Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
1.
Neurogastroenterol Motil ; 22(2): 204-9, e66, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19754922

ABSTRACT

BACKGROUND: Somatostatin inhibits gall bladder contraction. Impaired gall bladder emptying is associated with gall bladder stone formation. The incidence of cholecystolithiasis is high in patients treated with a somatostatin agonist octreotide, which predominantly interacts with somatostatin receptor subtype 2 (SSTR2). Therefore, it is believed that SSTR2 regulates gall bladder contraction; however, evidence has not been provided. Here, we evaluate the effects of SSTR1-SSTR5-selective agonists on egg yolk-induced gall bladder contraction in mice. METHODS: Homozygous deletion of SSTR2 and SSTR5 was generated by cross-mating of SSTR2(-/-) with SSTR5(-/-) mice. Mice of different genotypes were injected with SSTR1-5-selective agonists or octreotide 15 min before induction of gall bladder emptying by egg yolk. One hour later, gall bladders were removed and weighed. KEY RESULTS: Egg yolk-reduced gall bladder weights in all mice, irrespective of their genotype. Octreotide was the most potent inhibitor of gall bladder emptying in wild-type mice. In contrast, agonists with high selectivity for SSTR2 or SSTR5 inhibited gall bladder emptying by approximately 50-60%, whereas SSTR1-, SSTR3- and SSTR4-selective agonists failed to influence gall bladder contraction. In SSTR2(-/-) mice, octreotide and an SSTR5-selective agonist inhibited gall bladder emptying by approximately 50%, whereas SSTR2-selective agonists were inactive. Octreotide inhibited gall bladder emptying in SSTR5(-/-) mice by approximately 50%, without any effect in SSTR2(-/-)/SSTR5(-/-) mice. CONCLUSIONS & INFERENCES: Our study provides evidence for the role of SSTR2 and SSTR5 in regulating gall bladder emptying in mice.


Subject(s)
Gallbladder Emptying/physiology , Gallbladder/metabolism , Receptors, Somatostatin/metabolism , Analysis of Variance , Animals , Body Weight/genetics , Egg Yolk , Gallbladder/drug effects , Gallbladder Emptying/drug effects , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Knockout , Muscle Contraction/drug effects , Muscle Contraction/physiology , Octreotide/pharmacology , Proteins/metabolism , Proteinuria/metabolism , Receptors, Somatostatin/genetics , Somatostatin/metabolism
2.
Bioorg Med Chem Lett ; 16(13): 3489-94, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16632357

ABSTRACT

Synthesis and derivatization of a series of substituted tetrahydrofluorenone analogs giving potent, ERbeta subtype selective ligands are described. Several analogs possessing ERbeta binding affinities comparable to 17beta-estradiol but with greater than 75-fold selectivity over ERalpha are reported.


Subject(s)
Estrogen Receptor beta/drug effects , Fluorenes/chemical synthesis , Fluorenes/pharmacology , Cell Line , Crystallography, X-Ray , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/drug effects , Estrogen Receptor beta/chemistry , Fluorenes/classification , Humans , Ligands , Models, Molecular , Molecular Structure , Stereoisomerism , Structure-Activity Relationship
3.
J Mol Endocrinol ; 32(3): 987-95, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15171727

ABSTRACT

The effects of estrogen receptor (ER) ligands on the stability and transcriptional activity of ERbeta in the breast cancer cell lines MCF-7 and HeLa were examined. We found that ERbeta was degraded in the presence of 17beta-estradiol. Tamoxifen and Faslodex (ICI 182,780) prevented ERbeta receptor destabilization. In contrast to ERalpha, ERbeta degradation was not abolished by inhibitors of the proteasome-mediated protein degradation pathway. Furthermore, single point mutations in helix 12 of the receptor dramatically affected the stability and subsequent transcriptional activation of ERbeta.


