Associate editor: I. Kimura
Ghrelin is a physiological regulator of insulin release in pancreatic islets and glucose homeostasis

https://doi.org/10.1016/j.pharmthera.2008.02.008Get rights and content

Abstract

Ghrelin, an acylated 28-amino acid peptide, was isolated from the stomach as the endogenous ligand for the growth hormone (GH) secretagogue receptor (GHS-R). Circulating ghrelin is produced predominantly in the oxyntic mucosa of stomach. Ghrelin potently stimulates GH release and feeding, and exhibits positive cardiovascular effects, suggesting a possible clinical application. Low plasma ghrelin levels are associated with elevated fasting insulin levels and insulin resistance, suggesting both physiological and pathophysiological roles for ghrelin in glucose metabolism. Here, we review the physiological role of ghrelin in the regulation of insulin release and glucose metabolism, and a potential therapeutic avenue to treat type 2 diabetes by manipulating ghrelin and/or its signaling. Ghrelin inhibits insulin release in mice, rats and humans. The signal transduction mechanisms of ghrelin in islet β-cells are distinct from those utilized in GH-releasing and/or GHS-R-expressing cells. Ghrelin is expressed in pancreatic islets and released into pancreatic microcirculations. Pharmacological and genetic blockades of islet-derived ghrelin markedly augment glucose-induced insulin release in vitro. In high-fat diet-induced mildly obese mice, ghrelin-deficiency enhances insulin release and prevents impaired glucose tolerance. Thus, manipulation of insulinostatic function of ghrelin — GHS-R system, particularly that in islets, could optimize the amount of insulin release to meet the systemic demand, providing a potential therapeutic application to prevent type 2 diabetes.

Introduction

Ghrelin, a novel acylated 28-amino acid peptide, was isolated from the human and rat stomach as the endogenous ligand (Kojima et al., 1999) for the growth hormone (GH) secretagogue-receptor (GHS-R) (Howard et al., 1996). This novel peptide has an n-octanoylated serine residue at the third N-terminal position; this acylation is essential for ghrelin bioactivity (Kojima et al., 1999). Circulating ghrelin is produced predominantly in the stomach (Ariyasu et al., 2001), while substantially lower amounts are detected in the intestine, pancreas, kidney, immune system, placenta, testis, pituitary, lung, and hypothalamus (Kojima et al., 1999, Hosoda et al., 2000, Date et al., 2000a, Mori et al., 2000, Gualillo et al., 2001, Tanaka et al., 2001, Date et al., 2002, Gnanapavan et al., 2002, Hattori et al., 2001, Muccioli et al., 2002, Tena-Sempere et al., 2002, Volante et al., 2002a, Volante et al., 2002b). GHS-Rs are expressed in the hypothalamus-pituitary unit, and are also distributed in other central and peripheral tissues (Howard et al., 1996, Guan et al., 1997, Smith et al., 1997, Muccioli et al., 1998, Papotti et al., 2000, Cassoni et al., 2001, Ghigo et al., 2001, Gnanapavan et al., 2002, Katugampola et al., 2002, Muccioli et al., 2002). Ghrelin is a potent stimulator of GH release (Kojima et al., 1999, Arvat et al., 2000, Arvat et al., 2001, Date et al., 2000b, Peino et al., 2000, Takaya et al., 2000). Ghrelin also stimulates feeding; ghrelin injected either centrally (Kamegai et al., 2001, Nakazato et al., 2001, Shintani et al., 2001, Wren et al., 2001b) or peripherally (Nakazato et al., 2001, Wren et al., 2001a) potently stimulates food intake. In humans, ghrelin peaks before meals, suggesting its role as a hunger signal (Cummings et al., 2002, Cummings and Overduin, 2007). Cardiovascular actions of ghrelin are also reported (Nagaya et al., 2001a, Nagaya et al., 2001b, Nagaya et al., 2001c, Nagaya and Kangawa, 2003a, Nagaya and Kangawa, 2003b, Lin et al., 2004, Matsumura et al., 2002). Given this wide spectrum of biological activities, the discovery of ghrelin opened many new perspectives within neuroendocrine, metabolic and cardiovascular research, thus suggesting its possible clinical application (Kojima & Kangawa, 2006). Ghrelin and GHS-R are also located in pancreatic islets (Date et al., 2002, Gnanapavan et al., 2002, Volante et al., 2002a, Wierup et al., 2002, Dezaki et al., 2004, Prado et al., 2004, Wierup et al., 2004, Wierup and Sundler, 2005). Ghrelin inhibits insulin release in mice, rats and humans (Broglio et al., 2001, Egido et al., 2002, Reimer et al., 2003, Dezaki et al., 2004). Low plasma ghrelin levels are associated with elevated fasting insulin levels and insulin resistance in humans (Ikezaki et al., 2002, Poykko et al., 2003). These findings suggest both physiological and pathophysiological roles for ghrelin in the regulation of insulin release. In addition, the plasma ghrelin level correlates inversely with obesity (Tschop et al., 2001, Ariyasu et al., 2002, Shiiya et al., 2002). Hence, ghrelin could be involved in energy and glucose metabolism, in which insulin plays a crucial role.

