Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Mol Pharmacol ; 59(1): 30-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11125021

ABSTRACT

A novel thiadiazole compound, SCH-202676 (N-(2,3-diphenyl-1,2, 4-thiadiazol-5-(2H)-ylidene)methanamine), has been identified as an inhibitor of both agonist and antagonist binding to G protein-coupled receptors (GPCRs). SCH-202676 inhibited radioligand binding to a number of structurally distinct, heterologously expressed GPCRs, including the human mu-, delta-, and kappa-opioid, alpha- and beta-adrenergic, muscarinic M1 and M2, and dopaminergic D1 and D2 receptors, but not to the tyrosine kinase epidermal growth factor receptor. SCH-202676 had no direct effect on G protein activity as assessed by [35S]guanosine-5'-O-(gamma-thio)triphosphate binding to purified recombinant G(oalpha)- or G(betagamma)-stimulated ADP-ribosylation of G(oalpha) by pertussis toxin. In addition, SCH-202676 inhibited antagonist binding to the beta2-adrenergic receptor expressed in Escherichia coli, a system devoid of classical heterotrimeric G proteins. SCH-202676 inhibited radiolabeled agonist and antagonist binding to the alpha2a-adrenergic receptor with an IC50 value of 0.5 microM, decreased the Bmax value of the binding sites with a slight increase in the KD value, and inhibited agonist-induced activation of the receptor. The effects of SCH-202676 were reversible. Incubation of plasma membranes with 10 microM SCH-202676 did not alter subsequent radioligand binding to the alpha2a-adrenergic receptor and the dopaminergic D1 receptor. Taken together, our data suggest that SCH-202676 has the unique ability to allosterically regulate agonist and antagonist binding to GPCRs in a manner that is both selective and reversible. The scope of the data presented suggests this occurs by direct interaction with a structural motif common to a large number of GPCRs or by activation/inhibition of an unidentified accessory protein that regulates GPCR function.


Subject(s)
Adrenergic Agents/pharmacology , Receptors, Adrenergic/metabolism , Thiazoles/pharmacology , Allosteric Regulation , Escherichia coli , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , HT29 Cells , Humans , Radioligand Assay , Receptors, Adrenergic/drug effects , Thiadiazoles , Tumor Cells, Cultured
2.
Endocrinology ; 141(12): 4524-32, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11108264

ABSTRACT

The receptor for melanin-concentrating hormone (MCH) was recently identified as the orphan G protein-coupled receptor SLC-1. In this study, a CHO cell line expressing the MCH receptor (Kd = 1.3 nM; binding capacity, 3.6 pmol/mg protein) is used to assess the ability of the MCH receptor to couple to Gi, Go, and Gq proteins. The results demonstrate that MCH inhibits forskolin-stimulated cAMP production in a pertussis toxin- (PTX)-sensitive manner in CHO-MCHR cells (EC50 = 100 pM), indicating that the MCH receptor couples to one or more members of the Gi subfamily of G proteins. In addition, MCH stimulates increases in phosphoinositide metabolism (EC50 = 50 nM) and in intracellular free Ca2+ levels (EC50 = 10 nM). MCH-stimulated inositol phosphate production and increases in intracellular free Ca2+ are partially inhibited (60% and 40%, respectively) by PTX pretreatment, demonstrating that there are at least two components of each of these signaling pathways. One component is PTX sensitive and therefore mediated through a Gi/Go protein. A distinct G protein-coupled (probably Gq type) mediates the PTX-insensitive component. To distinguish Gi vs. Go coupling, MCH-stimulated mitogen-activated protein (MAP) kinase activity was examined. Gi and Go use separate signaling pathways to mediate MAP kinase activation in CHOcells. Protein kinase C (PKC) activity is essential in the Go-dependent MAP kinase signaling pathway, but is not required in the GC-dependent MAP kinase signaling pathway. MCH stimulated MAP kinase activity is decreased (50%), but not abolished, by inhibition of PKC activity or depletion of cellular PKC, indicating that MCH-stimulated MAP kinase activity is mediated through both Gi- and Go-dependent signaling mechanisms. The results of this study are the first to clearly demonstrate that the MCH receptor couples to multiple G proteins to mediate several diverse intracellular signaling pathways.