Subject(s)
Acetylcysteine/analogs & derivatives , Breast Neoplasms/metabolism , Estradiol/analogs & derivatives , Estradiol/metabolism , Estrogen Antagonists/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Acetylcysteine/metabolism , Animals , Cell Line, Tumor , Cysteine Proteinase Inhibitors/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/chemistry , Estrogen Receptor beta/genetics , Female , Fulvestrant , Gene Expression Regulation , Genes, Reporter , Humans , Ligands , Point Mutation , Tamoxifen/metabolism
4.
J Med Chem ; 44(6): 917-22, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11300873

ABSTRACT

Extensive development of the structure-activity relationships of a screening lead determined three important pharmacophores for gonadotropin-releasing hormone (GnRH) receptor antagonist activity. Incorporation of the 3,4,5-trimethylphenyl group at the 3-position, 2-(2(S)-azetidinyl)ethoxy group at the 4-position, and N-4-pyrimidinylcarboxamide at the 6-position of the quinolone core resulted in the identification of 4-(2-(azetidin-2(S)-yl)ethoxy)-7-chloro-2-oxo-3-(3,4,5-trimethylphenyl)-1,2-dihydroquinoline-6-carboxylic acid pyrimidin-4-ylamide (1) as a potent antagonist of the GnRH receptor. A 10(4)-fold increase in in vitro binding affinity is observed for the GnRH receptor as compared to the initial screening lead. Compound 1 exhibits nanomolar binding activity and functional antagonism at the human receptor and is 7-fold less active at the rhesus receptor. Intravenous administration of compound 1 to rhesus monkeys results in a significant decrease of the serum levels of downstream hormones, luteinizing hormone (79% decrease in area under the curve) and testosterone (92% decrease in area under the curve), at a dose of 3 mg/kg. Quinolone 1 is a potent nonpeptidyl antagonist for the human GnRH receptor that is efficacious for the suppression of luteinizing hormone and testosterone in primates.


Subject(s)
Azetidines/chemical synthesis , Quinolones/chemical synthesis , Receptors, LHRH/antagonists & inhibitors , Animals , Azetidines/chemistry , Azetidines/pharmacokinetics , Azetidines/pharmacology , Binding, Competitive , CHO Cells , Cricetinae , Humans , In Vitro Techniques , Macaca mulatta , Pituitary Gland/metabolism , Quinolones/chemistry , Quinolones/pharmacokinetics , Quinolones/pharmacology , Radioligand Assay , Rats , Structure-Activity Relationship
5.
Bioorg Med Chem Lett ; 11(3): 415-7, 2001 Feb 12.
Article in English | MEDLINE | ID: mdl-11212124

ABSTRACT

N-Substituted nipecotic and iso-nipecotic amides of beta-methylTrpLys tert-butyl ester were found to be novel, selective and potent agonists of the somatostatin subtype-2 receptor in vitro. For example iso-nipecotic amide 8a showed high hsst2 binding affinity (Ki = 0.5 nM) and good selectivity (h5/h2 = 832).


Subject(s)
Nipecotic Acids/metabolism , Receptors, Somatostatin/agonists , Animals , Combinatorial Chemistry Techniques , Humans , Isomerism , Nipecotic Acids/chemical synthesis , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Protein Binding , Receptors, Somatostatin/metabolism , Structure-Activity Relationship
6.
J Physiol Paris ; 94(3-4): 211-5, 2000.
Article in English | MEDLINE | ID: mdl-11087999

ABSTRACT

High affinity, subtype selective non-peptide agonists of somatostatin receptor subtypes 1-5 were identified in combinatorial libraries constructed based on molecular modeling of known peptide agonists. Simultaneous traditional chemical synthesis yielded an additional series of somatostatin subtype-2 receptor (SSTR2) selective agonists. These compounds have been used to further define the physiological functions of the individual somatostatin receptor subtypes. In vitro experiments demonstrated the role of the SSTR2 in inhibition of glucagon release from mouse pancreatic alpha-cells and the somatostatin subtype-5 receptor (SSTR5) as a mediator of insulin secretion from pancreatic beta-cells. Both SSTR2 and SSTR5 regulated growth hormone release from the rat anterior pituitary gland. In vivo studies performed with SSTR2 receptor selective compounds demonstrated effective inhibition of pulsatile growth hormone release in rats. The SSTR2 selective compounds also lowered plasma glucose levels in normal and diabetic animal models. The availability of high affinity, subtype selective non-peptide agonists for each of the somatostatin receptors provides a direct approach to defining their physiological function both peripherally and in the central nervous system.


