Abstract
Background:
Ghrelin, a unique 28 amino acid peptide hormone secreted by the gastric X/A like cells, is an endogenous ligand of the growth hormone secretagogue receptor (GHSR). Ghrelin-GHSR signaling has been found to exert various physiological functions, including stimulation of appetite, regulation of body weight, lipid and glucose metabolism, and increase of gut motility and secretion. This system is thus critical for energy homeostasis.
Objective:
The objective of this review is to highlight the strategies of ghrelin-GHSR based intervention for therapy of obesity and its related metabolic diseases.
Results:
Therapeutic strategies of metabolic disorders targeting the ghrelin-GHSR pathway involve neutralization of circulating ghrelin by antibodies and RNA spiegelmers, antagonism of ghrelin receptor by its antagonists and inverse agonists, inhibition of ghrelin O-acyltransferase (GOAT), as well as potential pharmacological approach to decrease ghrelin synthesis and secretion.
Conclusion:
Various compounds targeting the ghrelin-GHSR system have shown promising efficacy for the intervention of obesity and relevant metabolic disorders in animals and in vitro. Further clinical trials to validate their efficacy in human beings are urgently needed.
Keywords: Ghrelin, growth hormone secretagogue receptor (GHSR), therapeutic strategies, obesity, body weight, food intake
1. INTRODUCTION
Obesity, a major risk factor of non-communicable diseases, reduces the expected life span by 5 to 20 years, depending on the severity of the condition and its associated comorbidities [1-4]. Current therapy of obesity focuses mainly on weight management, with limited effect. Defining a novel strategy that could provide a lasting effect of weight reduction is urgently needed.
Ghrelin, an acylated peptide hormone, was first discovered in the gastric X/A like endocrine cells through a reverse pharmacological approach in 1999 [5]. It is the endogenous ligand of growth hormone secretagogue receptor (GHSR), a seven-transmembraneG protein-coupled receptor, which is now named as ghrelin receptor (GRLN-R) [6]. This receptor has demonstrated high expression in the pituitary gland, pancreatic islets, heart, thyroid gland and a variety of brain regions [7-12]. X/A like cells are the second abundant type of endocrine cells in the stomach, found in the gastric fundus, accounting for 20-30% of the oxyntic endocrine cells. These cells synthesize and release ghrelin [13]. Ghrelin is also expressed in alpha cells of the embryonic pancreas [14], as well as in pituitary [15, 16], hypothalamus [17, 18], kidney [19], liver [9], spleen, heart [20], lung [21], adipose tissue, skin [9] and testis [9, 22-24] in lower amounts.
Ghrelin gene composing five exons and four introns have been demonstrated to encode prepro-ghrelin of 117-amino acid. Prepro-ghrelin is cleaved to proghrelin and a signal peptide of 23-amino acid subsequently [5]. In golgi apparatus, pro-ghrelin is cleaved by prohormone convertases 1/3 (pc1/3) to produce mature ghrelin containing 28 amino acids [25]. Ghrelin binds to GHSR to exert its biological action. The acylation of ghrelin by ghrelin O-acyl-transferase (GOAT) to attach octanoate to serine-3 of ghrelin is necessary for ghrelin to bind to GHSR [26, 27]. The acylated ghrelin could not be detected in the circulation of GOAT deficiency mice, indicating that GOAT is the only enzyme capable of octanoylate ghrelin in vivo [28, 29]. However, not all ghrelin is octanoylated. Actually, only a fraction (less than 10%) of the ghrelin in circulation is acylated [30]. The unmodified des-n-octanoyl form of ghrelin (desacyl ghrelin) does not activate GHSR [23]. The acylated ghrelin increases food intake, gastric emptying, lipogenesis, blood glucose and insulin resistance, as well as reduces thermogenesis and insulin secretion [31]. Conversely, desacyl ghrelin decreases food intake, gastric emptying, lipogenesis, blood glucose and insulin resistance, while stimulates thermogenesis and insulin secretion via a yet-to-be-defined receptor [32].
