Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Dec 1.
Published in final edited form as: Curr Gastroenterol Rep. 2012 Dec;14(6):480–488. doi: 10.1007/s11894-012-0291-3

Gastric Peptides and their Regulation of Hunger and Satiety

Andreas Stengel 1, Yvette Taché 2,*
PMCID: PMC3482275  NIHMSID: NIHMS409632  PMID: 23001831

Abstract

Ingestion of food affects the secretion of hormones from specialized endocrine cells scattered within the intestinal mucosa. Upon release, these hormones mostly decrease food intake by signaling information to the brain. Although enteroendocrine cells in the small intestine were thought to represent the predominant gut-brain regulators of food intake, recent advances also established a major role for gastric hormones in these regulatory pathways. First and foremost, the gastric endocrine X/A-like cell was in the focus of many studies due to the production of ghrelin which is until now the only known orexigenic hormone that is peripherally produced and centrally acting. Although X/A-cells were initially thought to only release one hormone that stimulates food intake, this view has changed with the identification of additional peptide products also derived from this cell, namely desacyl ghrelin, obestatin and nesfatin-1. Desacyl ghrelin may play a counter-regulatory role to the food intake stimulatory effect of ghrelin. The same property was suggested for obestatin; however, this hypothesis could not be confirmed in numerous subsequent studies. Moreover, the description of the stomach as the major source of the novel anorexigenic hormone nesfatin-1 derived from the NUCB2 gene further corroborated the assumption that the gastric X/A-like cell is not only a stimulator but also an inhibitor of feeding thereby acting as so far unique dual regulator of food intake located in a logistically important place where the gastrointestinal tract has initial contact with food.

Keywords: brain-gut axis, desacyl ghrelin, food intake, ghrelin, nesfatin-1, NUCB2, obesity, obestatin, X/A-like cell

Introduction

Specialized enteroendocrine cells located within the mucosa of the small but also the large intestine [1] were established as main regulators of food intake and intestinal functions due to the production and release of peptide hormones [2]. However, also the stomach contains distinct endocrine cells, namely gastrin-releasing cells (G cells), somatostatin-producing cells (D cells >20% of gastric oxyntic endocrine cells in humans, 5-10% in rats), enterochromaffin-like cells producing histamine (ECL, 30% in human and 65% in the rat) and in low abundance serotonin-containing enterochromaffin (EC) cells [1]. These cells’ products were mainly implicated in the regulation of acid production and secretion and motility. Moreover, a fifth endocrine cell type was described scattered in the gastric oxyntic mucosa and was named P/D1 cell in man and X/A-like cell in rats [1]. These cells account for 20-30% of the oxyntic endocrine cells thereby representing the second most abundant gastric endocrine cell type [1]. The function of these cells, unknown for a long time, was unraveled with the discovery of ghrelin in 1999 [3] identified as the only peripherally produced and centrally acting orexigenic peptide hormone [4]. This landmark discovery drew attention to this cell type in the context of regulating food intake and body weight. Subsequently, additional peptide products have been identified in this cell type including desacyl ghrelin and n-decanoyl ghrelin [4, 5], obestatin [6] and nucleobindin2/nesfatin-1 (NUCB2/nesfatin-1) [7]. Several studies were conducted to characterize these hormones with regard to their effects on food intake as recently reviewed for desacyl ghrelin [8], nesfatin-1 [9] and obestatin [10]. Overall, these studies point towards an anorexigenic effect of these additional peptide hormones thereby highlighting a unique role for the gastric X/A-like cell as dual regulator of food intake.

Peptide products of the gastric endocrine X/A-like cell

Ghrelin and desacyl ghrelin

More than a decade ago Kangawa and Kojima discovered the endogenous ligand of the long time orphan receptor, growth hormone (GH) secretagogue receptor 1a (GHS-R1a), which was named ghrelin [3, 11•]. The peptide ghrelin consists of 28 amino acids with a unique octanoyl fatty acid modification on the serine-3 residue that increases lipophilicity [3]. This modification is essential for binding to the GHS-R1a [3, 12], which was later renamed ghrelin receptor (GRLN-R) [13]. Subsequent structure-function studies established that the full length peptide is not necessary to stimulate the GRLN-R and identified the first five N-terminal amino acids containing the serine-3 fatty acid modification as the active core required to activate the receptor [14]. Recently, another form of ghrelin, called n-decanoyl ghrelin, was identified in rodents and shown to be the major circulating form of ghrelin in mice [5]. Interestingly, it was also shown that food derived medium chain fatty acids can be used as direct source for the acylation of ghrelin [14].

Desacyl ghrelin lacking the octanoyl group represents the major form of ghrelin in the circulation [15] although recent advances in improving blood processing for labile peptides changed the acyl/desacyl ghrelin ratio from 1:55 [16] to 1:5 [17•]. In contrast to ghrelin and n-decanoyl ghrelin, desacyl ghrelin does not bind to the GRLN-R [3] and the receptor of desacyl ghrelin is yet to be identified.

Innovative advancements came in 2008 when two independent groups identified the enzyme involved in catalyzing the acyl modification as the fourth member of membrane-bound O-acyltransferases (MBOATs) which was named ghrelin-O-acyltransferase (GOAT) [18, 19]. GOAT not only catalyzes the attachment of the octanoyl but also the decanoyl group resulting in octanoyl and decanoyl ghrelin respectively [19]. This acylation is assumed to occur before proghrelin is transported into the Golgi apparatus [18]. However, besides gastric GOAT mRNA and protein expression [20, 21], GOAT protein was also detected in the circulation of rats and mice [21] leading to the speculation about an extracellular acylation of ghrelin. This finding is yet to be confirmed in humans although first pilot data also show the occurrence of GOAT protein in human plasma (unpublished observations). Moreover, GOAT mRNA expression was shown to be widely present in human tissues including oesophagus, stomach, small intestine, colon, pancreas, liver, gallbladder, bile duct, spleen, kidney, cortex of adrenal gland, myocardium, lung, skeletal muscle, fat tissue, thyroid gland, testis, prostate, ovary, placenta and pituitary gland [22] suggesting possible additional functions of GOAT. A recent study suggested that GOAT plays a critical role in bile acid reabsorption [23].

Obestatin

Obestatin is a 23 amino acid peptide also derived from the ghrelin gene and was first described in 2005 based on a computer predicted splicing and post-translational processing site [6]. Like ghrelin, obestatin is expressed in rodent gastric X/A-like cells [24] and human endocrine P/D1 cells [25, 26]. This peptide was thought to act in opposition to ghrelin by antagonizing ghrelin’s orexigenic effect and reducing body weight [6, 27].

NUCB2/Nesfatin-1

Recently, another peptide hormone has been identified in rat X/A-like cells, namely nucleobindin2 (NUCB2)/nesfatin-1 [7]. Nesfatin-1, an 82 amino acid peptide, was first described in the rat hypothalamus as peptide product derived from NUCB2 [28]. NUCB2/nesfatin-1 is widely expressed in the rat brain [29-31] but also in the periphery including the pancreas, liver and white (subcutaneous and visceral fat) as well as brown (interscapular fat) adipose tissue [32]. Interestingly, NUCB2 mRNA expression is 10-times higher in the rat gastric mucosa than in the brain giving rise to a predominant production of nesfatin-1 in the stomach [7]. Phenotypical characterization in rats showed that nesfatin-1 immunoreactivity colocalized with ghrelin within the same gastric endocrine cells with a different subcellular cytoplasmatic localization in distinct and different pools of vesicles [7]. In addition, the expression of prohormone convertase (PC) 1/3 in X/A-like cells [18] also supports the coexpression of ghrelin and nesfatin-1 in one cell type as this hormone is required for the maturation of both peptide hormones [18, 33]. The colocalization of ghrelin and nesfatin-1 was recently also confirmed in the anterior intestine of goldfish [34] highlighting the phylogenetic stability of this expression pattern which gives rise to its physiological importance. Whether this finding can be translated into humans warrants further investigation. However, first pilot data in human stomach also show a colocalization of nesfatin-1 and ghrelin immunoreactivity in endocrine P/D1 cells (unpublished observation).