Subject(s)
Amides/chemistry , Indoles/chemistry , Naphthalenes/chemistry , Nitrobenzenes/chemistry , Pyridines/chemistry , Receptors, Somatostatin/agonists , Animals , Benzimidazoles/chemistry , CHO Cells , Cloning, Molecular , Combinatorial Chemistry Techniques , Cricetinae , Glucagon/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Membrane Proteins , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/genetics , Somatostatin/agonists
7.
J Pharmacol Exp Ther ; 295(3): 1051-60, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11082440

ABSTRACT

Twenty-five avermectin analogs were assessed in a mouse seizure model. The ED(50) against pentylenetetrazole-induced tonic seizures ranged from 0.48 mg/kg (L-676,893) to >160 mg/kg (L-685,869) cf. 0. 26 mg/kg for diazepam. Although avermectins are without acute toxic effects, they have been historically shown to have relative low LD(50) values in mammals. The mechanisms involved in the anticonvulsant effect and the toxicity were investigated. A series of avermectin analogs displaced [(3)H]ivermectin binding to rat brain membranes and recombinant GABA(A) receptors (alpha1beta3gamma2-subtype) with the same affinities, strongly suggesting that [(3)H]ivermectin labels the GABA(A) receptor in rodent brain. Avermectins, which were anticonvulsant, were also potent inhibitors of [(3)H]ivermectin binding in rat brain. However, the rank order for anticonvulsant activity did not parallel the rank order for affinity at the [(3)H]ivermectin site and it was reasoned that avermectins may have differential affinity or efficacy at subtypes of the GABA(A) receptor. All the active compounds tested potentiated the effects of GABA at recombinant GABA(A) receptors in oocytes and at native cortical GABA(A) receptors and the efficacy of avermectins at the GABA(A) receptor correlated best with their anticonvulsant potency. Although avermectins weakly inhibited [(3)H]strychnine binding in rat spinal cord, and inhibited glycine responses on primary cultured cortical neurons, activity at glycine receptors did not correlate with either anticonvulsant activity or toxicity. Because both anticonvulsant activity and toxicity correlated best with activity at GABA(A) receptors, it is unlikely that these effects can be separated, which may contraindicate the potential use of avermectins as anticonvulsants.


Subject(s)
Anticonvulsants/pharmacology , Ivermectin/analogs & derivatives , Receptors, GABA-A/drug effects , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Female , Ivermectin/adverse effects , Ivermectin/pharmacology , Male , Mice , Rats , Receptors, GABA-A/chemistry , Receptors, GABA-A/physiology , Receptors, Glycine/drug effects , Recombinant Proteins/drug effects , Xenopus
8.
Mol Endocrinol ; 14(5): 671-81, 2000 May.
Article in English | MEDLINE | ID: mdl-10809231

ABSTRACT

The dog GnRH receptor was cloned to facilitate the identification and characterization of selective nonpeptide GnRH antagonists. The dog receptor is 92% identical to the human GnRH receptor. Despite such high conservation, the quinolone-based nonpeptide GnRH antagonists were clearly differentiated by each receptor species. By contrast, peptide antagonist binding and functional activity were not differentiated by the two receptors. The basis of the differences was investigated by preparing chimeric receptors followed by site-directed mutagenesis. Remarkably, a single substitution of Phe313 to Leu313 in the dog receptor explained the major differences in binding affinities and functional activities. The single amino acid replacement of Phe313 of the human receptor with Leu313 resulted in a 160-fold decrease of binding affinity of the nonpeptide antagonist compound 1. Conversely, the replacement of Leu313 of the dog receptor with Phe313 resulted in a 360-fold increase of affinity for this compound. These results show that Phe313 of the GnRH receptor is critical for the binding of this structural class of GnRH antagonists and that the dog receptor can be "humanized" by substituting Leu for Phe. This study provides the first identification of a critical residue in the binding pocket occupied by nonpeptide GnRH antagonists and reinforces cautious extrapolation of ligand activity across highly conserved receptors.


