Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Cell Biochem ; 114(6): 1414-23, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23255434

ABSTRACT

RGS14 is a 60 kDa protein that contains a regulator of G protein signaling (RGS) domain near its N-terminus, a central region containing a pair of tandem Ras-binding domains (RBD), and a GPSM (G protein signaling modulator) domain (a.k.a. Gi/o-Loco binding [GoLoco] motif) near its C-terminus. The RGS domain of RGS14 exhibits GTPase accelerating protein (GAP) activity toward Gαi/o proteins, while its GPSM domain acts as a guanine nucleotide dissociation inhibitor (GDI) on Gαi1 and Gαi3. In the current study, we investigate the contribution of different domains of RGS14 to its biochemical functions. Here we show that the full-length protein has a greater GTPase activating activity but a weaker inhibition of nucleotide dissociation relative to its isolated RGS and GPSM regions, respectively. Our data suggest that these differences may be attributable to an inter-domain interaction within RGS14 that promotes the activity of the RGS domain, but simultaneously inhibits the activity of the GPSM domain. The RBD region seems to play an essential role in this regulatory activity. Moreover, this region of RGS14 is also able to bind to members of the B/R4 subfamily of RGS proteins and enhance their effects on GPCR-activated Gi/o proteins. Overall, our results suggest a mechanism wherein the RBD region associates with the RGS domain region, producing an intramolecular interaction within RGS14 that enhances the GTPase activating function of its RGS domain while disfavoring the negative effect of its GPSM domain on nucleotide dissociation.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , RGS Proteins/physiology , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Enzyme Activation , Humans , Muscarinic Antagonists/pharmacology , Protein Binding , Protein Interaction Domains and Motifs , RGS Proteins/chemistry , Receptor, Muscarinic M2/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Sf9 Cells , Signal Transduction , Tropicamide/pharmacology
2.
Cell Signal ; 23(8): 1375-86, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21447383

ABSTRACT

RGS2 (regulator of G protein signaling 2) is known to limit signals mediated via Gq- and Gs-coupled GPCRs (G protein coupled receptors), and it has been implicated in the differentiation of several cells types. The physiology of RGS2 knockout mice (rgs2(-/-)) has been studied in some detail, however, a metabolic phenotype has not previously been reported. We observed that old (21-24month) rgs2(-/-) mice weigh much less than wild-type C57BL/6 controls, and exhibit greatly reduced fat deposits, decreased serum lipids, and low leptin levels. Lower weight was evident as early as four weeks and continued throughout life. Younger adult male rgs2(-/-) mice (4-8months) were found to show similar strain-related differences as the aged animals, as well improved glucose clearance and insulin sensitivity, and enhanced beta-adrenergic and glucagon signaling in isolated hepatocytes. In addition, rgs2(-/-) pre-adipocytes had reduced levels of differentiation markers (Peroxisome proliferator-activated receptor γ (PPARγ); lipoprotein lipase (Lpl); CCAAT/enhancer binding protein α (CEBPα)) and also rgs2(-/-) white adipocytes were small relative to controls, suggesting altered adipogenesis. In wild-type animals, RGS2 mRNA was decreased in brown adipose tissue after cold exposure (7 h at 4 °C) but increased in white adipose tissue in response to a high fat diet, also suggesting a role in lipid storage. No differences between strains were detected with respect to food intake, energy expenditure, GPCR-stimulated lipolysis, or adaptive thermogenesis. In conclusion this study points to RGS2 as being an important regulatory factor in controlling body weight and adipose function.


Subject(s)
Aging , RGS Proteins/physiology , Weight Gain , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cold Temperature , Energy Metabolism , Female , Glucose/metabolism , Hepatocytes/metabolism , Insulin/metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/genetics , PPAR gamma/metabolism , RGS Proteins/deficiency , RGS Proteins/genetics , Receptors, Adrenergic, beta/metabolism , Signal Transduction
3.
Cell Signal ; 22(8): 1231-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20362664