Subcutaneous or intraventricular injection of ghrelin increases body weight by stimulating food intake [33-35] and inhibiting fat utilization in rodents [31, 36]. Subsequent studies have showed that ghrelin increases lipid deposition by concurrently increasing mRNA expression of enzymes responsible for fat storage and decreasing carnitine palmitoyltransferase-1a levels to reduce β-oxidation and lipolysis [37-39]. In the hypothalamic feeding center, GHSR1a is localized in neurons expressing neuropeptide Y (NPY) and agoutirelated peptide (AGRP), the orexigenic neuropeptides [40]. In humans, the anteprandial rise and postprandial fall of ghrelin indicate that ghrelin is a signal of hanger that promotes meal initiation [41]. In addition to stimulation of food intake and weight gain, the ghrelin-GHSR axis has also been demonstrated to regulate glucose and lipid metabolism via mTOR signal pathway [42-45], indicating a critical role in energy homeostasis. Thus, a strategy targeting the ghrelin-GHSR system may hold promise for the therapy of obesity and its related metabolic diseases. The present review summarizes the recent advances in ghrelin-GHSR based intervention of energy imbalance.
2. NEUTRALIZATION OF GHRELIN
2.1. Antibodies of Ghrelin
Traditionally, vaccination has been used to prevent infectious diseases. This notion has recently been redefined to allow vaccination as a pharmacological application to regulate body weight. Antibodies directed against acylated ghrelin were produced by active vaccination of rats through the injection of ghrelin immunoconjugates. The ghrelin antibody produced has been demonstrated to decrease significantly feeding efficiency, and weight gain [46]. A similar effect has been shown in pigs actively immunized against ghrelin, which reduces 10% of body weight and 15% of food intake relative to the control pigs at the end of the experiment [47]. Recently, another vaccine has been developed to lose weight targeting the ghrelin-GHSR system. The vaccine was developed in an advanced materialnanogel to allow intranasal administration. This approach reduces body weight in high fat diet-induced obese mice and ob/ob mice, thereby improving glucose tolerance and insulin sensitivity [48]. Unfortunately, ghrelin vaccine shows limited effect in the human phase IIIa clinical trial. Despite that strong antibody response to ghrelin reduced hunger, weight loss was not significant in 111 obese subjects [49]. Therefore, the clinical application of vaccination against ghrelin in the treatment of obesity remains to be proven [50].
2.2. RNA Spiegelmers
RNA spiegelmers (German Spiegel = mirror) are a unique brand of reagents to inhibit ghrelin action [51, 52]. Spiegelmers are oligonucleotides that are synthesized with unnatural L-enantiomers of ribose in the sugar-phosphate framework for stability. They arc designed to bind to acylated ghrelin, thereby blocking ghrelin receptor activation in vitro [53]. RNA spiegelmer has been shown to block the effect of ectogenic ghrelin on the activity of neurons in the hypothalamic arcuate nucleus [54]. Administration of ghrelin RNA spiegelmer into diet-induced obese mice results in weight loss and reduction of food intake and fat deposition relative to control mice during the first week of treatment. Spiegelmer NOX-B11-2 blocks the activation of GHSR1a in vitro and effectually accelerates weight loss in diet-induced obese mice [55]. However, the weight loss relapses during the second week of treatment. Another ghrelin RNA spiegelmer, NOX-B11-3, effectually blocks the stimulation of arcuate nucleus neurons induced by ghrelin in rats [54]. Further studies are required to explicate the therapeutic clinical effects of ghrelin RNA spiegelmers.
2.3. GHSR-Fc Fusion Protein
GHSR-Fc is a recombinant protein containing the ligand-binding domains of the GHSR1a fused with a human IgG constant region (Fc). The GHSR-Fc is secretable because its structure does not contain a sequence motif corresponding to the transmembrane domain of the receptor. In vivo expression of GHSR/Fc reduces levels of acylated ghrelin but not unacylated ghrelin. Thus, this fusion structure is able to deplete circulating acylated-ghrelin. Intramuscular injection of GHSR-Fc decreases weight gain in high fat diet-induced obese mice accompanied by reduction of peritoneal fat accumulation, alteration of gene expression in adipocyte and improvement of glucose tolerance [56]. GHSRl-Fc fusion protein may provide an alternative potential for the treatment of obesity.
3. PHARMACOLOGICAL TARGET OF GHRELIN RECEPTOR
3.1. Ghrelin Receptor Antagonists
Ghrelin is known to exercise its orexigenic and metabolic function through GHSR [57, 58]. Mice with GHSR deficiency show reduced fat deposition and healthier lipid structure, along with increasing energy expenditure and basal metabolic rate [59]. Thus, suppression of ghrelin-GHSR signaling is an attractive strategy for therapy of obesity, type 2 diabetes, and metabolic syndrome. Recently, various GHSR antagonists have been reported as new approaches for weight reduction (Table 1).