Subject(s)
Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/pharmacology , Oligopeptides/pharmacology , Phenylalanine/chemistry , Receptors, LHRH/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Cloning, Molecular , Dogs , Hormone Antagonists/chemistry , Humans , Leucine/chemistry , Models, Molecular , Molecular Sequence Data , Molecular Structure , Mutagenesis, Site-Directed , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Protein Binding , Quinolones/chemistry , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 10(1): 5-8, 2000 Jan 03.
Article in English | MEDLINE | ID: mdl-10636230

ABSTRACT

Quinazolinone derivatives were synthesized and evaluated as non-peptidic growth hormone secretagogues. Modeling guided design of quinazolinone compound 21 led to a potency enhancement of greater than 200-fold compared to human growth hormone secretagogue affinity of a screening lead 4.


Subject(s)
Drug Design , Human Growth Hormone/metabolism , Quinazolines/chemical synthesis , Quinazolines/pharmacology , Receptors, Cell Surface/agonists , Receptors, G-Protein-Coupled , Animals , Binding Sites , Humans , Inhibitory Concentration 50 , Kinetics , Models, Molecular , Quinazolines/chemistry , Quinazolines/metabolism , Rats , Receptors, Cell Surface/metabolism , Receptors, Ghrelin , Secretory Rate/drug effects , Structure-Activity Relationship
10.
Endocrinology ; 141(1): 111-7, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10614629

ABSTRACT

Somatostatin (SST) potently inhibits insulin and glucagon release from pancreatic islets. Five distinct membrane receptors (SSTR1-5) for SST are known, and at least two (SSTR2 and SSTR5) have been proposed to regulate pancreatic endocrine function. Our current understanding of SST physiology is limited by the receptor subtype selectivity of peptidyl SST analogs, making it difficult to assign a physiological function to an identified SST receptor subtype. To better understand the physiology of SSTRs we studied the in vitro effects of potent subtype-selective nonpeptidyl SST analogs on the regulation of pancreatic glucagon and insulin secretion in wild-type (WT) and in somatostatin receptor 2 knockout (SSTR2KO) mice. There was no difference in basal glucagon and insulin secretion between islets isolated from SSTR2KO and WT mice; however, potassium/arginine-stimulated glucagon secretion was approximately 2-fold higher in islets isolated from SSTR2KO mice. Neither SST nor any SSTR-selective agonist inhibited basal glucagon or insulin release. SST-14 potently inhibited stimulated glucagon secretion in islets from WT mice and much less effectively in islets from SSTR2KO mice. The SSTR2 selective analog L-779,976 inhibited glucagon secretion in islets from WT, but was inactive in islets from SSTR2KO mice. L-817,818, an SSTR5 selective analog, slightly reduced glucagon release in both animal groups, whereas SSTR1, -3, and -4 selective analogs were inactive. SST and L-817,818 inhibited glucose stimulated insulin release in islets from WT and SSTR2KO mice. L-779,976 much less potently reduced insulin secretion from WT islets. In conclusion, our data demonstrate that SST inhibition of glucagon release in mouse islets is primarily mediated via SSTR2, whereas insulin secretion is regulated primarily via SSTR5.


Subject(s)
Glucagon/metabolism , Hormone Antagonists/pharmacology , Indoles , Insulin Antagonists/pharmacology , Islets of Langerhans/metabolism , Naphthalenes , Receptors, Somatostatin/genetics , Somatostatin/pharmacology , Amides/pharmacology , Animals , Glucose/pharmacology , In Vitro Techniques , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Somatostatin/agonists , Somatostatin/analogs & derivatives , Somatostatin-28 , Stimulation, Chemical
11.
Nat Med ; 5(12): 1390-5, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10581081

ABSTRACT

Although insulin-like growth factor 1 (IGF-1) has been associated with retinopathy, proof of a direct relationship has been lacking. Here we show that an IGF-1 receptor antagonist suppresses retinal neovascularization in vivo, and infer that interactions between IGF-1 and the IGF-1 receptor are necessary for induction of maximal neovascularization by vascular endothelial growth factor (VEGF). IGF-1 receptor regulation of VEGF action is mediated at least in part through control of VEGF activation of p44/42 mitogen-activated protein kinase, establishing a hierarchical relationship between IGF-1 and VEGF receptors. These findings establish an essential role for IGF-1 in angiogenesis and demonstrate a new target for control of retinopathy. They also explain why diabetic retinopathy initially increases with the onset of insulin treatment. IGF-1 levels, low in untreated diabetes, rise with insulin therapy, permitting VEGF-induced retinopathy.


