptor (MC1R) is a highly polymorphic G protein-coupled receptor, which is known to modulate pigmentation and inflammation. In the current study, we investigated the pharmacological effects of select single-nucleotide polymorphisms (SNPs) (V60L, R163Q, and F196L). After transient expression of MC1Rs in human embryonic kidney 293 cells, basal and ligand-induced cAMP signaling and mitogen-activated protein kinase (MAPK) activation were assessed by using luciferase reporter gene assays and Western blot analysis, respectively. All receptor variants showed decreased basal cAMP activity. With the V60L and F196L variants, the decrease in constitutive activity was attributable, at least in part, to a reduction in surface expression. The F196L variant also displayed a significant reduction in potency for both the peptide agonist α-melanocyte-stimulating hormone (α-MSH) and the small-molecule agonist 1-[1-(3-methyl-L-histidyl-O-methyl-D-tyrosyl)-4-phenyl-4-piperidinyl]-1-butanone (BMS-470539). In MAPK signaling assays, the F196L variant showed decreased phospho-extracellular signal-regulated kinase levels after stimulation with either α-MSH or BMS-470539. In contrast, the R163Q variant displayed a selective loss of α-MSH-induced MAPK activation; whereas responsiveness to the small-molecule agonist BMS-470539 was preserved. Further assessment of MC1R variants in A549 cells, an in vitro model of inflammation, revealed an enhanced inflammatory response resulting from expression of the F196L variant (versus the wild-type MC1R). This alteration in function was restored by treatment with BMS-470539. Overall, these studies illustrate novel signaling profiles linked to distinct MC1R SNPs. Furthermore, our investigations highlight the potential for small-molecule drugs to rescue the function of MC1R variants that show reduced basal and/or α-MSH stimulated activity.


Subject(s)
Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/metabolism , Cell Line, Transformed , Cyclic AMP/metabolism , HEK293 Cells , Humans , Imidazoles/pharmacology , Inflammation/genetics , Inflammation/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Polymorphism, Single Nucleotide , Signal Transduction , alpha-MSH/pharmacology
2.
Am J Physiol Gastrointest Liver Physiol ; 291(1): G156-62, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16574983

ABSTRACT

Cholecystokinin (CCK), acting at CCK1 receptors (CCK1Rs) on intestinal vagal afferent terminals, has been implicated in the control of gastrointestinal function and food intake. Using CCK1R(-/-) mice, we tested the hypothesis that lipid-induced activation of the vagal afferent pathway and intestinal feedback of gastric function is CCK1R dependent. In anesthetized CCK1R(+/+) ("wild type") mice, meal-stimulated gastric acid secretion was inhibited by intestinal lipid infusion; this was abolished in CCK1R(-/-) mice. Gastric emptying of whole egg, measured by nuclear scintigraphy in awake mice, was significantly faster in CCK1R(-/-) than CCK1R(+/+) mice. Gastric emptying of chow was significantly slowed in response to administration of CCK-8 (22 pmol) in CCK1R(+/+) but not CCK1R(-/-) mice. Activation of the vagal afferent pathway was measured by immunohistochemical localization of Fos protein in the nucleus of the solitary tract (NTS; a region where vagal afferents terminate). CCK-8 (22 pmol ip) increased neuronal Fos expression in the NTS of fasted CCK1R(+/+) mice; CCK-induced Fos expression was reduced by 97% in CCK1R(-/-) compared with CCK1R(+/+) mice. Intralipid (0.2 ml of 20% Intralipid and 0.04 g lipid), but not saline, gavage increased Fos expression in the NTS of fasted CCK1R(+/+) mice; lipid-induced Fos expression was decreased by 47% in CCK1R(-/-) compared with CCK1R(+/+)mice. We conclude that intestinal lipid activates the vagal afferent pathway, decreases gastric acid secretion, and delays gastric emptying via a CCK1R-dependent mechanism. Thus, despite a relatively normal phenotype, intestinal feedback in response to lipid is severely impaired in these mice.


