Pharmacological characterisation of the goldfish somatostatin sst5 receptor

https://doi.org/10.1016/S0014-2999(01)01626-0Get rights and content

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≥16). Functional expression of the gfsst5 receptor was examined by measurement of agonist-induced luciferase expression and stimulation of [35S]GTPγS ([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]GTPγS 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]GTPγS binding. Pertussis toxin abolished the effect of SRIF-14 on luciferase expression and [35S]GTPγS binding suggesting coupling of the receptor to Gi/Go 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.

Introduction

Somatostatin or SRIF (somatotropin release inhibiting factor) is a hormone/neuropeptide which is found at high levels in the central and peripheral nervous system, and a number of peripheral tissues. It is involved in the regulation of multiple physiological processes, including endocrine and exocrine secretions, neurotransmission, neuromodulation of transmitter release, smooth muscle motility and cell proliferation, especially in tumours Reichlin, 1983, Patel, 1997.

In mammals, SRIF is found in two forms—SRIF-14, and the N-terminally extended SRIF-28. These two peptides are produced from a single gene which encodes preprosomatostatin (PSS); this is differentially cleaved to either SRIF-14 or SRIF-28 Pradayrol et al., 1980, Patel and O'Neil, 1988, Patel and Galanopoulou, 1995. SRIF-14 is highly conserved throughout evolution, being found in all mammalian species studied and in representatives from all vertebrate classes including birds, amphibians, reptiles and fish (Lin et al., 2000b). Although rare, variants of SRIF-14 are also found in some species including [Ser12]SRIF-14 in Sea Lamprey (Andrews et al., 1988), [Ser5]SRIF-14 in Pacific Ratfish (Conlon, 1990), [Pro2,Met13]SRIF-14 in European Green Frog (Vaudry et al., 1992) and [Pro2]SRIF-14 in Russian Sturgeon, African Lungfish and Goldfish Nishii et al., 1995, Lin et al., 1999b, Trabucchi et al., 1999.

Recently, it has been shown that SRIF is part of a multigene family. A second SRIF precursor gene (PSS-II) has been identified in teleost fish, that is processed to SRIF peptides of 28, 25, or 14 amino acids in length with [Tyr7,Gly10]SRIF-14 (or a variant of this depending on the species) at their C terminus Conlon et al., 1987, Zupanc et al., 1999, Lin et al., 2000b. In frog brain, a second PSS gene has been identified which is processed to [Pro2,Met13]SRIF-14 (Vaudry et al., 1992). A prepropeptide cDNA with structural similarity to that of preprosomatostatin has also been cloned in mammals. This is cleaved to produce cortistatin which is found in rat, mouse and human brain in various forms and has a 14 amino acid peptide at its C-terminus with 11 amino acids identical to SRIF-14 De Lecea et al., 1996, De Lecea et al., 1997, Fukusumi et al., 1997. Goldfish have three distinct SRIF precursor genes. These are termed PSS-I, PSS-II and PSS-III and are processed to SRIF-14, goldfish SRIF-28 which has [Glu1, Tyr7, Gly10]SRIF-14 at its C terminus, and [Pro2]SRIF-14, respectively Lin et al., 1999b, Lin et al., 2000b.

SRIF produces its effects by binding to high affinity membrane bound G protein-coupled receptors Schonbrunn and Tashjian, 1978, Jakobs et al., 1983, Bell and Reisine, 1993 of which five subtypes (sst1–5) have been identified from several mammalian species Bell and Reisine, 1993, Hoyer et al., 1994, Hoyer et al., 1995. The receptors display a subtype-specific distribution pattern in brain and peripheral tissues Bell and Reisine, 1993, Patel, 1997, Selmer et al., 2000. All five receptor subtypes bind SRIF-14 and SRIF-28 with high affinity, but differ in their ability to bind the short-chain synthetic analogues octreotide, seglitide and somatuline, producing two distinct subclasses according to pharmacology and structure: sst2, sst3 and sst5, with high affinity for the short analogues and sst1 and 4, with low affinity (Hoyer et al., 1995).