Regulation of peptide release

Ghrelin and desacyl ghrelin

The major source of circulating ghrelin is the stomach as indicated by a marked decrease of plasma ghrelin levels following gastrectomy [35]. Other, although quantitatively less important, sources of peripheral ghrelin encompass the small and large intestine [4], pancreas [36] and other organs such as kidney, liver, testis and adipose tissue [37, 38]. Blood levels of ghrelin are well established to be regulated by feeding status with a rise before food intake and a fall quickly thereafter [39, 40]. Recent studies also indicate the cephalic phase initiated by presenting pictures of food increases circulating ghrelin levels in humans [41]. In addition, fasting also results in an increase of circulating ghrelin levels associated with an increased gastric production and release of this peptide [42••-44]. Likewise, gastric GOAT mRNA expression and circulating GOAT protein concentration is increased under conditions of food restriction [21, 45] likely to further stimulate the acylation of ghrelin. Besides these short term influences on ghrelin, long term alterations of metabolic homeostasis also affect blood ghrelin levels with an increase under conditions of decreased body weight such as cachexia and anorexia and a marked decrease in obese subjects [46-48]. As ghrelin and desacyl ghrelin are derived from the same precursor, for a long time desacyl ghrelin was thought to merely represent the inactivated ghrelin peptide. However, recent studies provide evidence that both peptide forms are subject to differential regulation. In particular, the reduction of gastric pH results in the release of desacyl ghrelin but not ghrelin [49]. In addition, various stressors (injection of lipopolysaccharide as immunological stressor or abdominal surgery as physical stressor) influence the acyl/desacyl ghrelin ratio due to a rapid decline of ghrelin [50, 51]. Modification of dietary nutrition was also recently reported to influence deacyl ghrelin. High fish oil increased the circulating levels compared to standard high fat diet while high fat butter reduced it compared to other groups indicative that different levels of fatty acid saturation in the composition of high fat food influences fasting levels of desacyl ghrelin [52]. Although the underlying mechanisms remain to be fully characterized, a reduction of acylation due to a decrease in GOAT expression observed under these conditions [50] has been hypothesized. On a general note, one has to keep in mind that although the regulation of ghrelin under different metabolic and other conditions has been extensively described, the underlying mechanisms controlling these alterations of expression and release on a cellular basis are not well characterized. This is largely due to the fact that the low quantities of X/A-like cells hamper the enrichment to pure cell preparations and immortalized ghrelin producing MGN3-1 tumor cell lines [53] only allow a limited proportion of extrapolation to native X/A-like cells. This was recently shown by the demonstration that dopamine stimulates ghrelin release from MGN3-1 cells [54], while in vivo increased levels of dopamine in the circulation [55] or in the gastric mucosa [56] did not alter fasting ghrelin levels in rats. Animal models with fluorescent dye coupled to the ghrelin promoter allow to enrich ghrelin producing cells to higher purity as in the recent use of green fluorescent dye [57•• ]. With this approach the expression of GRP120 on isolated ghrelin cells was demonstrated along with the interaction of long chain free fatty acids with this receptor to reduce ghrelin release [57••].

Ghrelin binds to the GRLN-R widely expressed in the periphery (e.g. abdominal organs, vagal afferents, myenteric neurons, adipose tissue and pituitary) and the brain [37, 58-60]. After stimulation by ghrelin, the GRLN-R desensitizes through endocytosis via clathrin-coated pits [61] to prevent an overstimulation. Since ghrelin is the only known peripherally produced and centrally acting stimulator of food intake several studies attempted to block feeding and body weight gain using ghrelin antagonists. However, although initial data were promising none of the candidates moved towards clinical trials likely owing to the fact that the GRLN-R displays a high constitutive activity [62] even in the presence of antagonists. Based on these observations, focusing on the development of inverse agonists with high stability in blood and slow blood clearance as well as large diffusion in tissues [63•] may be a promising approach. Unlike ghrelin, desacyl ghrelin is unable to bind to the GRLN-R; hence the receptor for this peptide remains to be established. Since desacyl ghrelin exerts actions on sites where the GRLN-R is not expressed such as stimulation of insulin release from INS-1E cells [64] or inhibition of proliferation of breast cancer cells [65] the expression of a distinct desacyl ghrelin receptor is strongly suspected but is yet to be discovered.

Obestatin

Unlike ghrelin, plasma levels of obestatin do not change with metabolic status [6, 66], a finding that does not support the proposed role as negative modulator of food intake. In the study describing the discovery of obestatin, the authors reported the GPR39, a seven transmembrane domain G protein-coupled receptor, to mediate obestatin’s effects [6]. The same group also recently described expression and functional relevance of this receptor in gastrointestinal and adipose tissues to mediate the actions of obestatin [67]. However, various independent groups of investigators did not confirm these findings and showed that obestatin does not bind to the GPR39 [68-71] resulting in the conclusion that obestatin is not the endogenous ligand of the GPR39.

NUCB2/Nesfatin-1

As expected for a negative modulator of feeding, plasma levels of NUCB2/nesfatin-1 are affected by the feeding status with a decline during fasting and a restoration of levels upon refeeding [7]. As gastric NUCB2 mRNA expression is also decreased during fasting [7, 72], the lower circulating levels of NUCB2/nesfatin-1 may result from reduced gastric production and release. A recent study indicates that NUCB2/nesfatin-1 is directly regulated by the mammalian target of rapamycin (mTOR) as the inhibition of gastric mTOR signaling by rapamycin blunted the expression of gastric NUCB2/nesfatin-1 at the gene and protein levels in vivo and in vitro suggesting a direct regulatory effect of mTOR on NUCB2/nesfatin-1 [72]. Interestingly, these short term changes of circulating NUCB2/nesfatin-1 concentrations in response to feeding and fasting were not observed in healthy human subjects [73, 74]. However, under conditions of body mass index (BMI) alterations, plasma NUCB2/nesfatin-1 levels were lower in anorexic [75] and higher in obese patients [76, 77] which is opposite to the results observed for ghrelin before. This increased expression in obese could be directly related to the increased amount of white adipose tissue where expression of NUCB2 was recently reported [76]. Contrasting with our increasing knowledge on nesfatin-1’s regulation and effects [9, 78], the receptor mediating those actions is still unknown. A previous study suggested the mediation by a G-protein-coupled receptor due to the fact that nesfatin-1 increased [Ca2+] in isolated cultured hypothalamic cells which was linked with protein kinase A signaling [29]. The isolation of the receptor and description of its expression sites will represent a big leap forward in our understanding of the physiology of NUCB2/nesfatin-1.