Subject(s)
Gastric Emptying/physiology , Lipids/administration & dosage , Receptor, Cholecystokinin B/metabolism , Stomach/innervation , Stomach/physiology , Vagus Nerve/physiology , Animals , Feedback/drug effects , Feedback/physiology , Gastric Emptying/drug effects , Gene Silencing , Male , Mice , Mice, Knockout , Receptor, Cholecystokinin B/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Stomach/drug effects , Vagus Nerve/drug effects
3.
J Pharmacol Exp Ther ; 295(3): 1142-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11082451

ABSTRACT

The neuropeptide substance P (SP), apart from its traditional role in spinal nociceptive processing, is an important regulatory effector of opioid-dependent analgesic processes. The present study stems from our original findings indicating that 1) pharmacologically administered SP mediates a strong inhibitory activity on the development of morphine tolerance in rats, and that 2) a novel SP-opioid peptide chimera YPFFGLM-NH(2), designated ESP7, produces opioid-dependent analgesia without tolerance development. To further examine the effects of simultaneous activation of two distinct opposing spinal systems on opioid tolerance and the mechanisms underlying chimeric peptide function, a second SP-opioid chimera was synthesized. This chimera, designated ESP6 (YPFFPLM-NH(2)), contains overlapping domains of endomorphin-2 and SP, respectively. ESP6 is distinguished from ESP7 by a glycine to proline substitution at position 5. Intrathecal administration of morphine sulfate (MS) with ESP6 leads to a prolongation of MS analgesia over a 5-day period. The analgesia produced by ESP6 and MS is opioid receptor-dependent, due to the ability of naltrexone to block the analgesic response. Furthermore, when ESP6 and MS are administered with concurrent NK-1 receptor blockade, a decay in analgesic potency similar to that seen with MS alone results. The presence of a proline in ESP6 appears to reduce its conformational flexibility, limit its potency at the micro-opioid receptor, and hinder its analgesic effectiveness alone. However, ESP6 represents a novel adjuvant for the maintenance of opioid analgesia over time and provides a means to predict the pharmacological properties of a chimera from its structure.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Oligopeptides/pharmacology , Substance P/pharmacology , Animals , Drug Tolerance , Indoles/pharmacology , Isoindoles , Male , Naltrexone/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/physiology , Structure-Activity Relationship
4.
Trends Pharmacol Sci ; 21(9): 346-53, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973088

ABSTRACT

Polymorphisms in G-protein-coupled receptors can alter drug affinity and/or activity. In addition, genetic differences in amino acid sequences can induce ligand-independent signaling, which in turn can lead to disease. With growing efforts in the field of pharmacogenomics, it is anticipated that polymorphism-induced alterations in drug and/or receptor function will be a focus of increasing concern during the course of future drug-development efforts. In this review, the spectrum of pharmacological consequences that result from polymorphisms in the cholecystokinin CCK2 receptor will be discussed, thereby illustrating emerging themes in pharmacogenomics.


Subject(s)
Polymorphism, Genetic/physiology , Receptors, Cholecystokinin/physiology , Amino Acid Sequence , Animals , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Molecular Sequence Data , Polymorphism, Genetic/genetics , Receptors, Cholecystokinin/genetics , Species Specificity
5.
Mol Pharmacol ; 58(2): 399-406, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10908308

ABSTRACT

Recent efforts have focused on identifying small nonpeptide molecules that can mimic the activity of endogenous peptide hormones. Understanding the molecular basis of ligand-induced receptor activation by these divergent classes of ligands should expedite the process of drug development. Using the cholecystokinin-B/gastrin receptor (CCK-BR) as a model system, we have recently shown that both affinity and efficacy of nonpeptide ligands are markedly affected by amino acid alterations within a putative transmembrane domain (TMD) ligand pocket. In this report, we examine whether residues projecting into the TMD pocket determine the pharmacologic properties of structurally diverse CCK-BR ligands, including peptides and synthetic peptide-derived partial agonists (peptoids). Nineteen mutant human CCK-BRs, each including a single TMD amino acid substitution, were transiently expressed in COS-7 cells and characterized. Binding affinities as well as ligand-induced inositol phosphate production at the mutant CCK-BRs were assessed for peptides (CCK-8 and CCK-4) and for peptoids (PD-135,158 and PD-136, 450). Distinct as well as overlapping determinants of peptide and peptoid binding affinity were identified, supporting that both classes of ligands, at least in part, interact with the CCK-BR TMD ligand pocket. Eight point mutations resulted in marked increases or decreases in the functional activity of the synthetic peptoid ligands. In contrast, the functional activity of both peptides, CCK-8 and CCK-4, was not affected by any of the CCK-BR mutations. These findings suggest that the mechanisms underlying activation of G-protein-coupled receptors by endogenous peptide hormones versus synthetic ligands may markedly differ.