Table 1.
Ghrelin receptor antagonists.
| Chemical types | Compounds | Functions |
|---|---|---|
| Met-enkephalin-derived GH-releasing peptides | [D-Lys3]-growth hormone–releasing peptide 6 (GHRP-6) | 1. Increases cumulative food intake in non obese diabetic MKR mice [65]. 2. Reduces GH secretion, weight gain and plasma insulin [65]. 3. Attenuates alcohol-induced hepatic steatosis [69]. |
| Met-enkephalin-derived GRLN-R antagonists | JMV2810 JMV2844 JMV2959 JMV3002 JMV3021 |
Suppresses food intake [70-73]. |
| Ghrelin-derived ligand | BIM-28163 | Induces body weight gain [74]. |
| Small molecule GHSR antagonists-piperidine-substituted quinazolinone derivatives | YIL-781 | Improves glucose-stimulated insulin secretion and reduce food intake and weight loss [76]. |
| Compound D (CpdD) | Reduces food intake [77]. | |
| Compound B (CpdB) | Reduces body weight, white adipose tissue, hepatic steatosis and improves insulin sensitivity [77]. | |
| Carbohydrazide derivatives | GSK 1614343 | Increases weight gain and food intake [79]. |
[D-Lys3]-growth hormone-releasing peptide 6 ([D-Lys3]-GHRP-6) was discovered in 1992 during the study of met-enkephalin-derivatives of growth hormone-releasing peptide [60]. Its special amino acid sequence His-D-Trp-A1a-Trp-D-Phe-Lys-NH2 inhibits GHRP combining with the pituitary gland and membranes of hypothalamus [60]. In a recent study, [D-Lys3]-GHRP-6 has been shown to behave as a partial antagonist with a strong bias toward GHSR-1a-β-arrestin signaling [61]. Administration of [D-Lys3]-GHRP-6 reduces growth hormone secretion and weight gain in mice [62-64], as well as attenuates alcohol-induced hepatic steatosis by regulating lipid metabolism [65]. However, [D-Lys3]-GHRP-6 unexpectedly increases food intake and reduces plasma insulin levels, leading to increased blood glucose and insulin resistance [62]. This result casts a cloud on the use of [D-Lys3]-GHRP-6 as a new treatment option for weight control, mainly on account of its side effects relevant to glycemic control.
Another growth hormone secretagogue analogs, met-enkephalin-derived GHSR antagonists, with His-D2NaI-DLys-Trp-DPhe-LysNH2 amino acid sequence which carry 1, 2, 4-triazole structure, have also been studied in energy metabolism. For example, a subcutaneous injection of compound 21b (JMV2810) inhibits hexareline-stimulated food intake [66]. Similarly, JMV2844 (sc), JMV2959 (icv, sc, ip), JMV3002 (icv) and JMV3021 (sc) also suppress food intake in rats [66-69]. The effects of these antagonists on body weight control in humans remain unknown.
BIM-28163, a ghrelin-derived ligand, was identified as a GHSR antagonist blocking ghrelin-induccd GH secretion. However, BIM-28163 acts unexpectedly as a GHSR agonist to induce body weight gain in male SD rats [70].
The effect of small molecule GHSR antagonists, such as piperidine-substituted derivatives of quinazolinone seems promising [71]. YIL-781, a piperidine-substituted quinazolinone derivative, acts as an effective GHSR1a antagonist to improve insulin release and weight loss and to reduce food intake in obese mice [72]. Compound D (CpdD), another small molecule GHSR antagonist, leads to a transient reduction in food intake. Similarly, compound B (CpdB) imposes continuous weight loss on account of reduced fat mass, which is associated with improved insulin sensitivity and resistance to hepatic steatosis in diet-induced obese mice [73].
GHSR1a antagonists also include several derivatives of carbohydrazide [74]. However, GSK1614343, one of these compounds, increases weight gain and food intake in both rats and dogs [75]. Thus, their metabolic benefit requires further investigation.
Although short-term use of ghrelin receptor antagonists has shown promising effects for weight control, its long-term efficacy is limited.