Effects on short term food intake and long term energy homeostasis

Ghrelin and desacyl ghrelin

Ghrelin’s food intake stimulating effects have been early on described in animals [79, 80] and humans [81], an effect inhibited by GRLN-R antagonists [82]. In addition, mice lacking the GRLN-R do not respond with increased food ingestion following injection of ghrelin [83, 84] highlighting the role of this receptor in the mediation of ghrelin’s orexigenic effect. Similar to ghrelin’s effects, also n-decanoyl ghrelin stimulates feeding matching the binding affinity to the GRLN-R [5]. It became apparent over the past few years that ghrelin is not only involved in the short term regulation of food intake but also in the long term mediation of energy homeostasis thereby influencing body weight as also reflected in the negative correlation of ghrelin with BMI. When ghrelin is infused chronically, body weight gain is increased due to stimulated food intake but also increased fat storage and reduced mobilization of fat [46, 85, 86]. This is likely due to an increase in enzymes involved in the mediation of fat storage such as lipoprotein lipase, stearoyl-CoA desaturase-1, acetyl-CoA carboxylase alpha and fatty acid synthase along with a decrease in mRNA expression of an enzyme promoting fat oxidation, namely carnitine palmitoyl transferase-1 alpha [87]. Interestingly, these expression levels are reversed under conditions of ghrelin depletion in mice lacking ghrelin [87] supporting a physiological role of ghrelin in mediating alterations of these pathways. In addition to ghrelin, also GOAT is suggested to play a regulatory role as mice lacking GOAT displayed a lower body weight compared to wild type littermates under conditions of high fat diet feeding whereas when fed normal chow no differences were apparent [88••]. Moreover, in GOAT knock-out (KO) mice fed food enriched with medium-chain triglycerides, fat mass was decreased [88••] resulting in the hypothesis that GOAT can act as endogenous lipid sensor.

Ghrelin released from gastric X/A cells to stimulate feeding can act directly on food intake regulatory brain nuclei after crossing the blood-brain barrier [89, 90] or mediate its orexigenic action indirectly via the vagus nerve [91, 92]. Earlier studies showed that rats with subdiaphragmatic or gastric vagotomy do not respond with increased food intake following intravenous injection of ghrelin [91]. In contrast, another group reported that the orexigenic stimulus exerted by intraperitoneally injected ghrelin is still observed in rats after elective subdiaphragmatic vagal deafferentation [93]. These discrepant results could be explained by different surgical techniques, different doses of ghrelin used and also different routes of administration but add to the debate about the predominant route of orexigenic action of circulating ghrelin. In addition to its predominant source in the periphery, ghrelin is also produced in the brain in neurons adjacent to the third ventricle [94] and in the arcuate nucleus of the hypothalamus [95], a key brain center integrating food intake regulatory signals [96]. Ghrelin containing neurons in these nuclei are connected to other cells containing the orexigenic brain transmitters, neuropeptide Y (NPY) and agouti-related peptide (AgRP) [94, 97]. It has been shown early on that peripheral [98] as well as central [99] injection of ghrelin activates NPY neurons in the arcuate nucleus suggesting a downstream mediation of ghrelin’s orexigenic effect by these peptides. This was corroborated by pharmacological (anti-NPY and anti-AgRP antibodies) [100] as well as genetic (NPY and AgRP knockout) [101] approaches showing that inhibiting both, NPY and AgRP signaling blocks ghrelin’s orexigenic effects whereas single blockade of either one had no effect most likely related to a compensatory action.

While desacyl ghrelin was initially thought to represent an inactive or inactivated peptide, several sets of evidence now point towards a food intake modulatory action of this peptide although the data are less compelling compared to ghrelin [8]. Several studies reported a reduction of food intake following peripheral [102] and central (intracerebroventricular) injection [103], while one study described an increased food intake after intracerebroventricular injection of a low dose of desacyl ghrelin in rats or mice [104]. Furthermore, another study did not observe any food intake modulatory effects when desacyl ghrelin was injected alone but when co-injected with ghrelin, desacyl ghrelin abolished ghrelin’s orexigenic effect [105]. This observation led to the speculation of a counterbalancing role of desacyl ghrelin in the regulation of food intake. Similar to ghrelin, also desacyl ghrelin has been suggested to play a role in long term control of energy and body weight homeostasis since mice over-expressing desacyl ghrelin display a smaller body size and less white adipose tissue mass [106] resulting in reduced body weight [107] compared to wild type controls. Despite the fact that these findings give rise to a reduction of lipid accumulation by desacyl ghrelin, recent in vitro data indicated a stimulation of intracellular lipid accumulation in human adipocytes by desacyl ghrelin [108]. These conflicting results may be due to methodological differences (in vitro versus whole animals) but may also point towards species differences (human versus mouse) and warrant further investigations.

Obestatin

The authors discovering obestatin reported that this peptide is a physiological anorexigenic mediator of feeding counteracting ghrelin’s orexigenic effect [6]. However, very few subsequent studies were able to partially reproduce these data whereas, by far, most studies did not observe any modulatory effect on food intake, body weight (for review see [66]) or ghrelin’s orexigenic effect [109]. Based on this converging evidence obestatin is not considered to play a role in the regulation of hunger and satiety and the physiological role of this peptide is yet to be determined.

NUCB2/Nesfatin-1

The discovery of NUCB2/nesfatin-1 in the hypothalamus was paired with the description of the reduction of food intake after single injection and a blunted body weight gain after continuous third ventricular infusion of nesfatin-1 [28]. These effects are likely to present a physiological action of this peptide and not merely a pharmacological phenomenon as blockade of endogenous NUCB2/nesfatin-1 signaling by the use of a 3rd ventricle injection of an antisense oligonucleotide resulted in a stimulation of food intake and an increased body weight gain in rats [28]. This anorexigenic action upon central administration was also shown in several other studies by independent groups in rats [110, 111] and subsequently also in mice [112, 113] and goldfish [114]. The reduction of dark phase food intake was due to the stimulation of satiation (indicated by a reduction of meal size) and satiety (indicated by a decrease in meal frequency and prolongation of inter-meal intervals) as recently shown in mice using an automated episodic food intake monitoring device [113]. One has to note that nesfatin-1 selectively inhibits dark phase food intake in ad libitum fed animals [28, 110, 115, 116] whereas results were not robust during the light phase under fasting conditions [110, 115]. This observation may point towards the interaction with other brain transmitters specifically activated during the dark photoperiod. The anorexigenic action was also observed following intracerebroventricular injection of mid segment nesfatin-130-59 in mice giving rise to the active core of the peptide able to activate the yet unknown receptor [117]. After establishing the effects on food intake several studies investigated the brain site of nesfatin-1’s anorexigenic action. While after injection in cerebrospinal fluid at the level of the hypothalamus the effect on dark phase food intake was delayed in onset with a maximum reduction occurring during the third hour post injection [110], the effect was already observed during the first hour when the peptide was injected into the hindbrain at the level of the cisterna magna or the fourth ventricle [110]. Interestingly, while the corticotropin releasing factor receptor 2 (CRF2) antagonist, astressin2-B blocked the anorexigenic effect of nesfatin-1 injected at the level of the hypothalamus, its injection into the hindbrain did not influence nesfatin-1’s anorexic effect indicative of different forebrain versus hindbrain downstream signaling pathways [110]. Besides CRF2 signaling, the melanocortin and oxytocin pathways seem to be involved in nesfatin-1 food intake inhibitory action as injection of the melanocortin 3/4 receptor antagonist, SHU9119 [28, 115] and the oxytocin antagonist, ornithine vasotocin [111, 116] blocked the nesfatin-1-induced anorexigenic effect. Convergent reports also established that nesfatin-1’s action is independent of leptin [28, 116, 118] which is an important feature in the context of an anti-obesity target since leptin sensitivity is reduced under conditions of increased fat mass.