ABSTRACT

The chronic stimulation of certain G protein-coupled receptors promotes cardiomyocyte hypertrophy and thus plays a pivotal role in the development of human heart failure. The beta-adrenergic receptors (beta-AR) are unique among these in that they signal via Gs, whereas others, such as the alpha1-adrenergic (alpha1-AR) and endothelin-1 (ET-1) receptors, predominantly act through Gq. In this study, we investigated the potential role of regulator of G protein signalling 2 (RGS2) in modulating the hypertrophic effects of the beta-AR agonist isoproterenol (ISO) in rat neonatal ventricular cardiomyocytes. We found that ISO-induced hypertrophy in rat neonatal ventricular myocytes was accompanied by the selective upregulation of RGS2 mRNA, with little or no change in RGS1, RGS3, RGS4 or RGS5. The adenylyl cyclase activator forskolin had a similar effect suggesting that it was mediated through cAMP production. To study the role of RGS2 upregulation in beta-AR-dependent hypertrophy, cardiomyocytes were infected with adenovirus encoding RGS2 and assayed for cell growth, markers of hypertrophy, and beta-AR signalling. ISO-induced increases in cell surface area were virtually eliminated by the overexpression of RGS2, as were increases in alpha-skeletal actin and atrial natriuretic peptide. RGS2 overexpression also significantly attenuated ISO-induced extracellular signal-regulated kinases 1 and 2 (ERK1/2) and Akt activation, which may account for, or contribute to, its observed antihypertrophic effects. In contrast, RGS2 overexpression significantly activated JNK MAP kinase, while decreasing the potency but not the maximal effect of ISO on cAMP accumulation. In conclusion, the present results suggest that RGS2 negatively regulates hypertrophy induced by beta-AR activation and thus may play a protective role in cardiac hypertrophy.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Myocytes, Cardiac/metabolism , RGS Proteins/metabolism , Signal Transduction , Animals , Cell Enlargement , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelin-1/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Heart Ventricles/cytology , Heart Ventricles/growth & development , Isoproterenol/pharmacology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/enzymology , Rats , Rats, Sprague-Dawley
4.
Mol Cell Endocrinol ; 286(1-2): 26-34, 2008 May 14.
Article in English | MEDLINE | ID: mdl-18243519

ABSTRACT

Somatostatin (SRIF) and cortistatin (CST) are two endogenous peptides with high sequence similarities that act as hormones/neurotransmitters both in the CNS and the periphery; their genes although distinct result from gene duplication. Their receptors appear to be common, since the five known SRIF receptors (sst1-sst5) have similar subnanomolar affinity for SRIF and CST, whether the short (SRIF-14, CST-14, CST-17) or the long versions (SRIF-28, CST-29) of the peptides. Whether CST targets specific receptors not shared by SRIF, is still debated: MrgX2 has been described as a selective CST receptor, with submicromolar affinity for CST but devoid of affinity for SRIF; however the distribution of CST and MrgX2 is largely different, and there is no MrgX2 in rodents. A similar situation arises with the GHS receptor GHS-R1a, which displays some preferential affinity for CST over SRIF, but for which there is no evidence that it is activated by CST in vivo. In both cases, one may argue that submicromolar affinity is not the norm of a GPCR for its endogenous neuropeptide. On the other hand, all receptors known to bind SRIF have similar high affinity for CST and both peptides act as potent agonists at the sst1-sst5 receptors, whichever transduction pathway is considered. In addition, [(125)I][Tyr(10)]CST(14) labels sst1-sst5 receptors with subnanomolar affinity, and [(125)I][Tyr(10)]CST(14) binding in the brain is overlapping with that of [(125)I][Tyr(0)]SRIF(14). The functional differences reported that distinguish CST from SRIF, have not been explained convincingly and may relate to ligand-driven transductional selectivity, and other complicating factors such as receptor dimerisation, (homo or heterodimerisation), and/or the influence of accessory proteins (GIPs, RAMPS), which remain to be studied in more detail.


Subject(s)
Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Ghrelin/metabolism , Receptors, Neuropeptide/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Animals , Binding, Competitive , Humans , Nerve Tissue Proteins/agonists , Protein Multimerization , Radioligand Assay , Receptors, G-Protein-Coupled/agonists , Receptors, Neuropeptide/agonists , Receptors, Somatostatin/agonists
5.
J Biol Chem ; 281(43): 32684-93, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-16950788

ABSTRACT

Regulator of G protein signaling (RGS) proteins limit G protein signals. In this study, we investigated the role of RGS2 in the control of G protein signaling cascades in osteoblasts, the cells responsible for bone formation. Expression of RGS2 was up-regulated in primary cultures of mouse calvarial osteoblasts by parathyroid hormone-related peptide (PTHrP)-(1-34), which stimulates G(s) signaling. RGS2 was also up-regulated by extracellular ATP, which selectively activates G(q), as well as by forskolin and phorbol myristate acetate, which activate targets downstream of G(s) and G(q), respectively. To assess the role of endogenous RGS2, we characterized G(s) and G(q) signaling in osteoblasts derived from wild type and rgs2(-/-) mice. Under control conditions, nucleotide-stimulated calcium release, endothelin-stimulated accumulation of inositol phosphates, and PTHrP-stimulated cAMP accumulation were equivalent in osteoblasts isolated from wild type and rgs2(-/-) mice. Thus, basal levels of endogenous RGS2 do not appear to regulate G(s) or G(q) signaling in osteoblasts. Interestingly, forskolin treatment of wild type but not rgs2(-/-) osteoblasts suppressed both endothelin-stimulated accumulation of inositol phosphates and nucleotide-stimulated calcium release, indicating that up-regulation of RGS2 by G(s) signaling desensitizes G(q) signals. Furthermore, pretreatment with ATP suppressed PTHrP-dependent cAMP accumulation in wild type but not rgs2(-/-) osteoblasts, implying that up-regulation of RGS2 by G(q) signaling desensitizes G(s) signals. Our findings demonstrate that endogenously expressed RGS2 can limit G(s) signaling. Moreover, up-regulation of RGS2 contributes to cross-desensitization of G(s)- and G(q)-coupled signals.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , GTP-Binding Protein alpha Subunits, Gs/physiology , Osteoblasts/metabolism , RGS Proteins/metabolism , Signal Transduction , Up-Regulation , Adenosine Triphosphate/pharmacology , Adenoviridae/genetics , Animals , Cell Culture Techniques , Cells, Cultured , Colforsin/pharmacology , Fluorescent Antibody Technique, Indirect , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Osteoblasts/cytology , Osteoblasts/drug effects , Parathyroid Hormone/pharmacology , Peptide Fragments/pharmacology , Skull/cytology , Tetradecanoylphorbol Acetate/pharmacology
6.
Semin Cell Dev Biol ; 17(3): 390-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16765607

