Unacylated ghrelin is not a functional antagonist but a full agonist of the type 1a growth hormone secretagogue receptor (GHS-R)

https://doi.org/10.1016/j.mce.2007.05.010Get rights and content

Abstract

Recent findings demonstrate that the effects of ghrelin can be abrogated by co-administered unacylated ghrelin (UAG). Since the general consensus is that UAG does not interact with the type 1a growth hormone secretagogue receptor (GHS-R), a possible mechanism of action for this antagonistic effect is via another receptor. However, functional antagonism of the GHS-R by UAG has not been explored extensively. In this study we used human GHS-R and aequorin expressing CHO-K1 cells to measure [Ca2+]i following treatment with UAG. UAG at up to 10−5 M did not antagonize ghrelin induced [Ca2+]i. However, UAG was found to be a full agonist of the GHS-R with an EC50 of between 1.6 and 2 μM using this in vitro system. Correspondingly, UAG displaced radio-labeled ghrelin from the GHS-R with an IC50 of 13 μM. In addition, GHS-R antagonists were found to block UAG induced [Ca2+]i with approximately similar potency to their effect on ghrelin activation of the GHS-R, suggesting a similar mode of action. These findings demonstrate in a defined system that UAG does not antagonize activation of the GHS-R by ghrelin. But our findings also emphasize the importance of assessing the concentration of UAG used in both in vitro and in vivo experimental systems that are aimed at examining GHS-R independent effects. Where local concentrations of UAG may reach the high nanomolar to micromolar range, assignment of GHS-R independent effects should be made with caution.

Introduction

Ghrelin was discovered through its ability to activate the type 1a growth hormone secretagogue receptor (GHS-R) and stimulate growth hormone release in vivo (Smith et al., 2005, van der Lely et al., 2004). An evolutionarily conserved feature of ghrelin is the acylation of its third residue, usually with n-octanoic and, less commonly, with n-decanoic acid (Hosoda et al., 2003). Kojima et al. (1999) were the first to describe the requirement that ghrelin be acylated on its third serine residue for activation of the GHS-R in the nanomolar range, with an EC50 for increased [Ca2+]i of 2.5 × 10−9 M. In the circulation, ghrelin also occurs as a unacylated isoform (UAG) at 10–50 times the concentration of acylated ghrelin (Kojima and Kangawa, 2005, and our unpublished observations).

Of great interest to us has been the finding that in humans co-administration of UAG can antagonize the metabolic effects of ghrelin in vivo. Ghrelin administration causes hyperglycemia, hypoinsulinemia, increased circulating free fatty acids and worsening insulin sensitivity, but these effects are reversed or prevented by co-administration with UAG (Broglio et al., 2004, Gauna et al., 2004). These effects seem to be specific to ghrelin's metabolic activity since UAG has no impact on GH, PRL or ACTH secretion (Broglio et al., 2004). This suggested a direct action on the endocrine pancreas, and perhaps on hepatic glucose production. In relation to these in vivo findings, we have shown that UAG not only suppresses glucose output, but also blocks ghrelin induced glucose release by primary hepatocytes (Gauna et al., 2005). In support of these findings, a recent report demonstrated the antagonistic effect in fish where ghrelin's orexigenic effects were blocked by administration of UAG. Furthermore, this effect appears to occur both centrally and peripherally (Matsuda et al., 2006). There are now many reports of direct biological activity of UAG in vitro that suggest a receptor mediated cellular response, perhaps via a specific receptor that is not GHS-R (Baldanzi et al., 2002, Cassoni et al., 2004, Chen et al., 2005, Gauna et al., 2006, Muccioli et al., 2004, Nanzer et al., 2004, Thompson et al., 2003, Toshinai et al., 2006). Despite these findings, the current consensus appears to be that UAG is inactive as an agonist of the GHS-R. However, the possibility remains that UAG is somehow able to block the ghrelin response by antagonizing the GHS-R. Therefore, we have explored in more detail, in a defined in vitro system, the ability of UAG to antagonize activation of the GHS-R by ghrelin, the potency of UAG at the GHS-R, and the effects of GHS-R antagonists.

Section snippets

Peptides

Human UAG was obtained from NeoMPS (Strasbourg, France) and Thera Technologies (Montreal, Canada). Human ghrelin, [D-Lys3]GHRP-6, somatostatin28, obestatin and glucagon were obtained from NeoMPS. The ghrelin analog BIM28163, a potent antagonist of the GHS-R, was kindly provided by IPSEN Pharmaceuticals (Milford, MA). All peptides had been assessed for purity and integrity by high performance liquid chromatography and mass spectrometry.