Subject(s)
Meglumine/analogs & derivatives , Receptors, Cholecystokinin/metabolism , Amino Acid Sequence , Animals , Binding, Competitive , COS Cells , Humans , Indoles/pharmacology , Meglumine/pharmacology , Membrane Proteins/chemistry , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Sequence Data , Mutation , Peptides/metabolism , Peptoids , Radioligand Assay , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/chemistry , Receptors, Cholecystokinin/drug effects , Receptors, Cholecystokinin/genetics , Sequence Homology, Amino Acid
6.
Proc Natl Acad Sci U S A ; 97(13): 7621-6, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10852965

ABSTRACT

To elucidate mechanisms of acute and chronic pain, it is important to understand how spinal excitatory systems influence opioid analgesia. The tachykinin substance P (SP) represents the prototypic spinal excitatory peptide neurotransmitter/neuromodulator, acting in concert with endogenous opioid systems to regulate analgesic responses to nociceptive stimuli. We have synthesized and pharmacologically characterized a chimeric peptide containing overlapping NH(2)- and COOH-terminal functional domains of the endogenous opioid endomorphin-2 (EM-2) and the tachykinin SP, respectively. Repeated administration of the chimeric molecule YPFFGLM-NH(2), designated ESP7, into the rat spinal cord produces opioid-dependent analgesia without loss of potency over 5 days. In contrast, repeated administration of ESP7 with concurrent SP receptor (SPR) blockade results in a progressive loss of analgesic potency, consistent with the development of tolerance. Furthermore, tolerant animals completely regain opioid sensitivity after post hoc administration of ESP7 alone, suggesting that coactivation of SPRs is essential to maintaining opioid responsiveness. Radioligand binding and signaling assays, using recombinant receptors, confirm that ESP7 can coactivate mu-opioid receptors (MOR) and SPRs in vitro. We hypothesize that coincidental activation of the MOR- and SPR-expressing systems in the spinal cord mimics an ongoing state of reciprocal excitation and inhibition, which is normally encountered in nociceptive processing. Due to the ability of ESP7 to interact with both MOR and SPRs, it represents a unique prototypic, anti-tolerance-forming analgesic with future therapeutic potential.


Subject(s)
Analgesics/pharmacology , Oligopeptides/pharmacology , Pain/prevention & control , Recombinant Fusion Proteins/pharmacology , Substance P/pharmacology , Animals , Drug Design , Male , Oligopeptides/genetics , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Substance P/genetics
7.
J Biol Chem ; 275(25): 19115-20, 2000 Jun 23.
Article in English | MEDLINE | ID: mdl-10748042

ABSTRACT

The cholecystokinin (CCK) analog JMV-180 acts as a partial agonist in rats and a full agonist in mice. Whether this functional variability is due to species differences in CCK receptor structure or to alterations in the cellular environment is unknown. To address this question, an adenoviral construct encoding the rat CCK(A) receptor (AdCCK(A)R) was used to express the rat receptor in acini from CCK(A) receptor-deficient mice (CCK(A)R -/-). Infection of CCK(A)R -/- acini in vitro with pAdCCK(A)R led to a time-dependent increase in (125)I-CCK(8) binding. The affinity for JMV-180 of the adenovirally transferred rat and the endogenous mouse CCK(A) receptors was not different. In native mouse acini, JMV-180 acted as a full agonist (both stimulation and inhibition of amylase release). In contrast, in mouse acini expressing pAdCCK(A)R JMV-180 acted as a partial agonist (only stimulation of amylase release). In addition, the pattern of protein synthesis induced by JMV-180 in CCK(A)R -/- mouse acini infected with AdCCK(A)R resembled the pattern observed in wild-type rats (lack of inhibition) rather than the respective pattern in wild-type mice (inhibition). These data suggest that species differences in the CCK(A) receptor of rats and mice account for the observed divergence in the acinar cell response to JMV-180.