Here we review the physiological role of ghrelin in the regulation of insulin release and glucose metabolism, and present a potential therapeutic avenue to manipulate ghrelin signaling and thereby counteract the progression of type 2 diabetes.

Section snippets

Expressions of ghrelin and GHS-R in islets

Immunohistochemistry with antiserum against ghrelin demonstrated the immunoreactivity for ghrelin in a fraction of human and rat islet cells, which were observed mainly in the periphery of islets. Ghrelin-immunoreactive cells highly overlapped with glucagon-immunoreactive cells (Date et al., 2002, Dezaki et al., 2004, Kageyama et al., 2005), while some of glucagon-immunoreactive cells were not immunoreactive to ghrelin. Immunoreactive ghrelin was also observed in mouse islets (Iwakura et al.,

Insulinostatic function of islet-derived ghrelin in vitro

In isolated rat islets, GHS-R antagonists ([d-Lys3]-GHRP-6 and [d-Arg1, d-Phe5, d-Trp7,9, Leu11]-substance P) markedly increased insulin release in the presence of 5.6 mM glucose and this response was abolished in the absence of external Ca2+ (Dezaki et al., 2004), indicative of Ca2+-dependent insulin release by the receptor antagonists. Furthermore, antiserum against active ghrelin also increased insulin release, while control nonimmune serum had no effect (Dezaki et al., 2004). These results

Ghrelin attenuates [Ca2+]i in β-cells

In islet β-cells, cytosolic Ca2+ concentration ([Ca2+]i) is considered the major regulator of insulin secretion (Wollheim and Sharp, 1981, Prentki and Matschinsky, 1987). [Ca2+]i measured in islets by fura-2 microfluorometry was elevated mildly by increasing glucose concentration from 2.8 to 5.6 mM. In the presence of [d-Lys3]-GHRP-6, the peak of the first phase [Ca2+]i response was enhanced and, in some islets, oscillations of [Ca2+]i were induced (Dezaki et al., 2004). The peaks and

Exogenous ghrelin elevates plasma glucose and attenuates insulin levels

Systemic action of exogenous ghrelin to elevate blood glucose levels has been well documented in humans and rodents (Broglio et al., 2001, Broglio et al., 2002, Broglio et al., 2003a, Broglio et al., 2003b, Dezaki et al., 2004). In mice fasted overnight, intraperitoneal (i.p.) administration of ghrelin at concentrations of 1 and 10 nmol/kg significantly elevated blood glucose levels at 30 min after administration (Dezaki et al., 2004). The hyperglycemic effect of ghrelin was completely blocked

Increased plasma insulin and decreased blood glucose levels in ghrelin-KO mice

The effects of GHS-R antagonist and anti-ghrelin antiserum in the perfused pancreas, isolated islets, and the systemic insulin levels most likely result from counteraction of the action of endogenous ghrelin. To further confirm this hypothesis, ghrelin-knockout (Ghr-KO) mice were studied. In Ghr-KO mice, plasma ghrelin levels were undetectable. When fed standard chow, no significant differences between male Ghr-KO and wild-type (C57BL/6J) mice were observed at 8 weeks of age in body weights

Desacyl-ghrelin

The non-acylated form of ghrelin, desacyl-ghrelin, also exists at significant levels in pancreatic blood flow (Dezaki et al., 2006) as well as in systemic blood (Hosoda et al., 2000). In blood, desacyl-ghrelin circulates in amounts far greater than acylated ghrelin. The n-octanoyl group at serine 3 of the ghrelin molecule seems to be essential for the hormone's binding and bioactivity, at least in terms of endocrine actions, because the unacylated form of ghrelin does not activate the GHS-R (

Ghrelin-KO counteracts glucose intolerance in high-fat diet-fed and ob/ob mice

The enhanced insulin and suppressed glycemic responses to GTT in Ghr-KO mice could be beneficial under conditions of increased demand for insulin. When wild-type and Ghr-KO mice were fed high-fat diet (HFD) for 4 weeks, both mouse lines developed moderate increases in body weight to a similar extent (Dezaki et al., 2006). In an apparent controversy, it was reported that another line of Ghr-KO mice were protected from a rapid weight gain during post-weaning exposure to HFD for 3 weeks, which was

Conclusion

The notion that the islet-derived ghrelin plays a pivotal role in the regulation of insulin release at least in rodents is supported by the following findings. (1) mRNAs and proteins for ghrelin and GHS-R are expressed in pancreatic islets. (2) The ghrelin level is higher in the pancreatic vein than in the artery, indicative of release of ghrelin from pancreas. (3) Ghrelin immunoneutralization and GHS-R antagonists augment glucose-induced insulin release from perfused pancreas and isolated

Acknowledgments

We thank Drs. Kenji Kangawa and Hiroshi Hosoda (National Cardiovascular Center Research Institute) for providing ghrelin and ghrelin antiserum and for valuable discussion, and Dr. Masayasu Kojima (Kurume University) for providing ghrelin knockout mice and for valuable discussion.