Contrasting with consistent evidence from numerous studies reporting the anorexigenic effect of centrally injected nesfatin-1, although NUCB2/nesfatin-1 is more abundant in the stomach than in the brain [7] the effects of nesfatin-1 on food intake are not well established. Similar to the central effect, the mid segment nesfatin-130-59 also reduces food intake in mice when injected intraperitoneally although higher doses are needed [119]. This effect is likely to be vagally mediated as nesfatin-1 activates the Ca2+ influx in primary cultured nodose ganglion neurons in vitro [120] and the anorexigenic effect is not observed in animals pretreated with the neurotoxin capsaicin [119]. Similar to the study in rodents, also in goldfish only high doses of nesfatin-1 injected peripherally reduced food intake [114] suggesting that the major site of nesfatin-1 action is within the brain.

Summary & Conclusion

While gastric X/A-like cells were long thought to be exclusively involved in the stimulation of food intake due to the production of the only known peripherally produced and centrally acting orexigenic peptide hormone ghrelin, recent studies highlighted a function as anorexigenic modulator as well due to the production of desacyl ghrelin and nesfatin-1 in the same cells. Although our knowledge on the effects of these peptides markedly increased especially over the last three years. several important nuts have to be cracked over the next few years, especially the identification of the receptors for desacyl ghrelin and nesfatin-1 as well as the subcellular regulatory mechanisms controlling synthesis and release of the peptides produced in X/A-like cells.

Acknowledgments

Dr. Taché is in receipt of VA Research Career Scientist Award, VA Merit Award, Center Grant NIH DK-41301 (Animal Core) and R01 NIH DK 33061. Dr. Stengel is supported by the German Research Foundation Grant STE 1765/3-1 and Charité University Funding UFF 89-441-176.

Footnotes

Disclosure

No potential conflicts of interest relevant to this article were reported.

References and Recommended Reading

Papers of particular interest, published recently, have been highlighted as:

• Of importance

•• Of major importance

  • [1].Rindi G, Leiter AB, Kopin AS, et al. The “normal” endocrine cell of the gut: changing concepts and new evidences. Ann N Y Acad Sci. 2004;1014:1–12. doi: 10.1196/annals.1294.001. [DOI] [PubMed] [Google Scholar]
  • [2].Hameed S, Dhillo WS, Bloom SR. Gut hormones and appetite control. Oral Dis. 2009;15:18–26. doi: 10.1111/j.1601-0825.2008.01492.x. [DOI] [PubMed] [Google Scholar]
  • [3].Kojima M, Hosoda H, Date Y, et al. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature. 1999;402:656–660. doi: 10.1038/45230. [DOI] [PubMed] [Google Scholar]
  • [4].Date Y, Kojima M, Hosoda H, et al. Ghrelin, a novel growth hormone-releasing acylated peptide, is synthesized in a distinct endocrine cell type in the gastrointestinal tracts of rats and humans. Endocrinology. 2000;141:4255–4261. doi: 10.1210/endo.141.11.7757. [DOI] [PubMed] [Google Scholar]
  • [5].Hiejima H, Nishi Y, Hosoda H, et al. Regional distribution and the dynamics of n-decanoyl ghrelin, another acyl-form of ghrelin, upon fasting in rodents. Regul Pept. 2009;156:47–56. doi: 10.1016/j.regpep.2009.05.003. [DOI] [PubMed] [Google Scholar]
  • [6].Zhang JV, Ren PG, Avsian-Kretchmer O, et al. Obestatin, a peptide encoded by the ghrelin gene, opposes ghrelin’s effects on food intake. Science. 2005;310:996–999. doi: 10.1126/science.1117255. [DOI] [PubMed] [Google Scholar]
  • [7].Stengel A, Goebel M, Yakubov I, et al. Identification and characterization of nesfatin-1 immunoreactivity in endocrine cell types of the rat gastric oxyntic mucosa. Endocrinology. 2009;150:232–238. doi: 10.1210/en.2008-0747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Inhoff T, Wiedenmann B, Klapp BF, et al. Is desacyl ghrelin a modulator of food intake? Peptides. 2009;30:991–994. doi: 10.1016/j.peptides.2009.01.019. [DOI] [PubMed] [Google Scholar]
  • [9].Stengel A, Taché Y. Minireview: Nesfatin-1--An emerging new player in the brain-gut, endocrine, and metabolic axis. Endocrinology. 2011;152:4033–4038. doi: 10.1210/en.2011-1500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Lacquaniti A, Donato V, Chirico V, et al. Obestatin: an interesting but controversial gut hormone. Ann Nutr Metab. 2011;59:193–199. doi: 10.1159/000334106. [DOI] [PubMed] [Google Scholar]
  • [11•].Kojima M, Kangawa K. The discovery of ghrelin: with a little luck and great passion. Preface. Peptides. 2011;32:2153–2154. doi: 10.1016/j.peptides.2011.10.016. This article provides insight to historical steps that led to the discovery of ghrelin retraced by Kojima and Kangawa in a very personal way.
  • [12].Kojima M, Kangawa K. Ghrelin: structure and function. Physiol Rev. 2005;85:495–522. doi: 10.1152/physrev.00012.2004. [DOI] [PubMed] [Google Scholar]
  • [13].Davenport AP, Bonner TI, Foord SM, et al. International Union of Pharmacology. LVI. Ghrelin receptor nomenclature, distribution, and function. Pharmacol Rev. 2005;57:541–546. doi: 10.1124/pr.57.4.1. [DOI] [PubMed] [Google Scholar]
  • [14].Bednarek MA, Feighner SD, Pong SS, 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;43:4370–4376. doi: 10.1021/jm0001727. [DOI] [PubMed] [Google Scholar]
  • [15].Hosoda H, Kojima M, Matsuo H, Kangawa K. Ghrelin and des-acyl ghrelin: two major forms of rat ghrelin peptide in gastrointestinal tissue. Biochem Biophys Res Commun. 2000;279:909–913. doi: 10.1006/bbrc.2000.4039. [DOI] [PubMed] [Google Scholar]
  • [16].Raff H. Total and active ghrelin in developing rats during hypoxia. Endocrine. 2003;21:159–161. doi: 10.1385/ENDO:21:2:159. [DOI] [PubMed] [Google Scholar]
  • [17•].Stengel A, Keire D, Goebel M, et al. The RAPID method for blood processing yields new insight in plasma concentrations and molecular forms of circulating gut peptides. Endocrinology. 2009;150:5113–5118. doi: 10.1210/en.2009-0697. This article addresses a very relevant component of gut peptide determination: how blood processing after collection impacts on the concentrations and molecular forms of gut peptides detected after release (acyl vs. desacyl ghelin, PYY vs. PYY 3-36, CCK-8 vs. CCK-58).
  • [18].Yang J, Brown MS, Liang G, et al. Identification of the acyltransferase that octanoylates ghrelin, an appetite-stimulating peptide hormone. Cell. 2008;132:387–396. doi: 10.1016/j.cell.2008.01.017. [DOI] [PubMed] [Google Scholar]
  • [19].Gutierrez JA, Solenberg PJ, Perkins DR, et al. Ghrelin octanoylation mediated by an orphan lipid transferase. Proc Natl Acad Sci U S A. 2008;105:6320–6325. doi: 10.1073/pnas.0800708105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Sakata I, Yang J, Lee CE, et al. Colocalization of ghrelin O-acyltransferase and ghrelin in gastric mucosal cells. Am J Physiol Endocrinol Metab. 2009;297:E134–141. doi: 10.1152/ajpendo.90859.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Stengel A, Goebel M, Wang L, et al. Differential distribution of ghrelin-O-acyltransferase (GOAT) immunoreactive cells in the mouse and rat gastric oxyntic mucosa. Biochem Biophys Res Commun. 2010;392:67–71. doi: 10.1016/j.bbrc.2009.12.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Lim CT, Kola B, Grossman A, Korbonits M. The expression of ghrelin O-acyltransferase (GOAT) in human tissues. Endocr J. 2011;58:707–710. doi: 10.1507/endocrj.k11e-117. [DOI] [PubMed] [Google Scholar]
  • [23].Kang K, Schmahl J, Lee JM, et al. Mouse ghrelin-O-acyltransferase (GOAT) plays a critical role in bile acid reabsorption. Faseb J. 2012;26:259–271. doi: 10.1096/fj.11-191460. [DOI] [PubMed] [Google Scholar]
  • [24].Zhao CM, Furnes MW, Stenstrom B, et al. Characterization of obestatin- and ghrelin-producing cells in the gastrointestinal tract and pancreas of rats: an immunohistochemical and electron-microscopic study. Cell Tissue Res. 2008;331:575–587. doi: 10.1007/s00441-007-0514-3. [DOI] [PubMed] [Google Scholar]
  • [25].Gronberg M, Tsolakis AV, Magnusson L, et al. Distribution of obestatin and ghrelin in human tissues: immunoreactive cells in the gastrointestinal tract, pancreas, and mammary glands. J Histochem Cytochem. 2008;56:793–801. doi: 10.1369/jhc.2008.951145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Tsolakis AV, Grimelius L, Stridsberg M, et al. Obestatin/ghrelin cells in normal mucosa and endocrine tumours of the stomach. Eur J Endocrinol. 2009;160:941–949. doi: 10.1530/EJE-09-0001. [DOI] [PubMed] [Google Scholar]
  • [27].Soares JB, Leite-Moreira AF. Ghrelin, des-acyl ghrelin and obestatin: Three pieces of the same puzzle. Peptides. 2008;29:1255–1270. doi: 10.1016/j.peptides.2008.02.018. [DOI] [PubMed] [Google Scholar]
  • [28].Oh-I S, Shimizu H, Satoh T, et al. Identification of nesfatin-1 as a satiety molecule in the hypothalamus. Nature. 2006;443:709–712. doi: 10.1038/nature05162. [DOI] [PubMed] [Google Scholar]
  • [29].Brailoiu GC, Dun SL, Brailoiu E, et al. Nesfatin-1: distribution and interaction with a G protein-coupled receptor in the rat brain. Endocrinology. 2007;148:5088–5094. doi: 10.1210/en.2007-0701. [DOI] [PubMed] [Google Scholar]
  • [30].Foo K, Brismar H, Broberger C. Distribution and neuropeptide coexistence of nucleobindin-2 mRNA/nesfatin-like immunoreactivity in the rat CNS. Neuroscience. 2008;156:563–579. doi: 10.1016/j.neuroscience.2008.07.054. [DOI] [PubMed] [Google Scholar]
  • [31].Kohno D, Nakata M, Maejima Y, et al. Nesfatin-1 neurons in paraventricular and supraoptic nuclei of the rat hypothalamus coexpress oxytocin and vasopressin and are activated by refeeding. Endocrinology. 2008;149:1295–1301. doi: 10.1210/en.2007-1276. [DOI] [PubMed] [Google Scholar]