Subject(s)
Receptors, Cholecystokinin/agonists , Sincalide/analogs & derivatives , Adenoviridae/genetics , Amino Acid Sequence , Animals , Male , Mice , Molecular Sequence Data , Protein Binding , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A , Receptors, Cholecystokinin/chemistry , Receptors, Cholecystokinin/genetics , Receptors, Cholecystokinin/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Sincalide/pharmacology , Species Specificity
8.
J Clin Invest ; 103(3): 383-91, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9927499

ABSTRACT

Food intake and body weight are determined by a complex interaction of regulatory pathways. To elucidate the contribution of the endogenous peptide cholecystokinin, mice lacking functional cholecystokinin-A receptors were generated by targeted gene disruption. To explore the role of the cholecystokinin-A receptor in mediating satiety, food intake of cholecystokinin-A receptor-/- mice was compared with the corresponding intakes of wild-type animals and mice lacking the other known cholecystokinin receptor subtype, cholecystokinin-B/gastrin. Intraperitoneal administration of cholecystokinin failed to decrease food intake in mice lacking cholecystokinin-A receptors. In contrast, cholecystokinin diminished food intake by up to 90% in wild-type and cholecystokinin-B/gastrin receptor-/- mice. Together, these findings indicate that cholecystokinin-induced inhibition of food intake is mediated by the cholecystokinin-A receptor. To explore the long-term consequences of either cholecystokinin-A or cholecystokinin-B/gastrin receptor absence, body weight as a function of age was compared between freely fed wild-type and mutant animals. Both cholecystokinin-A and cholecystokinin-B/gastrin receptor-/- mice maintained normal body weight well into adult life. In addition, each of the two receptor-/- strains had normal pancreatic morphology and were normoglycemic. Our results suggest that although cholecystokinin plays a role in the short-term inhibition of food intake, this pathway is not essential for the long-term maintenance of body weight.


Subject(s)
Body Weight/physiology , Eating/physiology , Receptors, Cholecystokinin/physiology , Animals , Cholecystokinin/physiology , Female , Gastrins/physiology , Male , Mice , Mice, Knockout , Receptor, Cholecystokinin A
9.
Mol Pharmacol ; 54(5): 857-63, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9804620

ABSTRACT

We have reported previously that the transmembrane domains of the cholecystokinin-B/gastrin receptor (CCK-BR) comprise a putative ligand binding pocket. In the present study, we examined whether amino acid substitutions within the CCK-BR pocket altered the affinities and/or functional activities of L-365,260 (the prototypical nonpeptide CCK-BR antagonist) and two structural derivatives, YM022 (a higher affinity antagonist) and L-740,093S (a partial agonist). Eight amino acids that project into the CCK-BR pocket were individually replaced by alanine, using site-directed mutagenesis. Affinities for the nonpeptide molecules, as well as ligand-induced inositol phosphate production, were assessed with the wild-type and mutant receptors. For each of the nonpeptide ligands examined, a distinct series of mutations altered the affinity, suggesting that each ligand possessed a characteristic pattern of interactions within the CCK-BR pocket. Basal signaling levels and inositol phosphate formation induced by the full agonist CCK octapeptide were comparable for the wild-type receptor and all of the mutant CCK-BR forms. In contrast to the peptide agonist CCK octapeptide, the functional activities of the nonpeptide molecules were selectively altered by single point mutations within the CCK-BR pocket, resulting in interconversion of agonists and antagonists. These findings suggest that interactions between nonpeptide molecules and transmembrane domain amino acids of the CCK-BR can determine the functional activity and affinity of the ligands.


Subject(s)
Mutation , Receptors, Cholecystokinin/drug effects , Receptors, Cholecystokinin/genetics , Animals , Benzodiazepines/metabolism , Benzodiazepines/pharmacology , Benzodiazepinones/metabolism , Benzodiazepinones/pharmacology , Binding Sites , COS Cells/metabolism , COS Cells/physiology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/ultrastructure , DNA, Complementary/genetics , DNA, Complementary/metabolism , Hormone Antagonists/metabolism , Hormone Antagonists/pharmacology , Humans , Inositol Phosphates/biosynthesis , Iodine Radioisotopes , Ligands , Mutagenesis, Site-Directed , Phenylurea Compounds/metabolism , Phenylurea Compounds/pharmacology , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/metabolism , Sincalide/metabolism
10.
J Biol Chem ; 273(44): 28779-84, 1998 Oct 30.
Article in English | MEDLINE | ID: mdl-9786876