3.2. Inverse Ghrelin Receptor Agonists
Inverse GHSR1a agonists may provide an alternative strategy for the intervention of obesity because of the constitutive ghrelin receptor activity, which could be reduced by its inverse agonists [76-78]. In the hypothalamus, prolonged fasting results in an increase of GHSR1a expression, leading to higher appetite and lower energy expenditure. Thus, reducing the constitutive activity of GHSR1a through inverse agonists improves the sensitivity to anorectic signals such as leptin, PYY or insulin and eliminates the hunger between meals and the craving for snacks [79]. [D-Arg-1, D-Phe-5, D-Trp-7, 9 and Leu-11]-substance P was identified as the first inverse agonist for GHSR1a [63]. This substance P derivate reduces food intake in lean, diet-induced obese or ob/ob mice [63].
Liver enriched antimicrobial peptide 2 (LEAP-2) is a potent antagonist of the GHSR. LEAP-2 blocks the major effect of ghrelin on food intake, and maintenance of glucose levels in a steady state during fasting in mice [80]. The n-terminus sequence of LEAP-2 acts as an inverse agonist of the GHSR. Subcutaneous administration of the n-terminus region of LEAP-2 inhibits the food intake induced by ghrelin [81]. In a recent study, Mani et al. found that LEAP-2 regulates body mass and food intake by changing the LEAP-2/acyl-ghrelin ratio in humans and mice [82]. Therefore, decreasing LEAP-2 degradation or increasing LEAP-2 expression may be another promising strategy to antagonize ghrelin’s effect.
A series of 2-alkylamino nicotinamide analogs has been defined as the inverse ghrelin receptor agonist with oral bioactivity. Among these, compound 33 (Table 2) decreases weight gain in obese rats [83]. As a GHSR inverse agonist, the core structure of compound 33 is a diazabicyclo ring at the 5-position of the pyridine ring. Oral administration of compound 33 displays a moderate concentration in brain, suggesting its brain permeability [84].
Table 2.
Inverse ghrelin receptor agonists.
| Chemical types | Compounds | Functions |
|---|---|---|
| Substance P derivate | [D-Argl, D-Phe5, D-Trp7,9, Leul 1]-substance P | Reduces food intake [66] |
| Endogenous full antagonist of the GHSR | Antimicrobial peptide 2 (LEAP-2) | Reduces food intake and body mass [84-86] |
| 2-alkylamino nicotinamide analogs | Compound 33 | Decreases weight gain [87] |
| Acylurea series of ghrelin modulators | Non-CNS penetrant inverse agonist 22 (AZ-GHS-22) and the CNS penetrant inverse agonist 38 (AZ-GHS-38) | Unknown [89] |
| Synthetic ghrelin receptor inverse agonists | GHSR-IA GHSR-1A2 |
Reduces food intake, glucose levels, blood triglyceride levels and body weight [90] |
| Spiro-azetidino-piperidine | PF-5190457 | Increases glucose-stimulated insulin secretion, inhibits ghrelin-induced secretion of GH, delays gastric emptying, and reduces postprandial glucose [92] |
An acylurea series of ghrelin receptor agonists have been identified by high throughput screening and affinity optimizing through structural modifications. Two chemical compounds; the non-CNS penetrant inverse agonist 22 (AZ-GHS-22) and the CNS penetrant inverse agonist 38 (AZ-GHS-38), have been developed through modification of specific substructure and optimization of physical and chemical properties to convert partial agonist activity to inverse agonist activity [85]. The effects of these compounds in vivo remain unknown.
Novel synthetic inverse ghrelin receptor agonists GHSR-IA and GHSR-IA2 have also been developed. Both compounds are able to reduce food intake in mice. Oral GHSR-IA2 decreases glucose levels, blood triglyceride levels and body weight, with more efficacy [86].
Another small-molecule, PF-5190457, has been identified as a potent and selective GHSR inverse agonist with high oral bioavailability and potential of clinical application [87]. PF-5190457 is from a spiroazetidino-piperidine series. Through the evaluation of the pharmacokinetics, PF-5190457 appears to be a promising molecule. It increases insulin secretion in a glucose-dependent manner in human entire pancreas and dispersed islets. Initial clinical trial in healthy people shows that PF-5190457 is quickly absorbed orally, leading to attenuated secretion of GH, impaired motility and evacuation of stomach, and reduced postprandial glucose [88]. Recently, PF-5190457 has finished preclinical safety experiments and phase 1b of human laboratory study. The study demonstrates that the inverse ghrelin receptor agonist PF-5190457 is safe and tolerable [89].