ABSTRACT

Regulators of G-protein signaling (RGS proteins) comprise over 20 different proteins that have been classified into subfamilies on the basis of structural homology. The RZ/A family includes RGSZ2/RGS17 (the most recently discovered member of this family), GAIP/RGS19, RGSZ1/RGS20, and the RGSZ1 variant Ret-RGS. The RGS proteins are GTPase activating proteins (GAPs) that turn off G-proteins and thus negatively regulate the signaling of G-protein coupled receptors (GPCRs). In addition, some RZ/A family RGS proteins are able to modify signaling through interactions with adapter proteins (such as GIPC and GIPN). The RZ/A proteins have a simple structure that includes a conserved amino-terminal cysteine string motif, RGS box and short carboxyl-terminal, which confer GAP activity (RGS box) and the ability to undergo covalent modification and interact with other proteins (amino-terminal). This review focuses on RGS17 and its RZ/A sibling proteins and discusses the similarities and differences among these proteins in terms of their palmitoylation, phosphorylation, intracellular localization and interactions with GPCRs and adapter proteins. The specificity of these RGS protein for different Galpha proteins and receptors, and the consequences for signaling are discussed. The tissue and brain distribution, and the evolving understanding of the roles of this family of RGS proteins in receptor signaling and brain function are highlighted.


Subject(s)
GTP-Binding Proteins/metabolism , RGS Proteins , Signal Transduction/physiology , Animals , Calcium/metabolism , Cysteine/chemistry , GTPase-Activating Proteins/physiology , Humans , Models, Biological , Palmitic Acid/metabolism , Protein Binding , Protein Structure, Tertiary , RGS Proteins/chemistry , RGS Proteins/classification , RGS Proteins/genetics , RGS Proteins/metabolism , RGS Proteins/physiology , Subcellular Fractions/metabolism , Tissue Distribution , Ubiquitin/metabolism
7.
Cell Signal ; 18(5): 579-91, 2006 May.
Article in English | MEDLINE | ID: mdl-16226429

ABSTRACT

The intracellular regulator of G protein signalling (RGS) proteins were first identified as GTPase activating proteins (GAPs) for heterotrimeric G proteins, however, it was later found that they can also regulate G protein-effector interactions in other ways that are still not well understood. There is increasing evidence that some of the effects of RGS proteins occur due to their ability to interact with multiprotein signalling complexes. In this review, we will discuss recent evidence that supports the idea that RGS proteins can bind to proteins other than Galpha, such as G protein coupled receptors (GPCRs, e.g. muscarinic, dopaminergic, adrenergic, angiotensin, interleukin and opioid receptors) and effectors (e.g. adenylyl cyclase, GIRK channels, PDEgamma, PLC-beta and Ca(2+) channels). Furthermore, we will investigate novel RGS binding partners (e.g. GIPC, spinophilin, 14-3-3) that underlie the formation of signalling scaffolds or govern RGS protein availability and/or activity.