ABSTRACT

The enteroendocrine hormone, gastrin, exerts trophic effects on the gastric mucosa through the CCK-B/gastrin receptor (CCK-BR). To varying degrees in different species, excess circulating gastrin leads to proliferation of enterochromaffin-like cells and to the development of gastric carcinoid tumors. The African rodent, Mastomys natalensis, is distinguished from other mammals by its propensity toward CCK-BR-mediated growth even in the absence of hypergastrinemia. Here, we report that the Mastomys CCK-BR, when expressed in COS-7 cells, differs from the respective human, canine, and rat receptor homologs by its ability to trigger ligand-independent (i.e., constitutive) inositol phosphate formation. To define the molecular basis of this observation, a series of Mastomys-human chimeric receptors was investigated. Functional characterization of these constructs revealed that a limited segment of the Mastomys CCK-BR, transmembrane domain VI through the C-terminal end, is sufficient to confer constitutive activity to the human protein. Mutagenesis studies within this CCK-BR region defined a combination of three Mastomys amino acids that, when introduced into the human receptor, together conferred a level of ligand-independent signaling comparable with the Mastomys CCK-BR. Complementing prior observations that single point mutations can lead to ligand-independent signaling, our findings suggest that multiple naturally occurring amino acid polymorphisms and/or mutations may together result in an enhanced basal level of receptor activity.


Subject(s)
Polymorphism, Genetic , Receptors, Cholecystokinin/metabolism , Animals , COS Cells , Dogs , Humans , Muridae , Rats , Receptors, Cholecystokinin/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Species Specificity
11.
J Biol Chem ; 273(23): 14146-51, 1998 Jun 05.
Article in English | MEDLINE | ID: mdl-9603914

ABSTRACT

The development of non-peptide agonists for peptide hormone receptors would markedly expand the treatment options for a large number of diseases. However, difficulty in identifying non-peptide molecules which possess intrinsic activity has been a major obstacle in achieving this goal. At present, most of the known non-peptide ligands for peptide hormone receptors appear in standard functional assays to be antagonists. Here, we report that a constitutively active mutant of the human cholecystokinin-B/gastrin receptor, Leu325 --> Glu, offers the potential to detect even trace agonist activity of ligands which, at the wild type receptor isoform, appear to lack efficacy. The enhanced functional sensitivity of the mutant receptor enabled us to detect intrinsic activity of L-365,260, an established non-peptide antagonist for the cholecystokinin-B/gastrin receptor. Extending from this observation, we were able to demonstrate that minor structural modifications could convert L-365, 260 into either: (i) an agonist or (ii) an inverse agonist (attenuates ligand-independent signaling). The ability to confer functional activity to small non-peptide ligands suggests that the properties of endogenous peptide hormones can be mimicked, and even extended, by considerably less complex molecules.


Subject(s)
Receptors, Cholecystokinin/genetics , Animals , Benzodiazepines/pharmacology , Benzodiazepinones/chemistry , Benzodiazepinones/pharmacology , Binding, Competitive , COS Cells , Hormone Antagonists/pharmacology , Inositol Phosphates/metabolism , Molecular Structure , Mutation/genetics , Peptides/metabolism , Phenylurea Compounds/pharmacology , Receptors, Cholecystokinin/agonists , Receptors, Cholecystokinin/antagonists & inhibitors , Signal Transduction/physiology , Transfection/genetics
12.
Yale J Biol Med ; 71(3-4): 337-46, 1998.
Article in English | MEDLINE | ID: mdl-10461364

ABSTRACT

The gastric cholecystokinin-B/gastrin receptor (CCK-BR) is a key regulator of enterochromaffin-like cell function and proliferation. Over the last decade, a number of small non-peptide CCK-BR "antagonists" have been discovered. Here, we demonstrate that some of these non-peptide ligands in fact possess significant ability to activate the human CCK-BR, and are, therefore, more properly categorized as partial agonists. When tested in COS-7 cells transiently expressing the recombinant human CCK-BR, saturating concentrations of the small "peptoid" ligands PD 135,158 and PD 136,450 stimulated inositol phosphate formation to 23 and 43 percent, respectively, of the maximum response induced by a considerably larger endogenous peptide agonist, cholecystokinin octapeptide. In contrast, the benzodiazepine-derived CCK-BR ligand, YM022, acted as a "true" high-affinity antagonist of cholecystokinin-induced inositol phosphate formation (pA2 = 9.69). Consistent with recent findings in animal experiments, our data reveal that small synthetic ligands have the potential to function as either CCK-BR agonists or antagonists. These dual properties of synthetic molecules must be considered when evaluating candidate drugs for human disease.