  • [32].Osaki A, Shimizu H, Ishizuka N, et al. Enhanced expression of nesfatin/nucleobindin-2 in white adipose tissue of ventromedial hypothalamus-lesioned rats. Neurosci Lett. 2012 doi: 10.1016/j.neulet.2012.05.056. [DOI] [PubMed] [Google Scholar]
  • [33].Shimizu H, Oh IS, Okada S, Mori M. Nesfatin-1: An Overview and Future Clinical Application. Endocr J. 2009;56:537–543. doi: 10.1507/endocrj.k09e-117. [DOI] [PubMed] [Google Scholar]
  • [34].Kerbel B, Unniappan S. Nesfatin-1 suppresses energy intake, co-localises ghrelin in the brain and gut, and alters ghrelin, cholecystokinin and orexin mRNA expression in goldfish. J Neuroendocrinol. 2012;24:366–377. doi: 10.1111/j.1365-2826.2011.02246.x. [DOI] [PubMed] [Google Scholar]
  • [35].Jeon TY, Lee S, Kim HH, et al. Changes in plasma ghrelin concentration immediately after gastrectomy in patients with early gastric cancer. J Clin Endocrinol Metab. 2004;89:5392–5396. doi: 10.1210/jc.2004-0872. [DOI] [PubMed] [Google Scholar]
  • [36].Date Y, Nakazato M, Hashiguchi S, et al. Ghrelin is present in pancreatic alpha-cells of humans and rats and stimulates insulin secretion. Diabetes. 2002;51:124–129. doi: 10.2337/diabetes.51.1.124. [DOI] [PubMed] [Google Scholar]
  • [37].Gnanapavan S, Kola B, Bustin SA, et al. The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in humans. J Clin Endocrinol Metab. 2002;87:2988. doi: 10.1210/jcem.87.6.8739. [DOI] [PubMed] [Google Scholar]
  • [38].Barreiro ML, Gaytan F, Caminos JE, et al. Cellular location and hormonal regulation of ghrelin expression in rat testis. Biol Reprod. 2002;67:1768–1776. doi: 10.1095/biolreprod.102.006965. [DOI] [PubMed] [Google Scholar]
  • [39].Cummings DE, Purnell JQ, Frayo RS, et al. A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes. 2001;50:1714–1719. doi: 10.2337/diabetes.50.8.1714. [DOI] [PubMed] [Google Scholar]
  • [40].Tschöp M, Wawarta R, Riepl RL, et al. Post-prandial decrease of circulating human ghrelin levels. J Endocrinol Invest. 2001;24:RC19–21. doi: 10.1007/BF03351037. [DOI] [PubMed] [Google Scholar]
  • [41].Schussler P, Kluge M, Yassouridis A, et al. Ghrelin levels increase after pictures showing food. Obesity (Silver Spring) 2012;20:1212–1217. doi: 10.1038/oby.2011.385. [DOI] [PubMed] [Google Scholar]
  • [42••].Xu G, Li Y, An W, et al. Gastric mammalian target of rapamycin signaling regulates ghrelin production and food intake. Endocrinology. 2009;150:3637–3644. doi: 10.1210/en.2009-0372. This is the first demonstration of the reciprocal relationship between gastric mTOR signaling and ghrelin during changes in energy status which provides a link between the gastric fuel sensor and the impact on energy intake through ghrelin release.
  • [43].Toshinai K, Mondal MS, Nakazato M, et al. Upregulation of ghrelin expression in the stomach upon fasting, insulin-induced hypoglycemia, and leptin administration. Biochem Biophys Res Commun. 2001;281:1220–1225. doi: 10.1006/bbrc.2001.4518. [DOI] [PubMed] [Google Scholar]
  • [44].Kim MS, Yoon CY, Park KH, et al. Changes in ghrelin and ghrelin receptor expression according to feeding status. Neuroreport. 2003;14:1317–1320. doi: 10.1097/01.wnr.0000078703.79393.d2. [DOI] [PubMed] [Google Scholar]
  • [45].Gonzalez CR, Vazquez MJ, Lopez M, Dieguez C. Influence of chronic undernutrition and leptin on GOAT mRNA levels in rat stomach mucosa. J Mol Endocrinol. 2008;41:415–421. doi: 10.1677/JME-08-0102. [DOI] [PubMed] [Google Scholar]
  • [46].Tschöp M, Smiley DL, Heiman ML. Ghrelin induces adiposity in rodents. Nature. 2000;407:908–913. doi: 10.1038/35038090. [DOI] [PubMed] [Google Scholar]
  • [47].Cummings DE, Weigle DS, Frayo RS, et al. Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. N Engl J Med. 2002;346:1623–1630. doi: 10.1056/NEJMoa012908. [DOI] [PubMed] [Google Scholar]
  • [48].Tschöp M, Weyer C, Tataranni PA, et al. Circulating ghrelin levels are decreased in human obesity. Diabetes. 2001;50:707–709. doi: 10.2337/diabetes.50.4.707. [DOI] [PubMed] [Google Scholar]
  • [49].Mizutani M, Atsuchi K, Asakawa A, et al. Localization of acyl ghrelin- and des-acyl ghrelin-immunoreactive cells in the rat stomach and their responses to intragastric pH. Am J Physiol Gastrointest Liver Physiol. 2009;297:G974–980. doi: 10.1152/ajpgi.00147.2009. [DOI] [PubMed] [Google Scholar]
  • [50].Stengel A, Goebel-Stengel M, Wang L, et al. Abdominal surgery inhibits circulating acyl ghrelin and ghrelin-O-acyltransferase levels in rats: role of the somatostatin receptor subtype 2. Am J Physiol Gastrointest Liver Physiol. 2011;301:G239–248. doi: 10.1152/ajpgi.00018.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Stengel A, Goebel M, Wang L, et al. Lipopolysaccharide differentially decreases plasma acyl and desacyl ghrelin levels in rats: Potential role of the circulating ghrelin-acylating enzyme GOAT. Peptides. 2010;31:1689–1696. doi: 10.1016/j.peptides.2010.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Saidpour A, Zahediasl S, Kimiagar M, et al. Fish oil and olive oil can modify insulin resistance and plasma desacyl-ghrelin in rats. J Res Med Sci. 2011;16:862–871. [PMC free article] [PubMed] [Google Scholar]
  • [53].Iwakura H, Li Y, Ariyasu H, et al. Establishment of a novel ghrelin-producing cell line. Endocrinology. 2010;151:2940–2945. doi: 10.1210/en.2010-0090. [DOI] [PubMed] [Google Scholar]
  • [54].Iwakura H, Ariyasu H, Hosoda H, et al. Oxytocin and dopamine stimulate ghrelin secretion by the ghrelin-producing cell line, MGN3-1 in vitro. Endocrinology. 2011;152:2619–2625. doi: 10.1210/en.2010-1455. [DOI] [PubMed] [Google Scholar]
  • [55].Wang L, Murphy NP, Stengel A, et al. Ghrelin prevents levodopa-induced inhibition of gastric emptying and increases circulating levodopa in fasted rats. Neurogastroenterol Motil. 2012 doi: 10.1111/j.1365-2982.2012.01904.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].de la Cour CD, Norlen P, Hakanson R. Secretion of ghrelin from rat stomach ghrelin cells in response to local microinfusion of candidate messenger compounds: a microdialysis study. Regul Pept. 2007;143:118–126. doi: 10.1016/j.regpep.2007.05.001. [DOI] [PubMed] [Google Scholar]
  • [57 ••].Lu X, Zhao X, Feng J, et al. Postprandial Inhibition of Gastric Ghrelin Secretion by Long-chain Fatty Acid (LCFA) Through GPR 120 in Isolated Gastric Ghrelin Cells and Mice. Am J Physiol Gastrointest Liver Physiol. 2012 doi: 10.1152/ajpgi.00541.2011. This is one of the first generations of transgenic mice with green fluorescent protein (GFP) expressing ghrelin cells enabling to show the regulation of ghrelin release by long chain free fatty acids in a pure population of isolated gastric ghrelin expressing GFP cells.
  • [58].Hattori N, Saito T, Yagyu T, et al. GH, GH receptor, GH secretagogue receptor, and ghrelin expression in human T cells, B cells, and neutrophils. J Clin Endocrinol Metab. 2001;86:4284–4291. doi: 10.1210/jcem.86.9.7866. [DOI] [PubMed] [Google Scholar]
  • [59].Schellekens H, Dinan TG, Cryan JF. Lean Mean Fat Reducing “Ghrelin” Machine: Hypothalamic Ghrelin and Ghrelin Receptors as Therapeutic Targets in Obesity. Neuropharmacology. 2010;58:2–16. doi: 10.1016/j.neuropharm.2009.06.024. [DOI] [PubMed] [Google Scholar]
  • [60].Xu L, Depoortere I, Tomasetto C, et al. Evidence for the presence of motilin, ghrelin, and the motilin and ghrelin receptor in neurons of the myenteric plexus. Regul Pept. 2005;124:119–125. doi: 10.1016/j.regpep.2004.07.022. [DOI] [PubMed] [Google Scholar]
  • [61].Camina JP, Carreira MC, El Messari S, et al. Desensitization and endocytosis mechanisms of ghrelin-activated growth hormone secretagogue receptor 1a. Endocrinology. 2004;145:930–940. doi: 10.1210/en.2003-0974. [DOI] [PubMed] [Google Scholar]