Overall, several GHSR inverse agonists have been shown to be efficient for weight control in animals (Table 2). Additional researches are necessary to evaluate the long-term use of these compounds in the treatment of obesity and related metabolic diseases.
4. INHIBITION OF GOAT
Octanoylated pentapeptides were identified as the first inhibitor of ghrelin O-acyltransferase (GOAT) [29]. Another specific GOAT inhibitor, GO-CoA-Tat, a bisubstrate analog that antagonizes GOAT, was developed in a subsequent study [90]. In wild-type mice, intraperitoneal injection of GO-CoA-Tat reduces body weight and improves glucose tolerance. This reagent does not improve metabolic conditions in ghrelindeficient mice [90], Circulating acyl ghrelin levels and weight gain are reduced by GO-CoA-Tat in diet-induced obese mice [91]. Among mice treated with GO-CoA-Tat, body weight, triglyceride (TG), total cholesterol (TC) and glucose are reduced. Indicators of liver injury, such as glutamate pyruvate transaminase (GPT) and glutamic oxalacetic transaminase (GOT) are also significantly decreased [92]. Thus, GO-CoA-Tat may hold promise for the intervention of metabolic diseases. However, it is worth noting that this GOAT inhibitor has shown limited efficacy in human beings.
Compound B ((4-chloro-6-[[2-methyl-6-(tntluoromethyl) pyridin-3-yl]methoxy}-1-benzothiophen-3-yl) acetic acid), another GOAT inhibitor discovered by high-throughput screening, decreases gastric acylghrelin of mice after oral administration [93]. These novel discoveries suggest that GOAT is a therapeutic target, and the effects of GOAT inhibition by compound B on body weight warrant further investigation.
5. POTENTIAL PHARMACOLOGICAL TARGET TO REDUCE GHRELIN SECRETION
The discovery that ghrelin activates its receptor GHSR to regulate food intake stimulates the development of over 20 drugs targeting the ghrelin system as interventions for obesity. While ghrelin-GHSR antagonists initially held promise, long term efficacy has been minimal. In a recent study [94, 95], we have demonstrated that long term administration of the ghrelin receptor antagonist, D-Lys-3-GHRP6, leads to significant increases in ghrelin production. An increase in ghrelin production with long term use of receptor antagonists may explain the clinical inefficacy of ghrelin receptor antagonists. Shifting focus from ghrelin receptor antagonism to direct blockade of ghrelin production provides a novel strategy for the treatment of obesity and related diseases. For this goal, we have identified the mechanistic target of rapamycin (mTOR) as a critical molecule linking the production of ghrelin with organism energy levels within gastric X/A like cells [95]. Activation of mTOR signaling in these endocrine cells by deletion of tuberous sclerosis complex 1 (TSC1), an upstream inhibitor of mTOR complex, suppresses the synthesis and secretion of ghrelin, leading to the subsequent amelioration of liver steatosis in high fat diet-induced obese mice [42]. Further, these transgenic mice develop pancreatic fibrosis, which reduces the secretion of insulin [96]. Whether mTOR in gastric X/A like cells may serve as a novel target for the intervention of obesity and metabolic diseases requires further investigation.
Bitter taste receptor (T2R) has been demonstrated to stimulate ghrelin secretion to regulate food intake and gastric emptying [97]. Plasma ghrelin levels increase in an α-gustducin dependent manner in mice by intragastric administration of a mixture of T2R agonists [97]. A recent study has found that the sympathetic drive, controlling ghrelin release in the fundus, is impaired in obese mice. This attenuates the sensitivity of the X/A like cells to bitter and sweet signals in the gastrointestinal tract leading to the reduction of ghrelin level [98]. All these findings suggest that manipulation of endogenous ghrelin by T2R antagonists may provide a potential approach to treat obesity and its related metabolic diseases.