Subject(s)
Benzodiazepinones/pharmacology , Devazepide/pharmacology , Hormone Antagonists/pharmacology , Indoles/pharmacology , Inositol Phosphates/metabolism , Meglumine/analogs & derivatives , Receptors, Cholecystokinin/drug effects , Receptors, Cholecystokinin/metabolism , Animals , Benzodiazepines/pharmacology , COS Cells/drug effects , COS Cells/metabolism , Calcium/metabolism , Dose-Response Relationship, Drug , Humans , Indoles/metabolism , Ligands , Meglumine/metabolism , Meglumine/pharmacology , Molecular Mimicry , Peptoids , Phenethylamines/metabolism , Phenethylamines/pharmacology , Phenylurea Compounds/pharmacology , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
13.
Yale J Biol Med ; 71(3-4): 347-54, 1998.
Article in English | MEDLINE | ID: mdl-10461365

ABSTRACT

Mice lacking the cholecystokinin (CCK)-B/gastrin receptor have been generated by targeted gene disruption. The roles of this receptor in controlling gastric acid secretion and gastric mucosal growth have been assessed. The analysis of homozygous mutant mice vs. wild type included measurement of basal gastric pH, plasma gastrin concentrations as well as quantification of gastric mucosal cell types by immunohistochemistry. Mutant mice exhibited a marked increase in basal gastric pH (from 3.2 to 5.2) and about a 10-fold elevation in circulating carboxyamidated gastrin compared with wild-type controls. Histologic analysis revealed a decrease in both parietal and enterochromaffin-like (ECL) cells, thus explaining the reduction in acid output. Consistent with the elevation in circulating gastrin, antral gastrin cells were increased in number while somatostatin cells were decreased. These data support the importance of the CCK-B/gastrin receptor in maintaining the normal cellular composition and function of the gastric mucosa.


Subject(s)
Gastric Mucosa/metabolism , Receptors, Cholecystokinin/deficiency , Receptors, Cholecystokinin/genetics , Stomach/pathology , Animals , Enterochromaffin-like Cells/pathology , Female , Gastrins/metabolism , H(+)-K(+)-Exchanging ATPase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Mutant Strains , Somatostatin/metabolism
14.
Proc Natl Acad Sci U S A ; 94(20): 11043-8, 1997 Sep 30.
Article in English | MEDLINE | ID: mdl-9380756

ABSTRACT

The brain cholecystokinin-B/gastrin receptor (CCK-BR) is a major target for drug development because of its postulated role in modulating anxiety, memory, and the perception of pain. Drug discovery efforts have resulted in the identification of small synthetic molecules that can selectively activate this receptor subtype. These drugs include the peptide-derived compound PD135,158 as well as the nonpeptide benzodiazepine-based ligand, L-740,093 (S enantiomer). We now report that the maximal level of receptor-mediated second messenger signaling that can be achieved by these compounds (drug efficacy) markedly differs among species homologs of the CCK-BR. Further analysis reveals that the observed differences in drug efficacy are in large part explained by single or double aliphatic amino acid substitutions between respective species homologs. This interspecies variability in ligand efficacy introduces the possibility of species differences in receptor-mediated function, an important consideration when selecting animal models for preclinical drug testing. The finding that even single amino acid substitutions can significantly affect drug efficacy prompted us to examine ligand-induced signaling by a known naturally occurring human CCK-BR variant (glutamic acid replaced by lysine in position 288; 288E --> K). When examined using the 288E --> K receptor, the efficacies of both PD135,158 and L-740, 093 (S) were markedly increased compared with values obtained with the wild-type human protein. These observations suggest that functional variability resulting from human receptor polymorphisms may contribute to interindividual differences in drug effects.


Subject(s)
Benzodiazepinones/pharmacology , Indoles/pharmacology , Meglumine/analogs & derivatives , Phenylurea Compounds/pharmacology , Polymorphism, Genetic , Receptors, Cholecystokinin/genetics , Animals , COS Cells , Dogs , Enzyme Activation , Humans , Meglumine/pharmacology , Mice , Radioligand Assay , Receptors, Cholecystokinin/agonists , Receptors, Cholecystokinin/antagonists & inhibitors , Recombinant Proteins/genetics , Species Specificity , Type C Phospholipases/metabolism
15.
Gastroenterology ; 113(3): 1015-25, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9287997