Subject(s)
Endothelial Growth Factors/physiology , Lymphokines/physiology , Neovascularization, Pathologic/physiopathology , Receptor, IGF Type 1/physiology , Retinal Vessels/physiology , Animals , Growth Inhibitors/pharmacology , Humans , Insulin/pharmacology , Insulin-Like Growth Factor I/analogs & derivatives , Insulin-Like Growth Factor I/pharmacology , Ischemia/etiology , Ischemia/physiopathology , Ischemia/prevention & control , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/prevention & control , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/physiology , Receptor, IGF Type 1/antagonists & inhibitors , Receptors, Growth Factor/physiology , Receptors, Vascular Endothelial Growth Factor , Retinal Vessels/drug effects , Signal Transduction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Biochem Biophys Res Commun ; 263(2): 276-80, 1999 Sep 24.
Article in English | MEDLINE | ID: mdl-10491284

ABSTRACT

Somatostatin (SST) regulates growth hormone (GH) secretion from pituitary somatotrophs by interacting with members of the SST family of G-protein-coupled receptors (sst1-5). We have used potent, nonpeptidyl SST agonists with sst2 and sst5 selectivity to determine whether these receptor subtypes are involved in regulating growth hormone releasing hormone (GHRH) stimulated secretion. GHRH stimulated GH release from pituitary cells in a dose-dependent manner, and this secretion was inhibited by Tyr(11)-SST-14, a nonselective SST analog. A sst2 selective agonist, L-779,976, potently inhibited GHRH-stimulated GH release. In addition, L-817, 818, a potent sst5 receptor selective agonist, also inhibited GH secretion, but was approximately 10-fold less potent (P < 0.01, ANOVA) in inhibiting GH release than either Tyr(11)-SST-14 or L-779, 976. These results show that both sst2 and sst5 receptor subtypes regulate GHRH-stimulated GH release from rat pituitary cells.


Subject(s)
Growth Hormone-Releasing Hormone/pharmacology , Growth Hormone/metabolism , Indoles , Pituitary Gland, Anterior/drug effects , Receptors, Somatostatin/agonists , Somatostatin/agonists , Amides/pharmacology , Animals , Dose-Response Relationship, Drug , Male , Pituitary Gland, Anterior/cytology , Rats , Rats, Wistar , Somatostatin/analogs & derivatives , Somatostatin/pharmacology
13.
Endocrinology ; 140(8): 3790-6, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10433240

ABSTRACT

Somatostatin, also known as somatotropin release-inhibiting factor (SRIF), is secreted by pancreatic delta-cells and inhibits the secretion of both insulin and glucagon. SRIF initiates its actions by binding to a family of six G protein-coupled receptors (sst1, -2A, -2B, -3, -4, and -5) encoded by five genes. Messenger RNA for both sst2 and sst5 have been reported in the rat pancreas, and the sst2A receptor protein has been localized to rat pancreatic alpha and pancreatic polypeptide-secreting cells in the islets as well as to pancreatic acinar cells. In this study we have used double immunostaining to show that the sst5 protein is expressed exclusively in the beta-cells of rat pancreatic islets and localizes with insulin-secreting alpha-cells. The sst5 receptor is not colocalized with sst2A. Thus, in the rat SRIF inhibits pancreatic insulin and glucagon secretion via different sst receptor subtypes.