Subject(s)
GTP-Binding Proteins/physiology , Receptors, Pituitary Hormone/physiology , Signal Transduction , Adenylyl Cyclases/metabolism , Animals , CHO Cells , Cell Membrane/metabolism , Colforsin/pharmacology , Cricetinae , Cyclic AMP/biosynthesis , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Gene Expression , Heterotrimeric GTP-Binding Proteins/physiology , Humans , Hypothalamic Hormones/metabolism , Hypothalamic Hormones/pharmacology , Inositol Phosphates/biosynthesis , Lysophospholipids/pharmacology , Melanins/metabolism , Melanins/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Pertussis Toxin , Pituitary Hormones/metabolism , Pituitary Hormones/pharmacology , Protein Kinase C/antagonists & inhibitors , Receptors, Pituitary Hormone/genetics , Transfection , Virulence Factors, Bordetella/pharmacology
3.
Peptides ; 21(7): 961-7, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10998529

ABSTRACT

The recent identification of the nociceptin receptor-nociceptin system and the description of its role in nociceptive processing has produced numerous investigative studies. A fundamental part of this research is to understand the cellular signaling events (i.e. the building blocks) upon which the pharmacology of this intriguing system is based. As anticipated, nociceptin receptor activation inhibits the formation of cAMP formation via a pertussis toxin-sensitive G-protein. This indicates that nociceptin receptor couples to the G(i)/G(o) class of G-protein(s). However, there is now growing evidence for nociceptin activation of additional signaling pathways, including MAP kinase and phospholipase C/[Ca(2+)](i). These signaling events are discussed in this review.


Subject(s)
Opioid Peptides/metabolism , Receptors, Opioid/metabolism , Adenylyl Cyclases/metabolism , Animals , CHO Cells , COS Cells , Calcium/metabolism , Cricetinae , Cyclic AMP/metabolism , GTP-Binding Proteins/metabolism , Humans , Inhibitory Concentration 50 , Ion Channels , Kinetics , MAP Kinase Signaling System , Models, Biological , Opioid Peptides/chemistry , Receptors, Opioid/chemistry , Signal Transduction , Tumor Cells, Cultured , Type C Phospholipases/metabolism , Vasodilator Agents/chemistry , Vasodilator Agents/metabolism , Nociceptin Receptor , Nociceptin
4.
Mol Pharmacol ; 58(1): 217-25, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10860944

ABSTRACT

A molecular model of the human melanin-concentrating hormone (MCH) peptide was constructed and docked into a helical, bacteriorhodopsin-based model of the recently identified human MCH receptor. From this hormone-receptor complex, potential sites of agonist-receptor interaction were identified, and site-directed mutagenesis was used to substitute residues predicted to reside within the receptor binding pocket. Substitution of Asp(123)(3.32) in the third transmembrane domain of the receptor resulted in a loss of detectable (125)I-MCH binding and of MCH-stimulated Ca(2+) flux; cell surface expression of the mutant receptor was not affected. Arg(11) and Arg(14) of the MCH ligand were identified as potential sites of interaction with Asp(123)(3.32). [Ala(14)]-MCH was equipotent to native MCH in its ability to bind to and activate the wild-type MCH receptor, whereas [Ala(11)]-MCH displayed a 3000-fold reduction in binding affinity and a complete loss of measurable functional activity. Furthermore, [Lys(11)]-MCH and [D-Arg(11)]-MCH displayed reduced affinity for the receptor. [Lys(11)]-MCH was observed to be a partial agonist, eliciting approximately 67% of the native peptide's activity in a Ca(2+) flux assay, and [D-Arg(11)]-MCH was determined to be a functional antagonist with a K(b) valve of 15.8 microM. These data provide evidence that a basic moiety with specific stereochemical requirements at this site is needed for receptor activation. We conclude that both Asp(123)(3.32) in the MCH receptor and Arg(11) in the MCH peptide are required for the formation of the MCH peptide/receptor complex and propose that they form a direct interaction that is critical for receptor function.


Subject(s)
Hypothalamic Hormones/metabolism , Melanins/metabolism , Pituitary Hormones/metabolism , Receptors, Pituitary Hormone/metabolism , Amino Acid Sequence , Animals , Binding, Competitive , CHO Cells , COS Cells , Cricetinae , Flow Cytometry , Humans , Models, Molecular , Mutagenesis, Site-Directed , Peptides/chemistry , Peptides/metabolism , Receptors, Pituitary Hormone/antagonists & inhibitors , Receptors, Pituitary Hormone/genetics , Transfection
5.
J Neurochem ; 71(6): 2252-9, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9832122