Subject(s)
RGS Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , 14-3-3 Proteins/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Adenylyl Cyclases/metabolism , Animals , Calcium Channels/metabolism , Calmodulin/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Guanylate Cyclase/metabolism , Isoenzymes/metabolism , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Phospholipase C beta , Type C Phospholipases/metabolism
8.
Circ Res ; 97(3): 268-76, 2005 Aug 05.
Article in English | MEDLINE | ID: mdl-16002744

ABSTRACT

Cardiac responsiveness to neurohumoral stimulation is altered in congestive heart failure (CHF). In chronic CHF, the left ventricle has become sensitive to serotonin because of appearance of Gs-coupled 5-HT4 receptors. Whether this also occurs in acute CHF is unknown. Serotonin responsiveness may develop gradually or represent an early response to the insult. Furthermore, serotonin receptor expression could vary with progression of the disease. Postinfarction CHF was induced in male Wistar rats by coronary artery ligation with nonligated sham-operated rats as control. Contractility was measured in left ventricular papillary muscles and mRNA quantified by real-time reverse-transcription PCR. Myosin light chain-2 phosphorylation was determined by charged gel electrophoresis and Western blotting. Ca2+ transients in CHF were measured in field stimulated fluo-4-loaded cardiomyocytes. A novel 5-HT2A receptor-mediated inotropic response was detected in acute failing ventricle, accompanied by increased 5-HT2A mRNA levels. Functionally, this receptor dominated over 5-HT4 receptors that were also induced. The 5-HT2A receptor-mediated inotropic response displayed a triphasic pattern, shaped by temporally different activation of Ca2+-calmodulin-dependent myosin light chain kinase, Rho-associated kinase and inhibitory protein kinase C, and was accompanied by increased myosin light chain-2 phosphorylation. Ca2+ transients were slightly decreased by 5-HT2A stimulation. The acute failing rat ventricle is, thus, dually regulated by serotonin through Gq-coupled 5-HT2A receptors and Gs-coupled 5-HT4 receptors.


Subject(s)
Heart Failure/physiopathology , Myocardial Contraction , Receptor, Serotonin, 5-HT2A/physiology , Receptors, Serotonin, 5-HT4/physiology , Acute Disease , Animals , Calcium/metabolism , Calmodulin/antagonists & inhibitors , Calmodulin/physiology , Cardiac Myosins/metabolism , Dose-Response Relationship, Drug , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System , Male , Myocytes, Cardiac/metabolism , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/physiology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT2A/genetics , Receptors, Serotonin, 5-HT4/genetics , Serotonin/pharmacology , rho-Associated Kinases
9.
Br J Pharmacol ; 142(1): 150-60, 2004 May.
Article in English | MEDLINE | ID: mdl-15037513

ABSTRACT

1. Somatostatin (somatotropin release inhibiting factor; SRIF) acts via five G protein-coupled receptors (sst(1)-sst(5)) that modulate multiple cellular effectors. The aim of this study was to compare two functional effects of the human sst(2) receptor stably expressed in CHO-K1 cells in a single experiment using a duplex assay for intracellular calcium and serum response element (SRE)-driven luciferase expression. 2. Intracellular calcium was measured using a fluorometric imaging plate reader II (FLIPR II). SRIF-14 rapidly and transiently increased intracellular calcium with a pEC(50) of 8.74+/-0.03 (n=52). At 5 h after FLIPR II measurements, luciferase expression was determined. SRIF-14 concentration-dependently increased luciferase expression (pEC(50)=9.06+/-0.03, n=52). 3. Natural and synthetic agonist/antagonist ligands for SRIF receptors were tested in the duplex assay. Correlation of agonist potencies and efficacies between the two responses were significant (r(2)=0.83 and 0.90, pEC(50) and E(max), respectively). 4. Pertussis toxin pretreatment reduced SRIF-14/octreotide-mediated intracellular calcium increases by 45-47% and luciferase expression by 95-98%. 5. Thapsigargin pretreatment abolished the SRIF-14/octreotide-mediated intracellular calcium increase but had no effect on luciferase expression. 6. In conclusion, SRIF stimulates an increase in intracellular calcium and SRE-luciferase expression via human sst(2) receptors in CHO-K1 cells. The increase in luciferase is mediated via G(i)/G(o) while intracellular calcium increase is mediated by both G(i)/G(o) proteins and pertussis toxin-insensitive G proteins, and is mainly via release of calcium from intracellular stores. SRIF ligands display a similar recognition profile suggesting that the ligand/receptor/G protein/effector interaction is similar for the two parameters.


Subject(s)
Calcium/metabolism , Intracellular Fluid/metabolism , Luciferases/biosynthesis , Receptors, Somatostatin/physiology , Animals , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Intracellular Fluid/drug effects , Luciferases/genetics , Protein Isoforms/agonists , Protein Isoforms/physiology , Receptors, Somatostatin/agonists , Somatostatin/metabolism , Somatostatin/pharmacology
10.
Br J Pharmacol ; 139(1): 109-21, 2003 May.
Article in English | MEDLINE | ID: mdl-12746229