In goldfish, cDNAs for two receptors corresponding to mammalian sst1, one corresponding to mammalian sst2 and one to mammalian sst5 have been cloned (Lin et al., 1999a, Lin et al., 2000a; submitted). The sst1 receptors (termed sst1A and sst1B) have 98% homology in their amino acid sequences and are thought to be the product of duplicate genes rather than spliced variants (Lin et al., 1999a). An sst3 subtype (fsst3) has been isolated in an electric fish Apteronotus albifrons Siehler et al., 1999b, Zupanc et al., 1999.

In the present study, the newly cloned goldfish sst5 (gfsst5) receptor (Lin et al., 2002, in press) was expressed in Chinese hamster lung fibroblast cells (CCL39), and its pharmacological features examined and compared to those of human and mouse sst5 using radioligand binding studies, and stimulation of [35S]guanosine 5′-O-(3-thiotriphosphate ([35S]GTPγS) binding and coupling via the serum responsive element to luciferase expression.

Section snippets

Cell culture

CCL39-SRE-Luci cells (established line of Chinese hamster lung fibroblasts; American Type Culture Collection), were cultured in a 1:1 mixture of Dulbecco's Modified Eagle's Medium (DMEM; Seromed, Biochrom, Berlin, Germany: 3.7 g l−1 NaHCO3, 1.0 g l−1 d-glucose, with stable glutamine) and Ham's F-12 Nutrient Mixture (Seromed: 1.176 g l−1 NaHCO3, with stable glutamine), supplemented with 10% (v/v) foetal bovine serum (Gibco BRL) and Hygromycin B (100 μg ml−1) (Calbiochem, La Jolla, CA, USA) at 37

Radioligand binding

[125I]LTT-SRIF-28 and [125I][Tyr10]cortistatin-14 labelled gfsst5 binding sites in CCL39-SRE-Luci cells with high affinity and in a saturable manner ([125I]LTT-SRIF-28: Bmax=303±15 fmol mg−1, pKd=9.99±0.19; [125I][Tyr10]cortistatin-14: Bmax=348±23 fmol mg−1, pKd=9.71±0.08) (Fig. 1). There was no significant difference between the sites labelled by the two radioligands (Data analysed using independent t-tests, P: 0.227 and 0.150; n=4 for pKd and Bmax, respectively). Non-specific binding was

Discussion

The somatostatin neuropeptide family exerts its effects by binding to G protein-coupled receptors of which five subtypes have been cloned from several mammalian species and sst1A, sst1B, sst2, sst3 and sst5 receptors have been cloned from teleost fish species (Bell and Reisine, 1993, Hoyer et al., 1994, Hoyer et al., 1995, Siehler et al., 1999b, Lin et al., 2002). In this study, a type 5 somatostatin receptor isolated from goldfish has been expressed and pharmacologically characterised in

Acknowledgements

Work supported in part by EC Contract QLG3-CT-1999-00908 and Swiss grant BBW 00-0427.

References (73)

  • D.A Horstman et al.

    An aspartate conserved among G-protein receptors confers allosteric regulation of alpha 2-adrenergic receptors by sodium

    J. Biol. Chem.

    (1990)
  • D Hoyer et al.

    Classification and nomenclature of somatostatin receptors

    Trends Pharmacol. Sci.

    (1995)
  • R.J Lefkowitz et al.

    Constitutive activity of receptors coupled to guanine nucleotide regulatory proteins

    Trends Pharmacol. Sci.

    (1993)
  • X Lin et al.

    Molecular cloning and expression of a type-two somatostatin receptor in goldfish brain and pituitary

    Mol. Cell. Endocrinol.

    (2000)
  • C Murga et al.

    Novel molecular mediators in the pathway connecting G-protein-coupled receptors to MAP kinase cascades

    Trends Endocrinol. Metab.

    (1999)
  • A Newman-Tancredi et al.