Subject(s)
Insulin/analysis , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Receptors, Somatostatin/analysis , Receptors, Somatostatin/genetics , Alternative Splicing , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Humans , Immunohistochemistry , Insulin/metabolism , Insulin Secretion , Mice , Molecular Sequence Data , RNA, Messenger/genetics , Rats , Receptors, Somatostatin/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transcription, Genetic , Transfection
14.
Bioorg Med Chem Lett ; 9(3): 491-6, 1999 Feb 08.
Article in English | MEDLINE | ID: mdl-10091708

ABSTRACT

Backbone cyclization of urea-based somatostatin agonists resulted in novel, orally bioavailable agonists. Binding assays confirmed that the resulting conformationally constrained cyclic ureas retained the potency of their acyclic counterparts. SAR studies subsequently led to highly potent analogs, selective for receptor subtype 2, and having good oral bioavailability.


Subject(s)
Somatostatin/agonists , Somatostatin/pharmacology , Urea/chemistry , Administration, Oral , Animals , Benzimidazoles , Biological Availability , Dogs , Indoles , Somatostatin/chemistry , Somatostatin/pharmacokinetics , Structure-Activity Relationship
15.
J Neurochem ; 72(1): 318-26, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9886084

ABSTRACT

Glutamate-gated chloride channels have been described in nematodes, insects, crustaceans, and mollusks. Subunits from the nematode and insect channels have been cloned and are phylogenetically related to the GABA and glycine ligand-gated chloride channels. Ligand-gated chloride channels are blocked with variable potency by the nonselective blocker picrotoxin. The first two subunits of the glutamate-gated chloride channel family, GluClalpha and GluClbeta, were cloned from the free living nematode Caenorhabditis elegans. In this study, we analyze the blockade of these novel channels by picrotoxin. In vitro synthesized GluClalpha and GluClbeta RNAs were injected individually or coinjected into Xenopus oocytes. The EC50 values for picrotoxin block of homomeric GluClalpha and GluClbeta were 59 microM and 77 nM, respectively. Picrotoxin block of homomeric GluClbeta channels was promoted during activation of membrane current with glutamate. In addition, recovery from picrotoxin block was faster during current activation by glutamate. A chimeric channel between the N-terminal extracellular domain of GluClalpha and the C-terminal membrane-spanning domain of GluClbeta localized the higher affinity picrotoxin binding site to the membrane-spanning domains of GluClbeta. A point mutation within the M2 membrane-spanning domain of GluClbeta reduced picrotoxin sensitivity >10,000-fold. We conclude that picrotoxin blocks GluCl channels by binding to a site accessible when the channel is open.


Subject(s)
Chloride Channels/physiology , GABA Antagonists/pharmacology , Glutamic Acid/pharmacology , Ion Channel Gating/drug effects , Picrotoxin/pharmacology , Amino Acid Sequence , Animals , Antinematodal Agents/pharmacology , Binding Sites/physiology , Caenorhabditis elegans , Chloride Channels/chemistry , Chloride Channels/genetics , Drug Interactions , Drug Resistance , Electrophysiology , Ivermectin/pharmacology , Kinetics , Membrane Potentials/drug effects , Molecular Sequence Data , Oocytes/physiology , Point Mutation , Protein Structure, Tertiary , Xenopus
16.
J Comb Chem ; 1(5): 388-96, 1999.
Article in English | MEDLINE | ID: mdl-10748735

ABSTRACT

The tetradecapeptide somatostatin is widely distributed throughout the body and is thought to be involved with a variety of regulatory functions. Recently, five human somatostatin receptors (hSSTR1-5) have been cloned and characterized. Several selective peptidal agonists of the hSSTR receptors are known, and we sought to apply this information to the design of novel non-peptide small molecule ligands for each receptor. Initial computational methods identified a 200 nM murine SSTR2 active compound via a database search of our sample collection. A combinatorial library was designed around the structural class of the compound with the goal of rapidly developing this initial lead into the desired subtype-selective small molecules in order to characterize the pharmacology of each of the receptor subtypes. The library was synthesized using the resin-archive, iterative deconvolution format. The total number of unique compounds in the library was expected to be 131,670, present in 79 mixtures of 1330 or 2660 compounds per mixture. Through sequences of screening and mixture deconvolution, the components of selective and highly active (Ki = 50 pM to 200 nM) non-peptide small molecule ligands for somatostatin subtypes 1, 2, 4, and 5 were identified. In addition to discovering compounds with the desired activity and selectivity, useful structure/activity information was generated which can be used in the design of new compounds and second-generation combinatorial libraries.