ABSTRACT

1. The mouse corticotroph tumour cell line AtT-20 is a useful model to investigate the physiological role of native somatostatin (SRIF, Somatotropin release inhibitory factor) receptor subtypes (sst(1) - sst(5)). The objective of this study was to characterise the pharmacological features and the functional effects of SRIF receptors expressed by AtT-20 cells using radioligand binding and cAMP accumulation. 2. [(125)I]LTT-SRIF-28, [(125)I]CGP 23996, [(125)I]Tyr(10)-cortistatin-14 and [(125)I]Tyr(3)-octreotide labelled SRIF receptor binding sites with high affinity and in a saturable manner (B(max)=315, 274, 239 and 206 fmol mg(-1), respectively). [(125)I]LTT-SRIF-28 labels significantly more sites than [(125)I]Tyr(10) -cortistatin-14 and [(125)I]Tyr(3) -octreotide as seen previously in cells expressing pure populations of sst(2) or sst(5) receptors. 3. SRIF analogues displaced the binding of the four radioligands. sst(2/5) receptor-selective ligands showed much higher affinity than sst(1/3/4) receptor-selective ligands. The binding profile of [(125)I]Tyr(3)-octreotide was different from that of [(125)I]LTT-SRIF-28, [(125)I]CGP 23996 and [(125)I]Tyr(10)-cortistatin-14. The sst(5/1) receptor-selective ligand L-817,818 identified two binding sites, one with subnanomolar affinity (sst(5) receptors) and one with micromolar affinity (sst(2) receptors); however, the proportions were different: 70 - 80% high affinity with [(125)I]LTT-SRIF-28, [(125)I]CGP 23996, [(125)I]Tyr(10)-cortistatin-14, but only 20% with [(125)I]Tyr(3)-octreotide. 4. SRIF analogues inhibited the forskolin-stimulated cAMP levels depending on concentration. sst(2/5) receptor-selective ligands were highly potent, whereas sst(1/3/4) receptor-selective ligands had no significant effects. The sst(2) receptor antagonist D-Tyr(8)-CYN 154806 competitively antagonised the effects of SRIF-14 and sst(2) receptor-preferring agonists, but not those of L-817,818. 5. The complex binding properties of SRIF receptor analogues indicate that sst(2) and sst(5) receptors are the predominant SRIF receptors expressed on AtT-20 cell membranes with no or only negligible presence of sst(1), sst(3) and sst(4) receptors. In the functional studies using cAMP accumulation, only sst(2) and sst(5) receptors appear to play a role. However, the "predominant" receptor appears to be the sst(2) receptor, although sst(5) receptors can also mediate the effect, when the ligand is not able to activate sst(2) receptors. This clearly adds flexibility to SRIF-mediated functional effects and suggests that the physiological role of SRIF and its analogues may be mediated preferentially via one subtype over another.


Subject(s)
Receptors, Somatostatin/metabolism , Animals , Binding, Competitive , Cell Line, Tumor , Colforsin/pharmacology , Cyclic AMP/biosynthesis , Mice , Radioligand Assay , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors , Somatostatin/pharmacology
11.
Eur J Pharmacol ; 465(3): 211-8, 2003 Apr 04.
Article in English | MEDLINE | ID: mdl-12681432

ABSTRACT

The availability of antagonist ligands for somatostatin receptors is very limited, with those that are available often displaying agonist properties or limited receptor subtype selectivity. Hay et al. [Bioorg. Med. Chem. Lett. 11 (2001) 2731] recently described the development of small-molecule somatostatin receptor subtype 2 (sst(2)) selective compounds. This study investigates the binding affinity and functional characteristics of two of those antagonists (2 and 3) and the agonist compound, from which they were derived (1). In radioligand binding studies using the agonist radioligands [125I][Tyr(11)]SRIF-14 (Ala-Gly-c[Cys-Lys-Asn-Phe-Phe-Trp-Lys-Thr-(125I-Tyr)-Thr-Ser-Cys]-OH), [125I]LTT-SRIF-28 ([Leu(8),DTrp(22),125I-Tyr(25)]SRIF-28; Ser-Ala-Asn-Ser-Asn-Pro-Ala-Leu-Ala-Pro-Arg-Glu-Arg-Lys-Ala-Gly-c[Cys-Lys-Asn-Phe-Phe-DTrp-Lys-Thr-(125I-Tyr)-Thr-Ser-Cys]-OH), [125I]CGP 23996 (c[Lys-Asu-Phe-Phe-Trp-Lys-Thr-(125I-Tyr)-Thr-Ser]), [125I][Tyr(3)]octreotide (DPhe-c[Cys-(125I-Tyr)-DTrp-Lys-Thr-Cys]-Thr-OH) and [125I][Tyr(10)]cortistatin-14 (Pro-c[Cys-Lys-Asn-Phe-Phe-Trp-Lys-Thr-(125I-Tyr)-Ser-Ser-Cys]-Lys) at human recombinant somatostatin receptors expressed in Chinese hamster lung fibroblast (CCL39) cells and native rat cortex, the compounds bound with high affinity (pK(d) 6.8-9.7) and selectivity to human sst(2) receptors. Some affinity was also observed for sst(5) labelled by [125I][Tyr(3)]octreotide and [125I]CGP 23996. In functional studies at human sst(2) receptors expressed in Chinese hamster ovary (CHO) cells, both the agonist 1 and the two putative antagonists 2 and 3 concentration dependently inhibited forskolin-stimulated adenylate cyclase and stimulated luciferase reporter gene expression, with similar efficacy to the natural ligand somatotropin release inhibiting factor (SRIF)-14. Compound 1 had similar potency to SRIF-14, which was in the nanomolar range, whereas 2 and 3 were 10-100-fold less potent. The intrinsic activity of 2 and 3 was too high to allow antagonist studies to be carried out. In conclusion, in contrast to previous findings, all three compounds are potent agonists at recombinant human sst(2) receptors.