6. SUMMARY AND FUTURE DIRECTIONS
Ghrelin and GFISR play important roles in the modulation of energy homeostasis. A great effort has been made to develop novel strategies targeting the ghrelin-GHSR system and to evaluate the potential clinical application in the intervention of obesity and metabolic diseases. These strategies include neutralization of ghrelin by antibodies, RNA spiegelmers and GFISR-Fc, antagonism of ghrelin receptor by its an-tagonists and inverse agonists, inhibition of ghrelin O-acyltransferase (GOAT) enzyme, as well as potential pharmacological target to decrease ghrelin secretion (Fig. 1). While short-term use of ghrelin or GFISR antagonism holds promise in animal experiments, a long-term application has shown limited efficacy. The mechanism underlying their minimal long-term effect may be attributed to the compensatory increment in the newly synthesized ghrelin associated with these treatments. Defining novel pathways to alter the production of acyl-ghrelin from gastric X/A like cells would shift therapeutic focus to novel gastric targets and ultimately provide a therapeutic benefit for people with metabolic diseases.
Fig. (1).
Summary of ghrelin-based strategies for intervention of metabolic dysfunction. Intervention of ghrelin synthesis, processing, release and activation of its receptor GHSR1a has been developed for the therapy of obesity and its related metabolic dysfunctions.
It is also worth noting that most of the tested compounds have been shown to be effective only in animals and in vitro. So far, only the ghrelin vaccine and PF-5190457 have been tested in humans. Clinical trials of other ghrelin-GHSR based interventions are urgently needed to prove their effectiveness in human beings, especially on reduction of appetite and body gain.
CONCLUSION
The long-term efficacy in weight reduction and therapy of metabolic dysfunctions by blocking the action of ghrelin has been limited, likely because of the negative feedback up-regulation in ghrelin production. Blockade of ghrelin production by direct targeting the gastric X/A like cells may provide an alternative strategy.
FUNDING
This study has been supported by the National Key R&D Program of China (2017YFC0908900); National Natural Science Foundation of China (81730020, 81930015 and 81700516); and National Institutes of Health (R01 DK112755 and R01-DK-110273-01A1).
LIST OF ABBREVIATIONS
- GHSR
Growth Hormone Secretagogue Receptor
- GOAT
Ghrelin O-Acyltransferase
- GRLN-R
Ghrelin Receptor
- NPY
Neuropeptide Y
- AGRP
Agouti Related Peptide
- [D-LYS3]-GHRP-6
[D-Lys3]-Growth Hormone-Releasing Peptide 6
- LEAP-2
Liver Enriched Antimicrobial Peptide 2
- AZ-GHS-22
Non-CNS Penetrant Inverse Agonist 22
- AZ-GHS-38
CNS Penetrant Inverse Agonist 38
- TG
Triglyceride
- TC
Total Cholesterol
- GPT
Glutamate Pyruvate Transaminase
- GOT
Glutamic Oxalacetic Transaminase
- mTOR
Mechanistic Target of Rapamycin
- TSC1
Tuberous Sclerosis Complex 1
- T2R
Bitter Taste Receptor
Footnotes
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or otherwise.
REFERENCES
- [1].Berrington de Gonzalez A; Hartge P; Cerhan JR; Flint AJ; Hannan L; MacInnis RJ; Moore SC; Tobias GS; Anton-Culver H; Freeman LB; Beeson WL; Clipp SL; English DR; Folsom AR; Freedman DM; Giles G; Hakansson N; Henderson KD; Hoffman-Bolton J; Hoppin JA; Koenig KL; Lee IM; Linet MS; Park Y; Pocobelli G; Schatzkin A; Sesso HD; Weiderpass E; Willcox BJ; Wolk A; Zeleniuch-Jacquotte A; Willett WC; Thun MJ Body-mass index and mortality among 1.46 million white adults. N. Engl. J. Med 2010. 363(23), 2211–2219. 10.1056/NEJMoa1000367 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Whitlock G; Lewington S; Sherliker P; Clarke R; Emberson J; Halsey J; Qizilbash N; Collins R; Peto R Prospective Studies Collaboration. Body-mass index and cause-specific mortality in 900,000 adults; collaborative analyses of 57 prospective studies. Lancet, 2009, 373(9669), 1083–1096. 10.1016/S0140-6736(09)60318-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Fontaine KR; Redden DT; Wang C; Westfall AO; Allison DB Years of life lost due to obesity. JAMA, 2003, 289(2), 187–193. 10.1001/jama.289.2.187 [DOI] [PubMed] [Google Scholar]