We also thank Ms. Minako Warashina, Ms. Seiko Ookuma and Ms. Megumi Motoshima for technical assistance, and Ms. Chizu Sakamoto for secretarial assistance. This work was supported by Grants-in-Aid for Scientific Research

References (148)

  • HosodaH. et al.

    Ghrelin and des-acyl ghrelin: two major forms of rat ghrelin peptide in gastrointestinal tissue

    Biochem Biophys Res Commun

    (2000)
  • HsuW.H. et al.

    Somatostatin inhibits insulin secretion by a G-protein-mediated decrease in Ca2+ entry through voltage-dependent Ca2+ channels in the beta cell

    J Biol Chem

    (1991)
  • IwakuraH. et al.

    Analysis of rat insulin II promoter-ghrelin transgenic mice and rat glucagon promoter-ghrelin transgenic mice

    J Biol Chem

    (2005)
  • KageyamaH. et al.

    Morphological analysis of ghrelin and its receptor distribution in the rat pancreas

    Regul Pept

    (2005)
  • KatadaT. et al.

    Islet-activating protein. Enhanced insulin secretion and cyclic AMP accumulation in pancreatic islets due to activation of native calcium ionophores

    J Biol Chem

    (1979)
  • KojimaM. et al.

    Ghrelin: discovery of the natural endogenous ligand for the growth hormone secretagogue receptor

    Trends Endocrinol Metab

    (2001)
  • LauwersE. et al.

    Obestatin does not activate orphan G protein-coupled receptor GPR39

    Biochem Biophys Res Commun

    (2006)
  • MacDonaldP.E. et al.

    Inhibition of Kv2.1 voltage-dependent K+ channels in pancreatic β-cells enhances glucose-dependent insulin secretion

    J Biol Chem

    (2002)
  • MagistrelliG. et al.

    Substitution of two variant residues in the protein tyrosine phosphatase-like PTP35/IA-2 sequence reconstitutes catalytic activity

    Biochem Biophys Res Commun

    (1996)
  • MatsumotoM. et al.

    Structure–activity relationship of ghrelin: pharmacological study of ghrelin peptides

    Biochem Biophys Res Commun

    (2001)
  • McKeeK.K. et al.

    Cloning and characterization of two human G protein-coupled receptor genes (GPR38 and GPR39) related to the growth hormone secretagogue and neurotensin receptors

    Genomics

    (1997)
  • MoecharsD. et al.

    Altered Gastrointestinal and metabolic function in the GPR39-obestatin receptor-knockout mouse

    Gastroenterology

    (2006)
  • MoriK. et al.

    Kidney produces a novel acylated peptide, ghrelin

    FEBS Lett

    (2000)
  • MuccioliG. et al.

    Ghrelin and des-acyl ghrelin both inhibit isoproterenol-induced lipolysis in rat adipocytes via a non-type 1a growth hormone secretagogue receptor

    Eur J Pharmacol

    (2004)
  • MuccioliG. et al.

    Neuroendocrine and peripheral activities of ghrelin: implications in metabolism and obesity

    Eur J Pharmacol

    (2002)
  • NagayaN. et al.

    Ghrelin improves left ventricular dysfunction and cardiac cachexia in heart failure

    Curr Opin Pharmacol

    (2003)
  • NagayaN. et al.

    Ghrelin, a novel growth hormone releasing peptide, in the treatment of chronic heart failure

    Regul Pept

    (2003)
  • Aguilar-BryanL. et al.

    Molecular biology of adenosine triphosphate-sensitive potassium channels

    Endocr Rev

    (1999)
  • AhrénB.

    Autonomic regulation of islet hormone secretion — implications for health and disease

    Diabetologia

    (2000)
  • AriyasuH. et al.

    Delayed short-term secretory regulation of ghrelin in obese animals: evidenced by a specific RIA for the active form of ghrelin

    Endocrinology

    (2002)
  • AriyasuH. et al.

    Stomach is a major source of circulating ghrelin, and feeding state determines plasma ghrelin-like immunoreactivity levels in humans

    J Clin Endocrinol Metab

    (2001)
  • ArvatE. et al.

    Preliminary evidence that Ghrelin, the natural GH secretagogue (GHS)-receptor ligand, strongly stimulates GH secretion in humans

    J Endocrinol Invest

    (2000)
  • ArvatE. et al.