Aequoscreen assay for ghrelin and UAG activity

Aequoscreen cells were kindly provided by Euroscreen s.a.

Results

The main reason for examining UAG modulation of GHS-R activity was to elucidate a mechanism for our finding that this peptide could antagonize the effects of ghrelin on hepatocyte glucose production in vitro (Gauna et al., 2005). Therefore, the first experiment that we ran was designed to determine if UAG had any antagonistic activity in an in vitro system where we could examine its rapid effects directly. We have used Aequoscreen cells transfected with the human GHS-R (CHO-A5-GHSR) for this

Discussion

Overall, our observations in a cell line in which we can measure the effects of peptides exclusively on GHSR activation suggest that UAG can reinforce the activity of ghrelin, depending on effective concentrations. However in other in vitro models, and in vivo, the situation appears to be more complex. Previous studies show that UAG can displace radiolabelled ghrelin from membrane extracts of cell-lines that lack expression of GHS-R1a mRNA (e.g. Cassoni et al., 2001, Muccioli et al., 2004), and

Acknowledgments

We thank Euroscreen s.a. (Gosselies, Belgium) for providing the Aequoscreen cells, and Dr. M. Culler (Endocrinology Research Group, IPSEN Pharmaceuticals, MA) for providing the BIM28163. The study was supported by the Netherlands Organization for Scientific Research (NWO grant 912-03022).

References (26)

  • G. Baldanzi et al.

    Ghrelin and des-acyl ghrelin inhibit cell death in cardiomyocytes and endothelial cells through ERK1/2 and PI 3-kinase/AKT

    J. Cell Biol.

    (2002)
  • M.A. Bednarek et al.

    Structure–function studies on the new growth hormone-releasing peptide, ghrelin: minimal sequence of ghrelin necessary for activation of growth hormone secretagogue receptor 1a

    J. Med. Chem.

    (2000)
  • F. Broglio et al.

    Non-acylated ghrelin counteracts the metabolic but not the neuroendocrine response to acylated ghrelin in humans

    J. Clin. Endocrinol. Metab.

    (2004)
  • P. Cassoni et al.

    Identification, characterization, and biological activity of specific receptors for natural (ghrelin) and synthetic growth hormone secretagogues and analogs in human breast carcinomas and cell lines

    J. Clin. Endocrinol. Metab.

    (2001)
  • P. Cassoni et al.

    Expression of ghrelin and biological activity of specific receptors for ghrelin and des-acyl ghrelin in human prostate neoplasms and related cell lines

    Eur. J. Endocrinol.

    (2004)
  • C.Y. Chen et al.

    Des-acyl ghrelin acts by CRF type 2 receptors to disrupt fasted stomach motility in conscious rats

    Gastroenterology

    (2005)
  • K. Cheng et al.

    The synergistic effects of His-D-Trp-Ala-Trp-D-Phe-Lys-NH2 on growth hormone (GH)-releasing factor-stimulated GH release and intracellular adenosine 3′,5′-monophosphate accumulation in rat primary pituitary cell culture

    Endocrinology

    (1989)
  • K. Dezaki et al.

    Blockade of pancreatic islet-derived ghrelin enhances insulin secretion to prevent high-fat diet-induced glucose intolerance

    Diabetes

    (2006)
  • C. Gauna et al.

    Administration of acylated ghrelin reduces insulin sensitivity, whereas the combination of acylated plus unacylated ghrelin strongly improves insulin sensitivity

    J. Clin. Endocrinol. Metab.

    (2004)
  • C. Gauna et al.

    Ghrelin stimulates, whereas des-octanoyl ghrelin inhibits, glucose output by primary hepatocytes

    J. Clin. Endocrinol. Metab.

    (2005)
  • C. Gauna et al.

    Unacylated ghrelin is active on the INS-1E rat insulinoma cell line independently of the growth hormone secretagogue receptor type 1a and the corticotropin releasing factor 2 receptor

    Mol. Cell. Endocrinol.

    (2006)
  • S. Gnanapavan et al.

    The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in humans

    J. Clin. Endocrinol. Metab.

    (2002)
  • H.A. Halem et al.

    A novel growth hormone secretagogue-1a receptor antagonist that blocks ghrelin-induced growth hormone secretion but induces increased body weight gain

    Neuroendocrinology

    (2005)
  • Cited by (72)

    • Ghrelin receptor signaling in health and disease: a biased view

      2023, Trends in Endocrinology and Metabolism
    View all citing articles on Scopus
    View full text