ABSTRACT

The diverse physiological actions of galanin are thought to be mediated through activation of galanin receptors (GalRs). We report the genomic and cDNA cloning of a mouse GalR that possesses a genomic structure distinct from that of GalR1 and encodes a functional galanin receptor. The mouse GalR gene consists of two exons separated by a single intron within the protein-coding region. The splicing site for the intron is located at the junction between the third transmembrane domain and the second intracellular loop. The cDNA encodes a 370-amino acid putative G protein-coupled receptor that is markedly different from human GalR1 and rat GalR3 (38 and 57%) but shares high homology with rat GalR2 (94%). In binding studies utilizing membranes from COS-7 cells transfected with mouse GalR2 cDNA, the receptor displayed high affinity (K(D) = 0.47 nM) and saturable binding with 125I-galanin (Bmax = 670 fmol/mg). The radioligand binding can be displaced by galanin and its analogues in a rank order: galanin approximately = M40 approximately = M15 approximately = M35 approximately = C7 approximately = galanin(2-29) approximately = galanin(1-16) >> galanin(10-29) approximately = galanin(3-29), which resembles the pharmacological profile of the rat GalR2. Receptor activation by galanin in COS-7 cells stimulated phosphoinositide metabolism, which was not reversed by pertussis toxin. Thus, the galanin receptor encoded in the cloned mouse GalR gene is the type 2 galanin receptor and is active in both ligand binding and signaling assays.


Subject(s)
Cloning, Molecular , DNA, Complementary/genetics , Genome , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Amino Acid Sequence , Animals , Base Sequence , Humans , Isomerism , Mice , Molecular Sequence Data , Rats , Receptor, Galanin, Type 2 , Receptors, Galanin , Sequence Homology, Amino Acid
6.
J Neurochem ; 71(3): 1024-33, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9721727

ABSTRACT

The recently identified 17-amino acid peptide nociceptin (orphanin FQ) is the endogenous ligand for the opioid receptor-like-1 (ORL-1) receptor. A physiologic role for nociceptin (OFQ) activation of the ORL-1 receptor (OFQR) may be to modulate opioid-induced analgesia. The molecular mechanism by which nociceptin (OFQ) and ORL-1 (OFQR) modify opioid-stimulated effects, however, is unclear. Both ORL-1 (OFQR) and opioid receptors mediate pertussis toxin (PTX)-sensitive signal transduction, indicating these receptors are capable of coupling to Gi/Go proteins. This study determines that nociceptin stimulates an intracellular signaling pathway, leading to activation of mitogen-activated protein (MAP) kinase in CHO cells expressing ORL-1 receptor (OFQR). Nociceptin (OFQ)-stimulated MAP kinase activation was inhibited by PTX or by expression of the carboxyl terminus of beta-adrenergic receptor kinase (betaARKct), which specifically blocks Gbetagamma-mediated signaling. Expression of the proline-rich domain of SOS (SOS-PRO), which inhibits SOS interaction with p21ras, also attenuated nociceptin (OFQ)-stimulated MAP kinase activation. The phosphatidylinositol 3-kinase (PI-3K) inhibitors wortmannin and LY294002 reduced nociceptin (OFQ)-stimulated MAP kinase activation, whereas inhibition of protein kinase C (PKC) activity by bisindolylmaleimide I or cellular depletion of PKC had no effect. In a similar manner, in cells expressing mu-opioid receptor, [D-Ala2,N-Me-Phe4,Gly-ol]-enkephalin (DAMGO; a mu-opioid receptor-selective agonist) stimulated PTX-sensitive MAP kinase activation that was inhibited by wortmannin, LY294002, betaARKct expression, or SOS-PRO expression but not affected by inhibition of PKC activity. These results indicate that both ORL-1 (OFQR) and mu-opioid receptors mediate MAP kinase activation via a signaling pathway using the betagamma-subunit of Gi, a PI-3K, and SOS, independent of PKC activity. In cells expressing both ORL-1 (OFQR) and mu-opioid receptors, pretreatment with nociceptin decreased subsequent nociceptin (OFQ)- or DAMGO-stimulated MAP kinase activation. In contrast, pretreatment of cells with DAMGO decreased subsequent DAMGO-stimulated MAP kinase but had no effect on subsequent nociceptin (OFQ)-stimulated MAP kinase activation. These results demonstrate that nociceptin (OFQ) activation of ORL-1 (OFQR) can modulate mu-opioid receptor signaling in a cellular system.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , GTP-Binding Proteins/physiology , Receptors, Opioid, mu/physiology , Receptors, Opioid/physiology , Signal Transduction/physiology , Animals , CHO Cells , Cricetinae , Enzyme Activation/physiology , Humans , Opioid Peptides/pharmacology , Receptors, Opioid/agonists , Receptors, Opioid/drug effects , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/drug effects , Nociceptin Receptor , Nociceptin
7.
Obes Res ; 6(2): 147-56, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9545022