    Endocrine activities of ghrelin, a natural growth hormone secretagogue (GHS), in humans: comparison and interactions with hexarelin, a nonnatural peptidyl GHS, and GH-releasing hormone

    J Clin Endocrinol Metab

    (2001)
  • AsakawaA. et al.

    Antagonism of ghrelin receptor reduces food intake and body weight gain in mice

    Gut

    (2003)
  • AstesanoA. et al.

    Cellular and subcellular expression of Golf/Gs and Gq/G11 α-subunits in rat pancreatic endocrine cells

    J Histochem Cytochem

    (1999)
  • BednarekM.A. 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)
  • Bresson-BepoldinL. et al.

    GHRP6-stimulated hormone secretion in somatotrophs: involvement of intracellular and extracellular calcium sources

    J Neuroendocrinol

    (1996)
  • BroglioF. et al.

    Ghrelin, a natural GH secretagogue produced by the stomach, induces hyperglycemia and reduces insulin secretion in humans

    J Clin Endocrinol Metab

    (2001)
  • BroglioF. et al.

    Endocrine activities of cortistatin-14 and its interaction with GHRH and ghrelin in humans

    J Clin Endocrinol Metab

    (2002)
  • BroglioF. et al.

    The endocrine response to ghrelin as a function of gender in humans in young and elderly subjects

    J Clin Endocrinol Metab

    (2003)
  • BroglioF. et al.

    Non-acylated ghrelin does not possess the pituitaric and pancreatic endocrine activity of acylated ghrelin in humans

    J Endocrinol Invest

    (2003)
  • BroglioF. et al.

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

    J Clin Endocrinol Metab

    (2004)
  • CassoniP. 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)
  • ChanC.B. et al.

    Increased uncoupling protein-2 levels in β-cells are associated with impaired glucose-stimulated insulin secretion: mechanism of action

    Diabetes

    (2001)
  • ChanC.B. et al.

    Overexpression of uncoupling protein 2 inhibits glucose-stimulated insulin secretion from rat islets

    Diabetes

    (1999)
  • ChanoineJ.P. et al.

    Ghrelin gene expression is markedly higher in fetal pancreas compared with fetal stomach: effect of maternal fasting

    Endocrinology

    (2004)
  • ChanoineJ.P. et al.

    Obestatin, acylated and total ghrelin concentrations in the perinatal rat pancreas

    Horm Res

    (2006)
  • ChiangM.K. et al.

    PTP-NP, a new member of the receptor protein tyrosine phosphatase family, implicated in development of nervous system and pancreatic endocrine cells

    Development

    (1996)
  • ColomboM. et al.

    Effects of ghrelin and other neuropeptides (CART, MCH, orexin A and B, and GLP-1) on the release of insulin from isolated rat islets

    Pancreas

    (2003)
  • CummingsD.E. et al.

    Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery

    N Engl J Med

    (2002)
  • Cited by (136)

    • Targeting appetite and satiety in diabetes and obesity, via G protein-coupled receptors

      2022, Biochemical Pharmacology
      Citation Excerpt :

      In one study, this effect is completely blocked by equivalent concentrations of des-acyl ghrelin, although this result differs from previous studies [66,67]. Several studies report that these effects are via atypical coupling and pharmacology of GHSR1a in pancreatic β-cells [63,64,67]. This is inconsistent with studies by 2 other groups that demonstrate mRNA, functional expression and typical coupling of GHSR1a on pancreatic δ-cells to cause release of somatostatin, which then acts on β-cells to suppress insulin secretion and are consistent with GHSR1a knockout data [68,69].

    • Ghrelin suppresses insulin secretion in human islets and type 2 diabetes patients have diminished islet ghrelin cell number and lower plasma ghrelin levels

      2020, Molecular and Cellular Endocrinology
      Citation Excerpt :

      Low ghrelin levels associate with T2D prevalence (Poykko et al., 2003) and pregnant women with T2D or gestational diabetes have lower ghrelin levels than non-diabetic women (Gomez-Diaz et al., 2016), but the underlying causes are unknown. It is established that ghrelin is a hormone with insulin-suppressing properties, both in vivo in healthy humans, as well as in isolated rodent islets, single rodent beta cells and beta cell lines (Gray et al., 2019; Dezaki et al., 2008). Blocking ghrelin signaling may be a therapeutic avenue for T2D treatment; an assumption supported by observations that oral administration of an antagonist to the ghrelin receptor, GHSR, improves rodent glucose homeostasis (Esler et al., 2007).

    • Pancreatic Hormones

      2020, Hormonal Signaling in Biology and Medicine: Comprehensive Modern Endocrinology
    • Pancreatic Hormones

      2019, Hormonal Signaling in Biology and Medicine: Comprehensive Modern Endocrinology
    View all citing articles on Scopus
    View full text