    Agonist and inverse agonist efficacy at human recombinant serotonin 5-HT1A receptors as a function of receptor:G-protein stoichiometry

    Neuropharmacology

    (1997)
  • M Nishii et al.

    Isolation and characterization of [Pro2]somatostatin-14 and melanotropins from Russian sturgeon, Acipenser gueldenstaedti Brandt

    Gen. Comp. Endocrinol.

    (1995)
  • Y.C Patel et al.

    Peptides derived from cleavage of prosomatostatin at carboxyl- and amino-terminal segments. Characterization of tissue and secreted forms in the rat

    J. Biol. Chem.

    (1988)
  • Y.C Patel et al.

    All five cloned human somatostatin receptors (hSSTR1–5) are functionally coupled to adenylyl cyclase

    Biochem. Biophys. Res. Commun.

    (1994)
  • P.J Pauwels et al.

    How efficacious are 5-HT1B/D receptor ligands: an answer from GTP gamma S binding studies with stably transfected C6-glial cell lines

    Neuropharmacology

    (1997)
  • L Pradayrol et al.

    N-terminally extended somatostatin: the primary structure of somatostatin-28

    FEBS Lett.

    (1980)
  • T Reisine

    Pertussis toxin in the analysis of receptor mechanisms

    Biochem. Pharmacol.

    (1990)
  • P Samama et al.

    A mutation-induced activated state of the beta 2-adrenergic receptor. Extending the ternary complex model

    J. Biol. Chem.

    (1993)
  • P Schoeffter et al.

    Inhibition of cAMP accumulation via recombinant human serotonin 5-HT1A receptors: considerations on receptor effector coupling across systems

    Neuropharmacology

    (1997)
  • A Schonbrunn et al.

    Characterization of functional receptors for somatostatin in rat pituitary cells in culture

    J. Biol. Chem.

    (1978)
  • I.S Selmer et al.

    Advances in understanding neuronal somatostatin receptors

    Regul. Pept.

    (2000)
  • S Siehler et al.

    [125I][Tyr3]octreotide labels human somatostatin sst2 and sst5 receptors

    Eur. J. Pharmacol.

    (1998)
  • S Siehler et al.

    Characterization of the fish sst3 receptor, a member of the SRIF1 receptor family: atypical pharmacological features

    Neuropharmacology

    (1999)
  • L.J Sim-Selley et al.

    Inhibitory effects of SR141716A on G-protein activation in rat brain

    Eur. J. Pharmacol.

    (2001)
  • J.A Stanton et al.

    Characterisation of a cloned human 5-HT1A receptor cell line using [35S]GTP gamma S binding

    Eur. J. Pharmacol.

    (1997)
  • H Vaudry et al.

    Isolation of [Pro2,Met13]somatostatin-14 and somatostatin-14 from the frog brain reveals the existence of a somatostatin gene family in a tetrapod

    Biochem. Biophys. Res. Commun.

    (1992)
  • Y Yamada et al.

    Cloning, functional expression and pharmacological characterization of a fourth (hSSTR4) and a fifth (hSSTR5) human somatostatin receptor subtype

    Biochem. Biophys. Res. Commun.

    (1993)
  • G.K.H Zupanc et al.

    Molecular cloning and pharmacological characterization of a somatostatin receptor subtype in the gymnotiform fish Apteronotus albifrons

    Gen. Comp. Endocrinol.

    (1999)
  • H Baumeister et al.

    Cloning, expression, pharmacology and tissue distribution of the mouse somatostatin receptor subtype 5

    J. Neuroendocrinol.

    (1998)
  • K.A Berg et al.

    Effector pathway-dependent relative efficacy at serotonin type 2A and 2C receptors: evidence for agonist-directed trafficking of receptor stimulus

    Mol. Pharmacol.

    (1998)
  • K.A Berg et al.

    Novel actions of inverse agonists on 5-HT2C receptor systems

    Mol. Pharmacol.