  • [62].Holst B, Cygankiewicz A, Jensen TH, et al. High constitutive signaling of the ghrelin receptor--identification of a potent inverse agonist. Mol Endocrinol. 2003;17:2201–2210. doi: 10.1210/me.2003-0069. [DOI] [PubMed] [Google Scholar]
  • [63•].Chollet C, Bergmann R, Pietzsch J, Beck-Sickinger AG. Design, evaluation, and comparison of ghrelin receptor agonists and inverse agonists as suitable radiotracers for PET imaging. Bioconjug Chem. 2012;23:771–784. doi: 10.1021/bc2005889. The authors discuss that the discoverey of novel ghrelin inverse agonists with featured of high stability in blood, slow blood clearance and large diffusion in tissues may be a promising approach to reduce ghrelin and ghrelin receptor signaling.
  • [64].Gauna C, Delhanty PJ, van Aken MO, et al. Unacylated ghrelin is active on the INS-1 E rat insulinoma cell line independently of the growth hormone secretagogue receptor type 1a and the corticotropin releasing factor 2 receptor. Mol Cell Endocrinol. 2006;251:103–111. doi: 10.1016/j.mce.2006.03.040. [DOI] [PubMed] [Google Scholar]
  • [65].Cassoni P, Papotti M, Ghe C, 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;86:1738–1745. doi: 10.1210/jcem.86.4.7402. [DOI] [PubMed] [Google Scholar]
  • [66].Gourcerol G, Taché Y. Obestatin--a ghrelin-associated peptide that does not hold its promise to suppress food intake and motility. Neurogastroenterol Motil. 2007;19:161–165. doi: 10.1111/j.1365-2982.2007.00916.x. [DOI] [PubMed] [Google Scholar]
  • [67].Zhang JV, Jahr H, Luo CW, et al. Obestatin induction of early-response gene expression in gastrointestinal and adipose tissues and the mediatory role of G protein-coupled receptor, GPR39. Mol Endocrinol. 2008;22:1464–1475. doi: 10.1210/me.2007-0569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Lauwers E, Landuyt B, Arckens L, et al. Obestatin does not activate orphan G protein-coupled receptor GPR39. Biochem Biophys Res Commun. 2006;351:21–25. doi: 10.1016/j.bbrc.2006.09.141. [DOI] [PubMed] [Google Scholar]
  • [69].Tremblay F, Perreault M, Klaman LD, et al. Normal food intake and body weight in mice lacking the G protein-coupled receptor GPR39. Endocrinology. 2007;148:501–506. doi: 10.1210/en.2006-1275. [DOI] [PubMed] [Google Scholar]
  • [70].Holst B, Egerod KL, Schild E, et al. GPR39 signaling is stimulated by zinc ions but not by obestatin. Endocrinology. 2007;148:13–20. doi: 10.1210/en.2006-0933. [DOI] [PubMed] [Google Scholar]
  • [71].Chartrel N, Alvear-Perez R, Leprince J, et al. Comment on “Obestatin, a peptide encoded by the ghrelin gene, opposes ghrelin’s effects on food intake”. Science. 2007;315:766. doi: 10.1126/science.1135047. author reply 766. [DOI] [PubMed] [Google Scholar]
  • [72].Li Z, Xu G, Li Y, et al. mTOR-dependent modulation of gastric nesfatin-1/NUCB2. Cell Physiol Biochem. 2012;29:493–500. doi: 10.1159/000338503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Tsuchiya T, Shimizu H, Yamada M, et al. Fasting concentrations of nesfatin-1 are negatively correlated with body mass index in non-obese males. Clin Endocrinol (Oxf) 2010;73:484–490. doi: 10.1111/j.1365-2265.2010.03835.x. [DOI] [PubMed] [Google Scholar]
  • [74].Li QC, Wang HY, Chen X, et al. Fasting plasma levels of nesfatin-1 in patients with type 1 and type 2 diabetes mellitus and the nutrient-related fluctuation of nesfatin-1 level in normal humans. Regul Pept. 2010;159:72–77. doi: 10.1016/j.regpep.2009.11.003. [DOI] [PubMed] [Google Scholar]
  • [75].Ogiso K, Asakawa A, Amitani H, et al. Plasma nesfatin-1 concentrations in restricting-type anorexia nervosa. Peptides. 2010;32:150–153. doi: 10.1016/j.peptides.2010.10.004. [DOI] [PubMed] [Google Scholar]
  • [76].Ramanjaneya M, Chen J, Brown JE, et al. Identification of nesfatin-1 in human and murine adipose tissue: a novel depot-specific adipokine with increased levels in obesity. Endocrinology. 2010;151:3169–3180. doi: 10.1210/en.2009-1358. [DOI] [PubMed] [Google Scholar]
  • [77].Tan BK, Hallschmid M, Kern W, et al. Decreased cerebrospinal fluid/plasma ratio of the novel satiety molecule, nesfatin-1/NUCB-2, in obese humans: evidence of nesfatin-1/NUCB-2 resistance and implications for obesity treatment. J Clin Endocrinol Metab. 2011;96:E669–673. doi: 10.1210/jc.2010-1782. [DOI] [PubMed] [Google Scholar]
  • [78].Garcia-Galiano D, Navarro VM, Gaytan F, Tena-Sempere M. Expanding roles of NUCB2/nesfatin-1 in neuroendocrine regulation. J Mol Endocrinol. 2010;45:281–290. doi: 10.1677/JME-10-0059. [DOI] [PubMed] [Google Scholar]
  • [79].Wren AM, Small CJ, Ward HL, et al. The novel hypothalamic peptide ghrelin stimulates food intake and growth hormone secretion. Endocrinology. 2000;141:4325–4328. doi: 10.1210/endo.141.11.7873. [DOI] [PubMed] [Google Scholar]
  • [80].Tang-Christensen M, Vrang N, Ortmann S, et al. Central administration of ghrelin and agouti-related protein (83-132) increases food intake and decreases spontaneous locomotor activity in rats. Endocrinology. 2004;145:4645–4652. doi: 10.1210/en.2004-0529. [DOI] [PubMed] [Google Scholar]
  • [81].Druce MR, Wren AM, Park AJ, et al. Ghrelin increases food intake in obese as well as lean subjects. Int J Obes (Lond) 2005;29:1130–1136. doi: 10.1038/sj.ijo.0803001. [DOI] [PubMed] [Google Scholar]
  • [82].Salome N, Haage D, Perrissoud D, et al. Anorexigenic and electrophysiological actions of novel ghrelin receptor (GHS-R1A) antagonists in rats. Eur J Pharmacol. 2009;612:167–173. doi: 10.1016/j.ejphar.2009.03.066. [DOI] [PubMed] [Google Scholar]
  • [83].Sun Y, Wang P, Zheng H, Smith RG. Ghrelin stimulation of growth hormone release and appetite is mediated through the growth hormone secretagogue receptor. Proc Natl Acad Sci U S A. 2004;101:4679–4684. doi: 10.1073/pnas.0305930101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Zigman JM, Nakano Y, Coppari R, et al. Mice lacking ghrelin receptors resist the development of diet-induced obesity. J Clin Invest. 2005;115:3564–3572. doi: 10.1172/JCI26002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Davies JS, Kotokorpi P, Eccles SR, et al. Ghrelin induces abdominal obesity via GHS-R-dependent lipid retention. Mol Endocrinol. 2009;23:914–924. doi: 10.1210/me.2008-0432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Strassburg S, Anker S, Castaneda T, et al. Long-term effects of ghrelin and ghrelin receptor agonists on energy balance in rats. Am J Physiol Endocrinol Metab. 2008;295:E78–84. doi: 10.1152/ajpendo.00040.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Theander-Carrillo C, Wiedmer P, Cettour-Rose P, et al. Ghrelin action in the brain controls adipocyte metabolism. J Clin Invest. 2006;116:1983–1993. doi: 10.1172/JCI25811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88 ••].Kirchner H, Gutierrez JA, Solenberg PJ, et al. GOAT links dietary lipids with the endocrine control of energy balance. Nat Med. 2009;15:741–745. doi: 10.1038/nm.1997. This study provides new insight to the ghrelin-GOAT system as a signaling pathway to indicate the presence of dietary calories, rather than their absence.
  • [89].Banks WA, Tschöp M, Robinson SM, Heiman ML. Extent and direction of ghrelin transport across the blood-brain barrier is determined by its unique primary structure. J Pharmacol Exp Ther. 2002;302:822–827. doi: 10.1124/jpet.102.034827. [DOI] [PubMed] [Google Scholar]
  • [90].Pan W, Tu H, Kastin AJ. Differential BBB interactions of three ingestive peptides: obestatin, ghrelin, and adiponectin. Peptides. 2006;27:911–916. doi: 10.1016/j.peptides.2005.12.014. [DOI] [PubMed] [Google Scholar]