ABSTRACT

Glucagon-like peptide (7-36) amide (GLP-1) acutely inhibits food and water consumption in rats after intracerebroventricular (icv) administration. To assess the potential for desensitization of these effects, we investigated the effects of chronic icv administration of GLP-1 on food consumption and body weight in Sprague-Dawley (SD) rats and Zucker (fa/fa) obese rats. In vitro functional densensitization of the GLP-1 receptor was not observed after overnight exposure of Rin m5F insulinoma cells to GLP-1 at concentrations up to 10 nM. Administration of GLP-1 to SD rats (30 microg icv twice a day for 6 days) resulted in significant reductions in 24-hour food consumption each day (25 +/- 1%). Continuous icv infusion of GLP-1 for 7 and 14 days significantly inhibited cumulative food consumption and reduced body weight in SD rats. In the genetically obese Zucker rat, chronic dosing with GLP-1 (30 microg icv) once a day for 6 days caused significant reductions in food consumption each day and a reduction in body weight. These results indicate that the GLP-1 pathways in the central nervous system controlling food consumption do not desensitize after chronic exposure to GLP-1 and suggest that agonists of the central GLP-1 receptor may be effective agents for the treatment of obesity.


Subject(s)
Body Weight/drug effects , Eating/drug effects , Neurotransmitter Agents/pharmacology , Obesity/physiopathology , Peptide Fragments/pharmacology , Animals , Glucagon , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Glucagon-Like Peptides , Injections, Intraventricular , Insulinoma/metabolism , Male , Pancreatic Neoplasms/metabolism , Peptide Fragments/administration & dosage , Rats , Rats, Sprague-Dawley , Rats, Zucker , Receptors, Glucagon/drug effects , Receptors, Glucagon/metabolism , Tumor Cells, Cultured
8.
J Recept Signal Transduct Res ; 18(4-6): 311-20, 1998.
Article in English | MEDLINE | ID: mdl-9879063

ABSTRACT

The rat GalR1 galanin receptor was used as a prototypic G protein-coupled receptor to test the feasibility of heterologous expression in a retrovirus-based system. The system utilizes an independent retroviral vector pMX, a virus-packaging cell line BOSC23 and a pre-B cell line BA/F3 as the host for expression. A polyclonal cell population that expresses high ligand affinity (KD = 0.18 nM) and high level (7 pmol/mg) of GalR1 was generated within days with no drug sensitivity-based selection. The expression represented a 20-fold increase over the expression level of GalR1 achieved in CHO cells. The affinity of galanin for the expressed receptor was decreased by 19-fold in the presence of GTP-gamma-S, suggesting that the expression system can produce active galanin receptor functionally coupled to G proteins. The fast and efficient method to generate stable cell lines and to prepared large quantities of receptors may provide a general application for expression of other G protein-coupled receptors.


Subject(s)
GTP-Binding Proteins/biosynthesis , Receptors, Neuropeptide/biosynthesis , Recombinant Proteins/biosynthesis , Retroviridae/genetics , Animals , Cell Line , GTP-Binding Proteins/genetics , Galanin/metabolism , Genetic Vectors , Rats , Receptors, Galanin , Receptors, Neuropeptide/genetics
9.
Am J Physiol ; 272(4 Pt 2): R1204-9, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9140021

ABSTRACT

Obesity occurs whenever energy intake exceeds energy expenditure. The ob gene product leptin is a potent anorectic agent when administered to ob/ob mice, but its effects on energy expenditure have not been investigated in detail. The present study was designed to analyze the acute metabolic effects of leptin in vivo. Analysis of oxygen consumption in ob/ob mice demonstrated a reduction in energy expenditure compared with lean controls; this reduction showed a diurnal fluctuation and was most evident during the light cycle. A single intraperitoneal dose of leptin increased oxygen consumption during the light cycle in ob/ob mice, ablating the circadian fluctuation in this parameter. In addition, leptin had a profound effect on fuel selection: the respiratory quotient was markedly reduced, indicating a reduction in carbohydrate oxidation and an increase in fat oxidation. These acute effects of leptin on metabolic parameters are consistent with the selective loss of body fat observed on chronic leptin treatment and suggest that increased energy utilization plays an important role in the anti-obese actions of leptin.


Subject(s)
Energy Metabolism/drug effects , Lipid Metabolism , Obesity/metabolism , Proteins/pharmacology , Analysis of Variance , Animals , Calorimetry, Indirect , Circadian Rhythm/drug effects , Cloning, Molecular , Darkness , Escherichia coli , Humans , Leptin , Light , Mice , Mice, Obese , Obesity/genetics , Oxygen Consumption , Recombinant Proteins/pharmacology , Thinness , Time Factors
10.
J Biol Chem ; 272(12): 7765-9, 1997 Mar 21.
Article in English | MEDLINE | ID: mdl-9065438