Subject(s)
Octreotide/analogs & derivatives , Octreotide/pharmacology , Receptors, Somatostatin/drug effects , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Cell Line , Cerebral Cortex/metabolism , Colforsin/pharmacology , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Genes, Reporter , In Vitro Techniques , Ligands , Luciferases/biosynthesis , Luciferases/genetics , Radioligand Assay , Rats , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors
12.
Naunyn Schmiedebergs Arch Pharmacol ; 367(1): 1-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12616335

ABSTRACT

The two forms (DTyr8 and LTyr8) of the putative somatostatin sst2 receptor antagonist CYN 154806 (Ac-4NO2-Phe-c(DCys-Tyr-DTrp-Lys-Thr-Cys)-D/LTyr-NH2) were investigated on recombinant human somatostatin receptors and endogenous guinea-pig ileum receptors. In radioligand binding studies using the agonist radioligands [125I]LTT-SRIF-28, [125I][Tyr10]cortistatin-14, [125I]CGP 23996 and [125I][Tyr3]octreotide in Chinese hamster lung fibroblast (CCL39) and Chinese hamster ovary (CHO) cells expressing human somatostatin receptors (hsst1-5), CYN 154806 binds to sst2 receptors with nanomolar affinity (pKD=8.14-8.89), 40- to 4500-fold higher than for sst1, sst3 or sst4. High affinity was also demonstrated for sst5 receptors, particularly for LTyr8CYN 154806 where the sst5 affinity was higher than for sst2 receptors when using [125I]CGP 23996 and [125I][Tyr3]octreotide. Functional properties of the compounds were examined in Chinese hamster ovary (CHO) cells expressing human sst2 receptors, in (1) inhibition of forskolin-stimulated adenylate cyclase, (2) stimulation of serum response element-driven luciferase expression and (3) [35S]guanosine 5'-O-(3-thiotriphosphate) ([35S]GTPS) binding. L- and DTyr8CYN 154806 showed full agonism at inhibition of forskolin-stimulated cAMP accumulation (pEC50=7.73 for both, Emax 104% and 78%, respectively), partial agonism at luciferase expression (pEC50=7.85 and 8.16, Emax=50% and 29%, respectively) and behaved as apparently silent antagonists at [35S]GTPS binding (no agonism observed, pKB=6.88 and 7.50, respectively). The agonist potential was confirmed in isolated guinea-pig ileum preparations via measurement of SRIF-induced inhibition of neurotransmission, where the L-isoform had marked agonism (pEC50=8.23, Emax=32%) whereas the D-isoform was apparently devoid of agonism. The present data suggest that CYN 154806 should be used with caution as an sst2 receptor antagonist tool, since it possesses intrinsic activity at sst2, and high affinity for both sst2 and sst5 receptors. The DTyr form, having lower intrinsic activity, especially in natural tissues, and greater selectivity for sst2 receptors, may be more reliable than LTyr CYN 154806.


Subject(s)
Oligopeptides/pharmacology , Receptors, Somatostatin/antagonists & inhibitors , Receptors, Somatostatin/physiology , Animals , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Guinea Pigs , Humans , Ileum/drug effects , Ileum/metabolism , In Vitro Techniques , Male , Oligopeptides/metabolism , Protein Binding/drug effects , Protein Binding/physiology , Receptors, Somatostatin/agonists
13.
Naunyn Schmiedebergs Arch Pharmacol ; 367(2): 95-103, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12595949