- [4].Blüher M. Obesity: global epidemiology and pathogenesis. Nat. Rev. Endocrinol, 2019, 15(5), 288–298. 10.1038/s41574-019-0176-8 [DOI] [PubMed] [Google Scholar]
- [5].Kojima M; Hosoda H; Date Y; Nakazato M; Matsuo H; Kangawa K Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature, 1999, 402(6762), 656–660. 10.1038/45230 [DOI] [PubMed] [Google Scholar]
- [6].Davenport AP; Bonner TI; Foord SM; Harmar AJ; Neubig RR; Pin JP; Spedding M; Kojima M; Kangawa K International Union of Pharmacology. LVI. Ghrelin receptor nomenclature, distribution, and function. Pharmacol. Rev, 2005, 57(4), 541–546. 10.1124/pr.57.4.1 [DOI] [PubMed] [Google Scholar]
- [7].Guan XM; Yu H; Palyha OC; McKee KK; Feighner SD; Sirinathsinghji DJ; Smith RG; Van der Ploeg LH; Howard AD Distribution of mRNA encoding the growth hormone secretagogue receptor in brain and peripheral tissues. Brain Res. Mol. Brain Res, 1997, 48(1), 23–29. 10.1016/SO169-328X(97)00071-5 [DOI] [PubMed] [Google Scholar]
- [8].Papotti M; Ghè C; Cassoni P; Catapano F; Deghenghi R; Ghigo E; Muccioli G Growth hormone secretagogue binding sites in peripheral human tissues. J. Clin. Endocrinol. Metab, 2000, 85(10), 3803–3807. 10.1210/jc.85.10.3803 [DOI] [PubMed] [Google Scholar]
- [9].Gnanapavan S; Kola B; Bustin SA; Morris DG; McGee P; Fairclough P; Bhattacharya S; Carpenter R; Grossman AB; Korbonits M The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in humans. J. Clin. Endocrinol. Metab 2002, 87(6), 2988. 10.1210/jcem.87.6.8739 [DOI] [PubMed] [Google Scholar]
- [10].Volante M; Allìa E; Gugliotta P; Funaro A; Broglio F; Deghenghi R; Muccioli G; Ghigo E; Papotti M Expression of ghrelin and of the GH secretagogue receptor by pancreatic islet cells and related endocrine tumors. J. Clin. Endocrinol. Metab 2002, 87(3), 1300–1308. 10.1210/jcem.87.3.8279 [DOI] [PubMed] [Google Scholar]
- [11].Prado CL; Pugh-Bemard AE; Elghazi L; Sosa-Pineda B; Sussel L Ghrelin cells replace insulin-producing beta cells in two mouse models of pancreas development. Proc. Natl Acad. Sci. USA, 2004, 101(9), 2924–2929. 10.1073/pnas.0308604100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Volante M; Allia E; Fulcheri E; Cassoni P; Ghigo E; Muccioli G; Papotti M Ghrelin in fetal thyroid and follicular tumors and cell lines: expression and effects on tumor growth. Am. J. Pathol, 2003, 162(2), 645–654. 10.1016/S0002-9440(10)63858-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Ariyasu H; Takaya K; Tagami T; Ogawa Y; Hosoda K; Akamizu T; Suda M; Koh T; Natsui K; Toyooka S; Shirakami G; Usui T; Shimatsu A; Doi K; Hosoda H; Kojima M; Kangawa K; Nakao K Stomach is a major source of circulating ghrelin, and feeding state determines plasma ghrelin-like immunoreactivity levels in humans. J. Clin. Endocrinol. Metab, 2001, 86(10), 4753–4758. 10.1210/jcem.86.10.7885 [DOI] [PubMed] [Google Scholar]
- [14].Date Y; Nakazato M; Hashiguchi S; Dezaki K; Mondal MS; Hosoda H; Kojima M; Kangawa K; Arima T; Matsuo H; Yada T; Matsukura S Ghrelin is present in pancreatic alpha-cells of humans and rats and stimulates insulin secretion. Diabetes, 2002, 51(1), 124–129. 10.2337/diabetes.51.1.124 [DOI] [PubMed] [Google Scholar]