  • [91].Date Y, Murakami N, Toshinai K, et al. The role of the gastric afferent vagal nerve in ghrelin-induced feeding and growth hormone secretion in rats. Gastroenterology. 2002;123:1120–1128. doi: 10.1053/gast.2002.35954. [DOI] [PubMed] [Google Scholar]
  • [92].Sakata I, Yamazaki M, Inoue K, et al. Growth hormone secretagogue receptor expression in the cells of the stomach-projected afferent nerve in the rat nodose ganglion. Neurosci Lett. 2003;342:183–186. doi: 10.1016/s0304-3940(03)00294-5. [DOI] [PubMed] [Google Scholar]
  • [93].Arnold M, Mura A, Langhans W, Geary N. Gut vagal afferents are not necessary for the eating-stimulatory effect of intraperitoneally injected ghrelin in the rat. J Neurosci. 2006;26:11052–11060. doi: 10.1523/JNEUROSCI.2606-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Cowley MA, Smith RG, Diano S, et al. The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis. Neuron. 2003;37:649–661. doi: 10.1016/s0896-6273(03)00063-1. [DOI] [PubMed] [Google Scholar]
  • [95].Lu S, Guan JL, Wang QP, et al. Immunocytochemical observation of ghrelin-containing neurons in the rat arcuate nucleus. Neurosci Lett. 2002;321:157–160. doi: 10.1016/s0304-3940(01)02544-7. [DOI] [PubMed] [Google Scholar]
  • [96].Schwartz MW, Woods SC, Porte D, Jr., et al. Central nervous system control of food intake. Nature. 2000;404:661–671. doi: 10.1038/35007534. [DOI] [PubMed] [Google Scholar]
  • [97].Guan JL, Wang QP, Kageyama H, et al. Synaptic interactions between ghrelin- and neuropeptide Y-containing neurons in the rat arcuate nucleus. Peptides. 2003;24:1921–1928. doi: 10.1016/j.peptides.2003.10.017. [DOI] [PubMed] [Google Scholar]
  • [98].Wang L, Saint-Pierre DH, Taché Y. Peripheral ghrelin selectively increases Fos expression in neuropeptide Y - synthesizing neurons in mouse hypothalamic arcuate nucleus. Neurosci Lett. 2002;325:47–51. doi: 10.1016/s0304-3940(02)00241-0. [DOI] [PubMed] [Google Scholar]
  • [99].Kamegai J, Tamura H, Shimizu T, et al. Chronic central infusion of ghrelin increases hypothalamic neuropeptide Y and agouti-related protein mRNA levels and body weight in rats. Diabetes. 2001;50:2438–2443. doi: 10.2337/diabetes.50.11.2438. [DOI] [PubMed] [Google Scholar]
  • [100].Nakazato M, Murakami N, Date Y, et al. A role for ghrelin in the central regulation of feeding. Nature. 2001;409:194–198. doi: 10.1038/35051587. [DOI] [PubMed] [Google Scholar]
  • [101].Chen HY, Trumbauer ME, Chen AS, et al. Orexigenic action of peripheral ghrelin is mediated by neuropeptide Y and agouti-related protein. Endocrinology. 2004;145:2607–2612. doi: 10.1210/en.2003-1596. [DOI] [PubMed] [Google Scholar]
  • [102].Chen CY, Inui A, Asakawa A, et al. Des-acyl ghrelin acts by CRF type 2 receptors to disrupt fasted stomach motility in conscious rats. Gastroenterology. 2005;129:8–25. doi: 10.1053/j.gastro.2005.04.015. [DOI] [PubMed] [Google Scholar]
  • [103].Asakawa A, Inui A, Fujimiya M, et al. Stomach regulates energy balance via acylated ghrelin and desacyl ghrelin. Gut. 2005;54:18–24. doi: 10.1136/gut.2004.038737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Toshinai K, Yamaguchi H, Sun Y, et al. Des-acyl ghrelin induces food intake by a mechanism independent of the growth hormone secretagogue receptor. Endocrinology. 2006;147:2306–2314. doi: 10.1210/en.2005-1357. [DOI] [PubMed] [Google Scholar]
  • [105].Inhoff T, Mönnikes H, Noetzel S, et al. Desacyl ghrelin inhibits the orexigenic effect of peripherally injected ghrelin in rats. Peptides. 2008;29:2159–2168. doi: 10.1016/j.peptides.2008.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Zhang W, Chai B, Li JY, et al. Effect of des-acyl ghrelin on adiposity and glucose metabolism. Endocrinology. 2008;149:4710–4716. doi: 10.1210/en.2008-0263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Ariyasu H, Takaya K, Iwakura H, et al. Transgenic mice overexpressing des-acyl ghrelin show small phenotype. Endocrinology. 2005;146:355–364. doi: 10.1210/en.2004-0629. [DOI] [PubMed] [Google Scholar]
  • [108].Rodriguez A, Gomez-Ambrosi J, Catalan V, et al. Acylated and desacyl ghrelin stimulate lipid accumulation in human visceral adipocytes. Int J Obes (Lond) 2009;33:541–552. doi: 10.1038/ijo.2009.40. [DOI] [PubMed] [Google Scholar]
  • [109].Chen CY, Tsai CY, Lee WJ, et al. Intracerebroventricular O-n-octanoylated ghrelin and its splice variant-induced feeding is blocked by insulin, independent of obestatin or CRF receptor, in satiated rats. Nutrition. 2012;28:812–820. doi: 10.1016/j.nut.2011.11.021. [DOI] [PubMed] [Google Scholar]
  • [110].Stengel A, Goebel M, Wang L, et al. Central nesfatin-1 reduces dark-phase food intake and gastric emptying in rats: differential role of corticotropin-releasing factor2 receptor. Endocrinology. 2009;150:4911–4919. doi: 10.1210/en.2009-0578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Yosten GL, Samson WK. The anorexigenic and hypertensive effects of nesfatin-1 are reversed by pretreatment with an oxytocin receptor antagonist. Am J Physiol Regul Integr Comp Physiol. 2010;298:R1642–1647. doi: 10.1152/ajpregu.00804.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Atsuchi K, Asakawa A, Ushikai M, et al. Centrally administered nesfatin-1 inhibits feeding behaviour and gastroduodenal motility in mice. Neuroreport. 2010;21:1008–1011. doi: 10.1097/WNR.0b013e32833f7b96. [DOI] [PubMed] [Google Scholar]
  • [113].Goebel M, Stengel A, Wang L, Taché Y. Central nesfatin-1 reduces the nocturnal food intake in mice by reducing meal size and increasing inter-meal intervals. Peptides. 2011;32:36–43. doi: 10.1016/j.peptides.2010.09.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Gonzalez R, Kerbel B, Chun A, Unniappan S. Molecular, cellular and physiological evidences for the anorexigenic actions of nesfatin-1 in goldfish. PLoS One. 2010;5:e15201. doi: 10.1371/journal.pone.0015201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Yosten GL, Samson WK. Nesfatin-1 exerts cardiovascular actions in brain: possible interaction with the central melanocortin system. Am J Physiol Regul Integr Comp Physiol. 2009;297:R330–336. doi: 10.1152/ajpregu.90867.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Maejima Y, Sedbazar U, Suyama S, et al. Nesfatin-1-regulated oxytocinergic signaling in the paraventricular nucleus causes anorexia through a leptin-independent melanocortin pathway. Cell Metab. 2009;10:355–365. doi: 10.1016/j.cmet.2009.09.002. [DOI] [PubMed] [Google Scholar]
  • [117].Stengel A, Goebel-Stengel M, Wang L, et al. Nesfatin-1(30-59) but not the N- and C-terminal fragments, nesfatin-1(1-29) and nesfatin-1(60-82) injected intracerebroventricularly decreases dark phase food intake by increasing inter-meal intervals in mice. Peptides. 2012;35:143–148. doi: 10.1016/j.peptides.2012.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Nonogaki K, Ohba Y, Sumii M, Oka Y. Serotonin systems upregulate the expression of hypothalamic NUCB2 via 5-HT2C receptors and induce anorexia via a leptin-independent pathway in mice. Biochem Biophys Res Commun. 2008;372:186–190. doi: 10.1016/j.bbrc.2008.05.010. [DOI] [PubMed] [Google Scholar]
  • [119].Shimizu H, Oh-I S, Hashimoto K, et al. Peripheral administration of nesfatin-1 reduces food intake in mice: the leptin-independent mechanism. Endocrinology. 2009;150:662–671. doi: 10.1210/en.2008-0598. [DOI] [PubMed] [Google Scholar]
  • [120].Iwasaki Y, Nakabayashi H, Kakei M, et al. Nesfatin-1 evokes Ca2+ signaling in isolated vagal afferent neurons via Ca2+ influx through N-type channels. Biochem Biophys Res Commun. 2009;390:958–962. doi: 10.1016/j.bbrc.2009.10.085. [DOI] [PubMed] [Google Scholar]

RESOURCES