ABSTRACT

BACKGROUND & AIMS: Gastrin is a peptide hormone important in the regulation of both acid secretion and differentiation of oxyntic mucosal cells of the stomach. To further elucidate the role of gastrin in the growth and development of the gastrointestinal tract, we have generated mice that are deficient in gastrin. METHODS: Gastrin-deficient mice were generated through targeted gene disruption. Gastric and colonic architecture were determined by routine histology and immunohistochemical techniques. Proliferation was assessed by 5-bromo-2'-deoxyuridine incorporation. RESULTS: Targeted disruption of the gastrin gene resulted in mice incapable of expressing gastrin messenger RNA (mRNA) or producing gastrin peptide. This deficiency led to a marked change in gastric architecture, with a decrease in number of parietal and enterochromaffin-like cells and an increase in number of mucous neck cells. There was no difference in the proliferation labeling index of the stomach in gastrin-deficient mice (3.04% +/- 0.33%) compared with wild-type littermates (3.15% +/- 0.18%). The colon of gastrin-deficient mice seemed normal histologically, although there was a decreased proliferation labeling index (2.97% +/- 0.52%) compared with wild-type littermates (4.71% +/- 0.44%; P < 0.01). CONCLUSIONS: Gastrin is important in regulating the differentiation of the gastric mucosa and is a trophic factor for the colonic mucosa.


Subject(s)
Colon/pathology , Gastrins/deficiency , Stomach/growth & development , Stomach/pathology , Animals , Atrophy , Bromodeoxyuridine/metabolism , Cell Division , Gastric Acid/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastrins/genetics , Gastrins/metabolism , Gene Targeting , Homozygote , Immunohistochemistry , Intestinal Mucosa/pathology , Mice , Parietal Cells, Gastric/pathology
16.
Gastroenterology ; 112(1): 280-6, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8978369

ABSTRACT

BACKGROUND & AIMS: The cholecystokinin (CCK)-B/gastrin receptor is one of several regulators of gastric acid secretion and mucosal growth. To elucidate the contribution of this receptor relative to other trophic and secretory factors, mice that lack the CCK-B/gastrin receptor have been generated and studied. METHODS: Both alleles of the CCK-B/gastrin receptor were inactivated by targeted gene disruption. Analysis of the mice included measurement of basal gastric pH and plasma gastrin levels. In addition, multiple gastric mucosal cell types were identified by immunostaining and quantified. RESULTS: Homozygous mutant mice were viable, fertile, and appeared grossly normal into adulthood. The receptor-deficient mice exhibited a marked increase in basal gastric pH (from 3.2 to 5.2) and an approximately 10-fold elevation in plasma gastrin concentration compared with wild-type controls. In the stomach of mutant animals, parietal and enterochromaffin-like cells were decreased, providing a likely explanation for the reduction in acid output. In the antrum, a decrease in somatostatin cell density and an increase in the gastrin cell number were observed, consistent with the concomitant elevation in circulating gastrin. CONCLUSIONS: Together, these findings demonstrate the importance of the CCK-B/gastrin receptor in maintaining the normal cellular composition and function of the gastric mucosa.


Subject(s)
Gastric Acid/metabolism , Gastric Mucosa/pathology , Receptors, Cholecystokinin/deficiency , Animals , Cell Division , Gastrins/blood , Genetic Vectors , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Parietal Cells, Gastric/pathology , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/genetics , Receptors, Cholecystokinin/physiology
17.
Mol Pharmacol ; 50(2): 436-41, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8700154

ABSTRACT

A hallmark of the mammalian brain cholecystokinin (CCK) receptor, CCK-B/gastrin (CCK-BR), is its high affinity for two structurally related peptides, CCK and gastrin. Previous radioligand binding experiments suggested that the predominant CCK receptor from Xenopus laevis brain shares high affinity for sulfated cholecystokinin octapeptide but has > or = 1000-fold lower affinity for gastrin. To determine the molecular basis for this pharmacological divergence between mammalian and lower vertebrate receptors, we isolated a cDNA encoding the X. laevis brain CCK receptor (CCK-XLR). CCK-XLR shares approximately 50% homology at the amino acid level with both the human CCK-BR and the peripheral CCK-A receptor subtypes. The recombinant X. laevis receptor has a distinct pharmacological profile of agonist and antagonist affinities and as such offers a useful tool for structure-function studies. We used CCK-XLR to map the human CCK-BR domains that confer high affinity for gastrin. A series of chimeric CCK-BR/CCK-XLR constructs was generated and pharmacologically characterized. While maintaining wild-type affinity for sulfated cholecystokinin octapeptide, receptors with increasing amino-terminal contributions from CCK-BR demonstrated a stepwise increase in gastrin affinity. Further dissection of the amino-terminal third of the human receptor, a domain that confers a > 250-fold increase in gastrin affinity, revealed the importance of interactions among at least three subdomains. Additional structural requirements for gastrin affinity mapped to a segment spanning transmembrane domains IV and V.