ABSTRACT

Deletion of residues 252-259 within the putative second intracellular loop of the human glucagon receptor results in a protein with high affinity for glucagon but with attenuated agonist activation of adenylyl cyclase. The Delta252-259 mutant has 4-fold higher affinity for glucagon than does the wild type receptor. The nonhydrolyzable GTP analog, guanosine 5'-(beta, gamma-imido)triphosphate (Gpp(NH)p), inhibits binding of 125I-glucagon to the wild type receptor but not to the Delta252-259 mutant. Divalent cations such as MgCl2 and CaCl2 stimulate the binding of 125I-glucagon to the wild type receptor by increasing glucagon affinity. The rate of dissociation of 125I-glucagon is decreased 4-fold by MgCl2 and increased 6-fold by Gpp(NH)p. However, divalent cations do not affect the binding of 125I-glucagon to the Delta252-259 mutant. The rate of dissociation of 125I-glucagon from the Delta252-259 mutant protein is equivalent to the rate of dissociation from the wild type receptor in the presence of MgCl2. These data suggest that at least three conformations of the glucagon receptor can exist in the membrane based on their differing affinities for 125I-glucagon. Deletion of residues 252-259 appears to lock the protein in the conformation promoted by divalent cations and prevents the protein from normal coupling to Gs.


Subject(s)
Receptors, Glucagon/metabolism , Amino Acid Sequence , Animals , COS Cells , Cations, Divalent , Cell Membrane/metabolism , Glucagon/metabolism , Humans , Kinetics , Molecular Sequence Data , Mutagenesis , Radioligand Assay , Receptors, Glucagon/agonists , Receptors, Glucagon/genetics , Recombinant Proteins/agonists , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion
11.
J Clin Invest ; 99(3): 385-90, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9022070

ABSTRACT

Leptin administration reduces obesity in leptin-deficient ob/ob mice; its effects in obese humans, who have high circulating leptin levels, remain to be determined. This longitudinal study was designed to determine whether diet-induced obesity in mice produces resistance to peripheral and/or central leptin treatment. Obesity was induced in two strains of mice by exposure to a 45% fat diet. Serum leptin increased in proportion to body weight (P < 0.00001). Whereas C57BL/6 mice initially responded to peripherally administered leptin with a marked decrease in food intake, leptin resistance developed after 16 d on high fat diet; mice on 10% fat diet retained leptin sensitivity. In AKR mice, peripheral leptin significantly decreased food intake in both 10 and 45% fat-fed mice after 16 d of dietary treatment. However, after 56 d, both groups became resistant to peripherally administered leptin. Central administration of leptin to peripherally leptin-resistant AKR mice on 45% fat diet resulted in a robust response to leptin, with a dose-dependent decrease in food intake (P < 0.00001) and body weight (P < 0.0001) after a single intracerebroventricular infusion. These data demonstrate that, in a diet-induced obesity model, mice exhibit resistance to peripherally administered leptin, while retaining sensitivity to centrally administered leptin.


Subject(s)
Obesity/drug therapy , Proteins/administration & dosage , Proteins/therapeutic use , Animals , Appetite Regulation/drug effects , Body Weight/drug effects , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Dose-Response Relationship, Drug , Drug Resistance , Eating/drug effects , Feeding Behavior/drug effects , Leptin , Longitudinal Studies , Male , Mice , Mice, Inbred AKR , Mice, Inbred C57BL , Obesity/blood , Proteins/analysis , Time Factors
12.
Peptides ; 18(2): 319-21, 1997.
Article in English | MEDLINE | ID: mdl-9149306

ABSTRACT

Exendin-4, a reptilian GLP-1 analogue, has been fluorescently labeled by covalently linking a fluorescein moiety onto the Trp residue yielding fluorescein-Trp25-exendin-4 (FLEX). FLEX is equipotent to GLP-1(7-36)-amide and exendin-4 as an inhibitor of [125I] GLP-1 binding to the human GLP-1 receptor stably expressed in CHO cells, and maintains full biological potency and efficacy as measured by the stimulation of cAMP accumulation in these cells. FLEX binding to CHO/hGLP-1R membranes results in an increase in fluorescence anisotropy. The binding is specific and saturable (Kd = 2.0 +/- 0.4 nM), and GLP-1(7-36)-amide and exendin-4 are equipotent inhibitors of FLEX binding to the human GLP-1 receptor. Thus, FLEX is a potent, biologically active ligand that is useful for the study of the binding and functional characteristics of the human GLP-1 receptor.