ABSTRACT

Four linear beta(2)/beta(3)-di- and alpha/beta(3)-tetrapeptides (1-4) were investigated as somatostatin sst(4) receptor agonists on recombinant human and mouse somatostatin receptors. Human somatostatin receptor subtypes 1-5 (sst(1-5)), and mouse somatostatin receptor subtypes 1,3,4 and 5, were characterised using the agonist radioligands [(125)I]LTT-SRIF-28, [(125)I][Tyr(10)]CST(14) and [(125)I]CGP 23996 in stably transfected Chinese hamster lung fibroblast (CCL39) cells. The peptides bound selectively to sst(4) receptors with nanomolar affinity (pK(d)=5.4-7.8). The peptides were investigated on second messenger systems both as agonists, and as antagonists to SRIF-14-mediated effects in CCL39 cells expressing mouse sst(4 )receptors, via measurement of inhibition of forskolin-stimulated adenylate cyclase activity, and stimulation of luciferase expression. The peptides showed full agonism or pronounced partial agonism (40 to 100% relative intrinsic activity) in both inhibition of forskolin-stimulated adenylate cyclase activity (pEC(50)=5.5-6.8), and luciferase expression (pEC(50)=5.5-6.5). The agonist potential was confirmed since antagonism was very difficult to establish. The data show that beta(2)/beta(3)-di- and alpha/beta(3)-tetrapeptide derivatives have agonist potential at recombinant somatostatin sst(4) receptors. Therefore, they may be used to elucidate physiological and biochemical effects mediated by sst(4), and may also have potential as therapeutic agents.


Subject(s)
Oligopeptides/pharmacology , Receptors, Somatostatin/agonists , Adenylyl Cyclases/metabolism , Animals , Cells, Cultured , Colforsin/pharmacology , Cricetinae , Cricetulus , Dipeptides/chemical synthesis , Dipeptides/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Ligands , Luciferases/biosynthesis , Lung/cytology , Membrane Proteins , Mice , Oligopeptides/chemical synthesis , Radioligand Assay , Receptors, Somatostatin/metabolism , Structure-Activity Relationship
14.
Mol Cell Endocrinol ; 189(1-2): 105-16, 2002 Mar 28.
Article in English | MEDLINE | ID: mdl-12039069

ABSTRACT

In this study, a somatostatin receptor (Sst) cDNA was cloned and sequenced from goldfish pituitary. The cDNA encodes a 390-amino acid type five-like Sst (designated as gfSst(5)). The amino acid sequence of the receptor has slightly higher homology to mammalian Sst(5'), compared with other mammalian Sst subtypes and recently identified fish Sst(1), Sst(2) and Sst(3). In CCL39-SRE-Luci cells stably expressing the cloned receptor, agonist radioligand [125I]LTT-SRIF(28'), a mammalian SRIF(28) analog, bound to a homogenous population of receptors with high affinity (nM K(d)). Competition binding studies showed that all three natural goldfish SRIF ligands, SRIF(14), [Pro(2)]SRIF(14), and goldfish SRIF(28) (gfSRIF(28)), and LTT-SRIF(28) bind the cloned gfSst(5) with high affinity and significantly stimulate [35S]GTPgammaS binding, with SRIF(28) peptides showing higher affinity in receptor binding and potency in [35S]GTPgammaS binding compared with SRIF(14) peptides. The receptor gene is highly and predominately expressed in pituitary; lower levels of the receptor mRNA were also detected in different brain regions by reverse transcriptor-polymerase chain reaction (RT-PCR) followed by Southern blot analysis.


Subject(s)
Goldfish/physiology , Pituitary Gland/metabolism , Receptors, Somatostatin/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , Goldfish/genetics , Humans , Molecular Sequence Data , Phylogeny , RNA, Messenger/metabolism , Radioligand Assay , Receptors, Somatostatin/classification , Receptors, Somatostatin/genetics , Sequence Alignment
15.
J Mol Neurosci ; 18(1-2): 15-27, 2002.
Article in English | MEDLINE | ID: mdl-11931345

ABSTRACT

Somatostatin (SRIF, somatotropin release inhibiting factor), discovered for its inhibitory action on growth hormone (GH) secretion from pituitary, is an abundant neuropeptide. Two forms, SRIF14 and SRIF28 exist. Recently, a second family of peptides with very similar sequences and features was described; the cortistatins (CST), CST17 and CST29 which are brain selective. The five cloned SRIF receptors (sst1-5) belong to the G-protein coupled/ heptathelical receptor family. Structural and operational features distinguish two classes of receptors; SRIF1 - sst2/sst3/sst5 (high affinity for octreotide or seglitide) and SRIF2 = sst1/sst4(very low affinitty for the aforementioned ligands). The affinity of SRIF receptors for somatostatins and cortistatins is equally high, and it is not clear whether selective receptors do exist for one or the other of the peptides. Several radiologlands label all SRIF receptors, e.g., [125]LTT-SRIF28' [l25I]CGP23996, [125]Tyr10cortistatin or [125I]Tyr11SRIF14. In contrast, [125I]Tyr3octreotide, [125I]BIM23027, [125I]MK678 or [125I]D-Trp8SRIF14 label predominantly SRIF1 sites, especially sst2 and possibly sst5 receptors. In brain, [125I]Tyr3octreotide binding equates with sst2 receptor mRNA distribution. Native SRIF2receptors can be labeled with [125I]SRIF14 in the presence of high NaCl in brain (sst1) or lung (sst4) tissue. Short cyclic or linear peptide analogs show selectivity for sst2/sst5 (octreotide, lanreotide, BIM 23027), sst1 (CH-275), sst3 (sst3-ODN-8), or sst5 receptors (BIM 23268); although claims for selectivity have not always been confirmed. Beta peptides ith affinity for SRIF receptors are also reported. The general lack of SRIF receptor antagonists is unique for peptide receptors, although CYN 154806 is a selective and potent sst2 antagonist. Nonpeptide ligands are still rare, although a number of molecules have been reported with selectivity and potency for sst1 (L 757,519), sst2 (L 779,976), sst3 (L 796,778), sst4 (NNC 26-9100, L 803,087) or sst1/sst5 receptors (L 817,018). Such molecules are essential to establish the role of SRIF receptors, e.g., sst1 in hypothalamic glutamate currents: sst2 in inhibiting release of GH, glucagon, TSH, gastric acid secretion, pain, seizures and tumor growth, and sst5 in vascular remodeling and inhibition of insulin and GH release.