Subject(s)
Gastrins/metabolism , Receptors, Cholecystokinin/chemistry , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Humans , Molecular Sequence Data , Receptors, Cholecystokinin/metabolism , Recombinant Fusion Proteins/chemistry , Xenopus laevis
18.
J Biol Chem ; 270(10): 5019-23, 1995 Mar 10.
Article in English | MEDLINE | ID: mdl-7890609

ABSTRACT

We have examined the role of transmembrane domain amino acids in conferring subtype-selective ligand affinity to the human cholecystokinin-B (CCK-B)/gastrin receptor. Fifty-eight residues were sequentially replaced by the corresponding amino acids from the pharmacologically distinct CCK-A receptor subtype. 125I-CCK-8 competition binding experiments were performed to compare all mutant CCK-B/gastrin receptor constructs with the wild type control. Affinities for the nonselective agonist, CCK-8, as well as the subtype-selective peptide (gastrin), peptide-derived (PD135,158), and nonpeptide (L365,260) and L364,718) ligands were assessed. All of the mutants retained relatively high affinity for CCK-8, suggesting that the tertiary structure of these receptors was well maintained. Only eight of the amino acid substitutions had a significant effect on subtype selective binding. When compared with the wild type, single point mutations in the CCK-B/gastrin receptor decreased affinity for gastrin, L365,260, and PD135,158 up to 17-,23-, and 61-fold, respectively. In contrast, the affinity for L364,718 increased up to 63-fold. None of the single amino acid substitutions, however, was sufficient to fully account for the subtype selectivity of any tested compound. Rather, CCK-B/gastrin receptor affinity appears to be influenced by multiple residues acting in concert. The 8 pharmacologically important amino acids cluster in the portion of the transmembrane domains adjacent to the cell surface. The spatial orientation of these residues was analyzed with a rhodopsin-based three-dimensional model of G-protein coupled receptor structure (Baldwin, J.M. (1993) EMBO J. 12, 1693-1703). This model predicts that the 8 crucial residues project into a putative ligand pocket, similar to the one which is well established for biogenic amine receptors (Caron, M. G., and Lefkowitz, R.J. (1993) Recent Prog. Horm. Res. 48, 277-290; Strader, C.D., Sigal, I.S., and Dixon, R.A. (1989) Trends Pharmacol. Sci. 10, Dec. Suppl., 26-30).


Subject(s)
Receptors, Cholecystokinin/metabolism , Amino Acid Sequence , Cell Membrane , Humans , Ligands , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Receptors, Cholecystokinin/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity
19.
J Biol Chem ; 269(42): 26121-6, 1994 Oct 21.
Article in English | MEDLINE | ID: mdl-7929324

ABSTRACT

We transfected COS cells with cDNA for rat cholecystokinin-A (CCK-A) and different CCK-B receptors and measured binding of 125I-CCK-8, [3H]L-364,718 and [3H]L-365,260 to characterize the different affinity states for each type of CCK receptor. Rat CCK-A and CCK-B receptors, canine CCK-B receptors and canine mutant CCK-B (M-CCK-B) receptors in which the leucine in position 355 was replaced by valine each existed in three different affinity states for CCK-8, high affinity, low affinity, and very low affinity. In rat CCK-A and probably CCK-B receptors, most were in the very low affinity state, whereas with canine CCK-B and M-CCK-B receptors, most were in the low affinity state. Studies with CCK receptor agonists, CCK-8, gastrin, and CCK-JMV-180, in conjunction with CCK receptor antagonists, L-364,718 and L-365,260, showed a different pattern of affinities for these ligands at the different CCK receptors. Thus, each transfected CCK receptor can exist in three different affinity states for CCK-8 and has a characteristic pattern of interaction with different ligands. This ability to exist in multiple affinity states is an intrinsic property of the CCK receptor molecule itself.


Subject(s)
Phenylurea Compounds , Receptors, Cholecystokinin/metabolism , Animals , Benzodiazepinones/metabolism , Cells, Cultured , Devazepide , Rats , Receptors, Cholecystokinin/chemistry , Sincalide/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...