Subject(s)
Fluoresceins/metabolism , Peptides/metabolism , Receptors, Glucagon/metabolism , Recombinant Proteins/metabolism , Animals , Binding, Competitive , CHO Cells , Cell Membrane/metabolism , Cricetinae , Cyclic AMP/metabolism , Fluoresceins/chemical synthesis , Fluoresceins/pharmacology , Gastrointestinal Hormones/metabolism , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Humans , Kinetics , Peptides/chemical synthesis , Peptides/pharmacology , Receptors, Glucagon/analysis , Receptors, Glucagon/biosynthesis , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Transfection
13.
Eur J Pharmacol ; 336(2-3): 233-42, 1997 Oct 08.
Article in English | MEDLINE | ID: mdl-9384238

ABSTRACT

Opioid receptor-like 1 (ORL1) receptor, a member of the superfamily of G-protein-coupled receptors has significant primary sequence homology to the mu-, delta- and kappa-opioid receptors. The ORL1 receptor is selectively activated by the recently discovered peptide nociceptin. To probe the functional homology amongst these receptors, a Chinese hamster ovary (CHO) cell line expressing the human ORL1 receptor has been characterized. Nociceptin inhibited forskolin-stimulated increases in intracellular cAMP with an IC50 of 70 pM. Stimulation by nociceptin caused a 2-fold increase in the rate of [35S]GTPgammaS binding to membranes derived from CHO cells expressing the ORL1 receptor. Following incubation with nociceptin mitogen-activated protein kinase activity was increased by 2-fold in cells expressing the ORL1 receptor. In non-transfected CHO cells, nociceptin had no effect on cAMP accumulation, the rate of [35S]GTPgammaS binding or mitogen-activated protein kinase activity. Human ORL1 receptors expressed in CHO cells selectively bound [125I][Tyr14]nociceptin with a Kd of 2.1 pM and a Bmax of 2.6 pmol/mg protein. Similar to opioid receptors, nociceptin binding to the ORL1 receptor was altered by Na+, GTPgammaS and dithiothreitol. Na+ increased the Kd of nociceptin binding to the ORL1 receptor. GTPgammaS decreased the apparent Bmax of [125I][Tyr14]nociceptin binding but had no effect on the Kd of the remaining sites. Pretreatment with dithiothreitol inhibited nociceptin binding to the ORL1 receptor. Nociceptin binding was insensitive to low nanomolar concentrations of opioid receptor-selective agonists and antagonists. However, high micromolar levels of opioid receptor-selective agents inhibited the binding. Morphine, naloxone, naltrindole, nor-Binaltorphimine and CTAP (D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2) inhibited nociceptin binding to ORL1 receptor with Ki values of 36, 24, 0.4, 8 and 28 microM, respectively. These results imply that ORL1 is a G-protein-coupled receptor with functional as well as structural homology to opioid receptors. In addition, opioid receptor ligands may serve as starting templates for the development of ORL1 specific ligands.


Subject(s)
Opioid Peptides/pharmacology , Receptors, Opioid/agonists , Animals , CHO Cells/drug effects , CHO Cells/metabolism , Cricetinae , Cyclic AMP/metabolism , Dithiothreitol/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Nociceptin Receptor , Nociceptin
14.
Horm Metab Res ; 28(12): 653-8, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9013736

ABSTRACT

Brain and whole body localization and distribution of 125I-leptin was determined after intraperitoneal administration to ob/ob and db/db mice, and was compared to inhibition of food intake. Food intake was not significantly inhibited at3 hours post-injection, but was decreased significantly at 6 h (p < 0.0007) and 24 h (p < 0.02) in ob/ob mice, times at which > 97 % of the radioactive dose was found in the urine. The highest concentrations of 125I-leptin at all time-points were found in the serum, liver and kidneys. These findings were verified by whole body autoradiography. Virtually no 125I-leptin was found in the CNS at later timepoints in either ob/ob or db/db mice. Coronal sectioning of entire brains from ob/ob and db/db mice revealed 125I radioactivity localized to the choroid plexus and in the ventricular space, but not in other CNS regions. No differences in localization, accumulation, or clearance of 125I-leptin in ob/ob vs. db/db mice were found in any of the tissues studied. The present studies demonstrate that the inhibitory effect of leptin on food intake in the ob/ob mouse persists for up to 24 hours after a single dose, despite the complete degradation and elimination of the labeled leptin during the first several hours after injection.


Subject(s)
Eating/drug effects , Obesity/metabolism , Proteins/pharmacology , Proteins/pharmacokinetics , Adipose Tissue/metabolism , Animals , Autoradiography , Brain/metabolism , Cerebral Ventricles/metabolism , Choroid Plexus/metabolism , Female , Humans , Injections, Intraperitoneal , Intestinal Mucosa/metabolism , Iodine Radioisotopes , Kidney/metabolism , Kinetics , Leptin , Liver/metabolism , Mice , Mice, Obese , Proteins/administration & dosage , Tissue Distribution
15.
Horm Metab Res ; 28(12): 694-7, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9013744