Subject(s)
Brain/metabolism , Drug Design , Hypothalamo-Hypophyseal System/metabolism , Neuropeptides/metabolism , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors , Somatostatin/analogs & derivatives , Animals , Binding, Competitive/drug effects , Binding, Competitive/physiology , Brain/drug effects , Humans , Hypothalamo-Hypophyseal System/drug effects , Ligands , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Receptors, Somatostatin/metabolism , Somatostatin/metabolism
16.
Eur J Pharmacol ; 436(3): 173-86, 2002 Feb 02.
Article in English | MEDLINE | ID: mdl-11858797

ABSTRACT

Somatostatin (somatotropin release inhibiting factor, SRIF), exerts its effects via specific G protein coupled receptors of which five subtypes have been cloned (sst1-5). Recently, SRIF receptors have also been cloned from fish tissues. In this study, goldfish sst5 receptors (gfsst5) were expressed and characterised in the Chinese hamster lung fibroblast cell line, that harbours the luciferase reporter gene driven by the serum responsive element (CCL39-SRE-Luci). The agonist radioligands [125I]-LTT-SRIF-28 ([Leu8, DTrp22, 125I-Tyr25]SRIF-28) and [125I][Tyr10]cortistatin-14 labelled similar receptor densities with high affinity and in a saturable manner (pKd: 9.99-9.71; Bmax: 300-350 fmol mg-1). 5'-Guanylyl-imidodiphosphate inhibited radioligand binding to some degree (38.5-57.9%). In competition binding studies, the pharmacological profile of SRIF binding sites defined with [125I]LTT-SRIF-28 and [125I][Tyr10]cortistatin-14 correlated significantly (r2=0.97, n=20). Pharmacological profiles of human and mouse sst5 receptors expressed in CCL39 cells correlated markedly less with those of the gfsst5 profile (r2=0.52-0.78, n > or = b16). Functional expression of the gfsst5 receptor was examined by measurement of agonist-induced luciferase expression and stimulation of [35S]GTPgammaS ([35S]guanosine 5'-O-(3-thiotriphosphate) binding. Profiles were similar to those achieved in radioligand binding studies (r2=0.81-0.93, n=20), although relative potency (pEC50) was reduced compared to pKd values. Relative efficacy profiles of luciferase expression and [35S]GTPgammaS binding, were rather divergent (r2=0.48, n=20) with peptides showing full agonism at one pathway and absence of agonism at the other. BIM 23056 (D-Phe-Phe-Tyr-D-Trp-Lys-Val-Phe-D-Nal-NH2) acted as an antagonist on the effects of SRIF-14 (pKB=6.74 +/- 0.23) on stimulation of [35S]GTPgammaS binding. Pertussis toxin abolished the effect of SRIF-14 on luciferase expression and [35S]GTPgammaS binding suggesting coupling of the receptor to G(i)/G(o) proteins. In summary, the present studies demonstrate that the gfsst5 receptor has a similar pharmacological profile and transductional properties to mammalian sst5 receptors. The difference in efficacy profiles defined using different functional assays suggests numerous, agonist specific, conformational receptor states, and/or ligand-dependent receptor trafficking.


Subject(s)
Receptors, Somatostatin/drug effects , Receptors, Somatostatin/metabolism , Animals , Binding, Competitive/drug effects , Cell Line , Dose-Response Relationship, Drug , Goldfish , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Ligands , Luciferases/drug effects , Luciferases/genetics , Luciferases/metabolism , Neuropeptides/metabolism , Neuropeptides/pharmacology , Radioligand Assay , Receptors, Somatostatin/genetics , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Somatostatin/metabolism , Somatostatin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...