Subject(s)
Combinatorial Chemistry Techniques/methods , Databases, Factual , Ligands , Receptors, Somatostatin/metabolism , Drug Design , Humans , Kinetics , Molecular Structure , Recombinant Proteins/metabolism , Somatostatin/chemistry , Structure-Activity Relationship
17.
Science ; 282(5389): 737-40, 1998 Oct 23.
Article in English | MEDLINE | ID: mdl-9784130

ABSTRACT

Nonpeptide agonists of each of the five somatostatin receptors were identified in combinatorial libraries constructed on the basis of molecular modeling of known peptide agonists. In vitro experiments using these selective compounds demonstrated the role of the somatostatin subtype-2 receptor in inhibition of glucagon release from mouse pancreatic alpha cells and the somatostatin subtype-5 receptor as a mediator of insulin secretion from pancreatic beta cells. Both receptors regulated growth hormone release from the rat anterior pituitary gland. The availability of high-affinity, subtype-selective agonists for each of the somatostatin receptors provides a direct approach to defining their physiological functions.


Subject(s)
Amides/pharmacology , Receptors, Somatostatin/agonists , Amides/metabolism , Amino Acid Sequence , Animals , Cell Line , Cells, Cultured , Cricetinae , Drug Design , Glucagon/metabolism , Growth Hormone/metabolism , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Ligands , Membrane Proteins , Mice , Models, Chemical , Molecular Sequence Data , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/metabolism , Rats , Receptors, Somatostatin/physiology
18.
Mol Endocrinol ; 12(10): 1594-604, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9773982

ABSTRACT

UNLABELLED: Ligand-dependent interactions between nuclear receptors and members of a family of nuclear receptor coactivators are associated with transcriptional activation. Here we used fluorescence resonance energy transfer (FRET) as an approach for detecting and quantitating such interactions. Using the ligand binding domain (LBD) of peroxisome proliferator-activated receptor (PPARgamma) as a model, known agonists (thiazolidinediones and delta12, 14-PGJ2) induced a specific interaction resulting in FRET between the fluorescently labeled LBD and fluorescently labeled coactivators [CREB-binding protein (CBP) or steroid receptor coactivator-1 (SRC-1)]. Specific energy transfer was dose dependent; individual ligands displayed distinct potency and maximal FRET profiles that were identical when results obtained using CBP vs. SRC-1 were compared. In addition, half-maximally effective agonist concentrations (EC59s) correlated well with reported results using cell-based assays. A site-directed AF2 mutant of PPARgamma (E471A) that abrogated ligand-stimulated transcription in transfected cells also failed to induce ligand-mediated FRET between PPARgamma LBD and CBP or SRC-1. Using estrogen receptor (ERalpha) as an alternative system, known agonists induced an interaction between ERalpha LBD and SRC-1, whereas ER antagonists disrupted agonist-induced interaction of ERalpha with SRC-1. In the presence of saturating agonist concentrations, unlabeled CBP or SRC-1 was used to compete with fluorescently labeled coactivators with saturation kinetics. Relative affinities for the individual receptor-coactivator pairs were determined as follows: PPARgamma-CBP = ERalpha-SRC-1 > PPARgamma-SRC-1 >> ERalpha-CBP. CONCLUSIONS: 1) FRET-based coactivator association is a novel approach for characterizing nuclear receptor agonists or antagonists; individual ligands display potencies that are predictive of in vivo effects and distinct profiles of maximal activity that are suggestive of alternative receptor conformations. 2) PPARgamma interacts with both CBP and SRC-1; transcriptional activation and coactivator association are AF2 dependent. 3) Nuclear receptor LBDs have distinct affinities for individual coactivators; thus, PPARgamma has a greater apparent affinity for CBP than for SRC-1, whereas ERalpha interacts preferentially with SRC-1 but very weakly with CBP.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/metabolism , Spectrometry, Fluorescence/methods , Thiazolidinediones , Transcription Factors/metabolism , Animals , Binding Sites , CREB-Binding Protein , Cricetinae , Energy Transfer , Estrogen Receptor alpha , Histone Acetyltransferases , Nuclear Proteins/metabolism , Nuclear Receptor Coactivator 1 , Pioglitazone , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Rosiglitazone , Thiazoles/pharmacology , Trans-Activators/metabolism , Transcription Factors/agonists , Transcription Factors/genetics
19.
Proc Natl Acad Sci U S A ; 95(18): 10836-41, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9724791