ABSTRACT

Leptin, the product of the obese (ob) gene, is a 16 kilodalton protein secreted from adipose tissue. Restoration of leptin to obese ob/ob mice leads to normalization of body weight. The effect of leptin in larger animals has not been explored, in part because of limited supplies of leptin. To date, the potency and yield of recombinant leptin purified from a variety of eukaryotic sources or from E. coli has been highly variable. While purification of leptin from E. coli inclusion bodies has afforded the greatest yield of protein, its potency is at least an order of magnitude lower than that of leptin secreted from E. coli or eukaryotic cells. The mechanistic basis of this difference in potency is not clear at present. The ability to purify significant quantities of highly active leptin will be crucial for the evaluation of leptin structure, as well as its function in additional animal models of obesity. We now report a facile protocol for the preparation of recombinant leptin using an E. coli expression system. 75-85 milligrams of leptin with a purity of greater than 97 % was prepared from a liter of recombinant E. coil. The procedure can be performed in less than 48 h and requires no chromatography. Intraperitoneal injection of 0.1 mg/kg renatured leptin into ob/ob mice results in a significant reduction in food consumption. The potency of this material is similar to the most potent recombinant leptin described to date. The ability to rapidly prepare large quantities of high specific activity material will hasten the definition of leptin's role in non-rodent models of obesity.


Subject(s)
Escherichia coli/chemistry , Proteins/isolation & purification , Animals , Eating/drug effects , Escherichia coli/genetics , Escherichia coli/ultrastructure , Gene Expression , Humans , Injections, Intraperitoneal , Leptin , Macromolecular Substances , Mass Spectrometry , Mice , Mice, Obese , Proteins/genetics , Proteins/pharmacology , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
16.
J Biol Chem ; 271(16): 9455-9, 1996 Apr 19.
Article in English | MEDLINE | ID: mdl-8621615

ABSTRACT

Obese (ob) is a recently identified gene involved in the regulation of energy balance in the mouse. We report here that AD-5075, a potent thiazolidinedione which lowered plasma glucose and triglyceride in Zucker diabetic fatty (ZDF) rats and db/db mice, decreased the expression of the ob gene in these animal models of obesity and non-insulin-dependent diabetes mellitus. The level of adipose ob mRNA in ZDF rats was 3-fold greater than that detected in the Zucker lean littermates. Chronic treatment with AD-5075 elicited a 67 and 70% reduction of ob mRNA in ZDF and control lean rats, respectively. Furthermore, the amount of adipose ob mRNA in db/db mice was 7 times higher than that detected in lean littermates. Treatment of db/db mice with AD-5075 resulted in a 78% reduction of the level of ob mRNA with parallel changes in circulating level of the ob gene product, leptin. The reduction of the ob mRNA in the Zucker lean rats was accompanied by significantly greater food intake and weight gain. However, in ZDF rats and db/db mice, there was profound increase in body weight without hyperphagia. The results demonstrate that the expression of the ob gene is up-regulated in these two rodent models of diabetes compared to their lean counterparts and that such overexpression is attenuated by treatment with an agent that improves insulin sensitivity and glucose homeostasis in vivo.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus/genetics , Gene Expression Regulation/drug effects , Hypoglycemic Agents/pharmacology , Obesity , Protein Biosynthesis , Proteins/genetics , Thiazoles/pharmacology , Thiazolidinediones , Adipose Tissue/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blood Glucose/metabolism , DNA Primers , DNA Probes , DNA, Complementary , Diabetes Mellitus/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Homeostasis/drug effects , Leptin , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Zucker , Reference Values , Triglycerides/blood
17.
Recept Channels ; 4(1): 9-17, 1996.
Article in English | MEDLINE | ID: mdl-8723643

ABSTRACT

Glucagon-like peptide-1 (GLP-1) and glucagon are peptide hormones involved in glucose homeostasis. The ligands are closely related (48% identical) and bind with different affinities to distinct, although highly homologous (47% identical), G protein coupled receptors on the surface of cells. By these criteria, glucagon and GLP-1 receptors can be considered receptor subtypes. A series of chimeric receptors in which 4-6 amino acids in the N-terminal extracellular domain of the human GLP-1 receptor were replaced with the analogous region of the human glucagon receptor were constructed and expressed in COS-7 cells. One of these chimeric receptors, C29-32 displays a 7 to 10-fold decrease in affinity for GLP-1 and the GLP-1 antagonist exendin 9-39 amide and a concomitant 7 to 9-fold increase in its affinity for glucagon. This change in affinity results in a 50-fold decrease in the selectivity of this receptor for GLP-1 over glucagon. Thus, the substitution of as few as four residues of the GLP-1 receptor profoundly affects its selectivity for the homologous peptide agonists GLP-1 and glucagon. These results suggest the extracellular N terminal domain of the GLP-1 receptor harbours molecular determinants for both agonist binding affinity and selectivity.