    (1999)
  • Cited by (22)

    • Somatostatin receptor-mediated suppression of gabaergic synaptic transmission in cultured rat retinal amacrine cells

      2014, Neuroscience
      Citation Excerpt :

      Effects of SRIF on the sIPSCs in the amacrine cells were first tested. As shown in Fig. 1B, external perfusion of 1 μM SRIF for 5 min significantly suppressed the sIPSCs, which was partially reversed by adding BIM (1 μM), a specific sst5 receptor antagonist (Wilkinson et al., 1996; Nunn et al., 2002). The cumulative probability curve of inter-event interval of these events in the presence of SRIF, as compared to that obtained in the control condition, was significantly shifted toward the right, indicating a decrease in sIPSC frequency (Fig. 1C).

    • Signalling pathway of goldfish melanin-concentrating hormone receptors 1 and 2

      2011, Regulatory Peptides
      Citation Excerpt :

      Consistent with the present study, we obtained preliminary data using a calcium mobilization assay that barfin flounder MCHR1 and MCHR2 have the same G protein-coupling system as goldfish receptors. To the best of our knowledge regarding studies on heterologous protein-expressing cells, MCHRs may be the first example of such a difference in peptide receptor-mediated signalling pathways between mammals and teleosts fish [43–45]. This observed feature of MCHRs is of particular interest, and may indicate that the MCHR-G protein interactions have changed during evolution from fish species to humans.

    • Goldfish brain somatostatin-28 differentially affects dopamine- and pituitary adenylate cyclase-activating polypeptide-induced GH release and Ca<sup>2+</sup> and cAMP signals

      2011, Molecular and Cellular Endocrinology
      Citation Excerpt :

      How these different inhibitory actions of SS-14 and gbSS-28 are manifested is unknown but the use of different SS receptor subtypes (Ssts) is a distinct possibility. For instance, SS-14 activation of goldfish Sst2 results in decreases in cAMP via Gi protein, whereas gbSS-28 attenuates cAMP formation via binding to Sst5 rather than Sst2 (Lin et al., 2000; Nunn et al., 2002). Interestingly, Sst signalling in mammals can also be altered as a consequence of these receptors physically interacting with other families of G-protein-coupled receptors, including DA receptors (Duran-Prado et al., 2008).

    • Involvement of protein kinase C and intracellular Ca<sup>2+</sup> in goldfish brain somatostatin-28 inhibitory action on growth hormone release in goldfish

      2010, General and Comparative Endocrinology
      Citation Excerpt :

      SS-14 and [Pro2]SS-14 bind goldfish Sst2s expressed in COS-7 cells with similar affinity and exhibit similar potency in inhibiting adenylate cyclase. On the other hand, gbSS-28 has no effect on forskolin-stimulated cAMP production in COS-7 cells expressing Sst2s but the gbSS-28-selective Sst5s expressed in CCL39 cells are coupled to Gi/o-protein (Lin et al., 2000; Nunn et al., 2002). Interestingly, gbSS-28 is the most potent among the three goldfish SSs in suppressing basal GH secretion, but it is less effective than SS-14 in inhibiting GnRH-induced GH release in static incubation of dispersed goldfish pituitary cells (Yunker et al., 2003).

    • Somatostatin and somatostatin receptors in fish growth

      2010, General and Comparative Endocrinology
      Citation Excerpt :

      As in mammals, fish possess multiple SSTR subtypes; to date four distinct subtypes have been characterized (SSTR 1–3 and 5) (Volkoff et al., 2005; Klein and Sheridan, 2008). Ligand-selective binding characteristics have been reported for some SSTRs of trout and goldfish (Nunn et al., 2002; Gong et al., 2004; Siehler et al., 2008). Knowledge of the mechanisms of SS action in fish is limited; however, cAMP, Ca2+/PKC, ERK, and PI3K/Akt appear to be involved (Yunker and Chang, 2001, 2004; Hagemeister and Sheridan, 2008).

    View all citing articles on Scopus
    View full text