ABSTRACT

A series of nonpeptide somatostatin agonists which bind selectively and with high affinity to somatostatin receptor subtype 2 (sst2) have been synthesized. One of these compounds, L-054,522, binds to human sst2 with an apparent dissociation constant of 0.01 nM and at least 3,000-fold selectivity when evaluated against the other somatostatin receptors. L-054,522 is a full agonist based on its inhibition of forskolin-stimulated adenylate cyclase activity in Chinese hamster ovary-K1 cells stably expressing sst2. L-054,522 has a potent inhibitory effect on growth hormone release from rat primary pituitary cells and glucagon release from isolated mouse pancreatic islets. Intravenous infusion of L-054,522 to rats at 50 microgram/kg per hr causes a rapid and sustained reduction in growth hormone to basal levels. The high potency and selectivity of L-054, 522 for sst2 will make it a useful tool to further characterize the physiological functions of this receptor subtype.


Subject(s)
Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Indoles/chemical synthesis , Indoles/pharmacology , Molecular Mimicry , Receptors, Somatostatin/agonists , Animals , CHO Cells , Cricetinae , Glucagon/antagonists & inhibitors , Glucagon/metabolism , Growth Hormone/metabolism , Humans , Insulin/metabolism , Insulin Antagonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Rats
20.
Gastroenterology ; 114(6): 1125-32, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9609748

ABSTRACT

BACKGROUND & AIMS: Somatostatin receptor subtype 2 (sst2) agonists inhibit gastric secretion. The role of sst2 in the regulation of acid secretion was assessed using sst2 knockout mice and urethane to induce somatostatin release. METHODS: Acid secretion was monitored every 10 minutes by gastric perfusion and backtitration of perfusates in fasted, urethane-anesthetized C57/129 sst2 (-/-) mice and wild-type (+/+) mice. The ileal vein was cannulated for drug injection. Intragastric pH and serum gastrin were monitored 1 hour after anesthesia without perfusion. RESULTS: Gastric pH values were lower in sst2 (-/-) mice (3.8 +/- 0.3) than in wild-type mice (7.1 +/- 0.1, P < 0.05), and there was no difference in gastrin levels. Basal acid output per 2 hours was 10-fold higher in sst2 knockout mice compared with wild-type mice. The gastrin antibody abolished the high basal acid secretion in sst2 (-/-) mice and had no effect in wild-type mice. The somatostatin antibody increased basal secretion by 4-fold in wild-type and had no effect in knockout mice. Somatostatin 14 or the sst2 agonist DC 32-87 inhibited pentagastrin-stimulated acid secretion in wild-type mice, but did not alter basal secretion in knockout mice. CONCLUSIONS: These results indicate that sst2 is the main subtype whereby endogenous somatostatin suppresses gastric acid secretion through inhibition of gastrin action.


Subject(s)
Gastric Acid/metabolism , Mice, Knockout/genetics , Mice, Knockout/metabolism , Receptors, Somatostatin/genetics , Receptors, Somatostatin/physiology , Animals , Antibodies, Monoclonal/pharmacology , Gastric Mucosa/metabolism , Gastrins/blood , Gastrins/immunology , Hydrogen-Ion Concentration , Mice , Mice, Inbred C57BL , Pentagastrin/pharmacology , Receptors, Somatostatin/agonists , Somatostatin/analogs & derivatives , Somatostatin/immunology , Somatostatin/pharmacology , Somatostatin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...