Subject(s)
Glucagon/metabolism , Peptide Fragments/metabolism , Protein Precursors/metabolism , Receptors, Glucagon/metabolism , Venoms , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , Cyclic AMP/biosynthesis , Exenatide , Glucagon/antagonists & inhibitors , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/antagonists & inhibitors , Peptides/metabolism , Protein Precursors/antagonists & inhibitors , Rats , Receptors, Glucagon/agonists , Receptors, Glucagon/chemistry , Receptors, Glucagon/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
18.
J Biol Chem ; 270(44): 26466-72, 1995 Nov 03.
Article in English | MEDLINE | ID: mdl-7592863

ABSTRACT

The human glucagon receptor was expressed at high density in Drosophila Schneider 2 (S2) cells. Following selection with G418 and induction with CuSO4, the cells expressed the receptor at a level of 250 pmol/mg of membrane protein. The glucagon receptor was functionally coupled to increases in cyclic AMP in S2 cells. Protein immunoblotting with anti-peptide antibodies revealed the expressed receptor to have an apparent molecular mass of 48 kDa, consistent with low levels of glycosylation in this insect cell system. Binding of [fluorescein-Trp25]glucagon to S2 cells expressing the glucagon receptor was monitored as an increase in fluorescence anisotropy along with an increase in fluorescence intensity. Anisotropy data suggest that the mobility of the fluorescein is restricted when the ligand is bound to the receptor. Kinetic analysis indicates that the binding of glucagon to its receptor proceeds via a bimolecular interaction, with a forward rate constant that is several orders of magnitude slower than diffusion-controlled. These data would be consistent with a conformational change upon the binding of agonist to the receptor. The combination of [fluorescein-Trp25]glucagon with the S2 cell expression system should be useful for analyzing glucagon receptor structure and function.


Subject(s)
Glucagon/analogs & derivatives , Receptors, Glucagon/metabolism , Amino Acid Sequence , Animals , Antibodies , Binding, Competitive , Cell Line , Cell Membrane/metabolism , Copper/pharmacology , Copper Sulfate , Drosophila melanogaster , Flow Cytometry , Fluorescence Polarization , Gene Expression/drug effects , Glucagon/metabolism , Humans , Immunoblotting , Kinetics , Mathematics , Models, Theoretical , Molecular Sequence Data , Peptides/chemistry , Peptides/immunology , Receptors, Glucagon/biosynthesis , Receptors, Glucagon/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Transfection
19.
FASEB J ; 9(9): 745-54, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7601339

ABSTRACT

The family of G-protein-coupled receptors can be defined by their similar structural and functional characteristics. Although their primary sequences are quite diverse, these proteins share several common structural features that reflect their common mechanism of action. Mutagenesis and biophysical analysis of several of these receptors indicate that small molecule agonists and antagonists bind to a hydrophobic pocket buried in the transmembrane core of the receptor. In contrast, peptide ligands bind to both the extracellular and transmembrane domains. The mechanisms by which these peptide and small molecule agonists cause receptor activation are being explored by various approaches, but are not yet well defined. A deeper understanding of structural basis for the functional activity of this large family of receptors will have important implications for drug design in a variety of therapeutic areas.


Subject(s)
GTP-Binding Proteins/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Binding Sites , Ligands , Peptides/metabolism , Receptors, Cell Surface/agonists , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/classification
20.
Biochem Biophys Res Commun ; 198(1): 328-34, 1994 Jan 14.
Article in English | MEDLINE | ID: mdl-7507321

ABSTRACT

A human glucagon receptor has been cloned from human liver tissue. The 1578-bp cDNA clone encodes a protein of 477 amino acids with 82% identity to the rat glucagon receptor. The predicted secondary structure and homology to known proteins places this receptor within the superfamily of seven transmembrane domain G protein coupled receptors. Transfection of the human glucagon receptor into COS-7 cells confers upon them high affinity binding for [125I] glucagon. In membranes prepared from COS-7 cells transfected with the human glucagon receptor, the binding of [125I] glucagon is inhibited with the rank order of potency glucagon > oxyntomodulin > glucagon-like peptide 1 (7-36) amide >> glucagon-like peptide 2 = gastric inhibitory peptide = secretin.


Subject(s)
Liver/metabolism , Receptors, Glucagon/biosynthesis , Amino Acid Sequence , Animals , Base Sequence , Binding, Competitive , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Cloning, Molecular/methods , DNA Primers , DNA, Complementary/metabolism , Gene Expression , Glucagon/metabolism , Humans , Kidney , Kinetics , Molecular Sequence Data , Poly A/isolation & purification , Poly A/metabolism , Polymerase Chain Reaction/methods , RNA/isolation & purification , RNA/metabolism , RNA, Messenger , Rats , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...