- [15].Korbonits M; Kojima M; Kangawa K; Grossman AB Presence of ghrelin in normal and adenomatous human pituitary. Endocrine, 2001. 14(1), 101–104. 10.1385/ENDO:14:1:101 [DOI] [PubMed] [Google Scholar]
- [16].Korbonits M; Bustin SA; Kojima M; Jordan S; Adams EF; Lowe DG; Kangawa K; Grossman AB The expression of the growth hormone secretagogue receptor ligand ghrelin in normal and abnormal human pituitary and other neuroendocrine tumors. J. Clin. Endocrinol. Metab, 2001, 86(2), 881–887. 10.1210/jc.86.2.881 [DOI] [PubMed] [Google Scholar]
- [17].Cowley MA; Smith RG; Diano S; Tschöp M; Pronchuk N; Grove KL; Strasburger CJ; Bidlingmaier M; Esterman M; Heiman ML; Garcia-Segura LM; Nillni EA; Mendez P; Low MJ; Sotonyi P; Friedman JM; Liu H; Pinto S; Colmers WF; Cone RD; Horvath TL The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis. Neuron, 2003, 37(4), 649–661. 10.1016/S0896-6273(03)00063-1 [DOI] [PubMed] [Google Scholar]
- [18].Hou Z; Miao Y; Gao L; Pan H; Zhu S Ghrelin-containing neuron in cerebral cortex and hypothalamus linked with the DVC of brainstem in rat. Regul. Pept, 2006, 134(2-3), 126–131. 10.1016/j.regpep.2006.02.005 [DOI] [PubMed] [Google Scholar]
- [19].Mori K; Yoshimoto A; Takaya K; Hosoda K; Ariyasu H; Yahata K; Mukoyama M; Sugawara A; Hosoda H; Kojima M; Kangawa K; Nakao K Kidney produces a novel acylated peptide, ghrelin. FEBS Lett., 2000, 486(3), 213–216. 10.1016/S0014-5793(00)02308-5 [DOI] [PubMed] [Google Scholar]
- [20].Beiras-Fernandez A; Kreth S; Weis F; Ledderose C; Pöttinger T; Dieguez C; Beiras A; Reichart B Altered myocardial expression of ghrelin and its receptor (GHSR-1a) in patients with severe heart failure. Peptides, 2010, 31(12), 2222–2228. 10.1016/j.peptides.2010.08.019 [DOI] [PubMed] [Google Scholar]
- [21].Volante M; Fulcheri E; Allìa E; Cerrato M; Pucci A; Papotti M Ghrelin expression in fetal, infant, and adult human lung. J. Histochem. Cytochem, 2002, 50(8), 1013–1021. 10.1177/002215540205000803 [DOI] [PubMed] [Google Scholar]
- [22].Barreiro ML; Gaytán F; Caminos JE; Pinilla L; Casanueva FF; Aguilar E; Diéguez C; Tena-Sempere M Cellular location and hormonal regulation of ghrelin expression in rat testis. Biol. Reprod, 2002, 67(6), 1768–1776. 10.1095/biolreprod.102.006965 [DOI] [PubMed] [Google Scholar]
- [23].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(3), 909–913. 10.1006/bbrc.2000.4039 [DOI] [PubMed] [Google Scholar]
- [24].Ueberberg B; Unger N; Saeger W; Mann K; Petersenn S Expression of ghrelin and its receptor in human tissues. Horm. Metab. Res, 2009, 41(11), 814–821. 10.1055/s-0029-1233462 [DOI] [PubMed] [Google Scholar]
- [25].Bang AS; Soule SG; Yandle TG; Richards AM; Pemberton CJ Characterisation of proghrelin peptides in mammalian tissue and plasma. J. Endocrinol, 2007, 192(2), 313–323. 10.1677/JOE-06-0021 [DOI] [PubMed] [Google Scholar]
- [26].Yang J; Brown MS; Liang G; Grishin NV; Goldstein JL Identification of the acyltransferase that octanoylates ghrelin, an appetite-stimulating peptide hormone. Cell 2008, 132(3), 387–396. 10.1016/j.cell.2008.01.017 [DOI] [PubMed] [Google Scholar]
- [27].Gutierrez JA; Solenberg PJ; Perkins DR; Willency JA; Knierman MD; Jin Z; Witcher DR; Luo S; Onyia JE; Hale JE Ghrelin octanoylation mediated by an orphan lipid transferase. Proc. Natl. Acad. Sci. USA, 2008, 105(17), 6320–6325. 10.1073/pnas.0800708105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Zhao TJ; Liang G; Li RL; Xie X; Sleeman MW; Murphy AJ; Valenzuela DM; Yancopoulos GD; Goldstein JL; Brown MS Ghrelin O-acyltransferase (GOAT) is essential for growth hormone-mediated survival of calorie-restricted mice. Proc. Natl. Acad. Sci. USA, 2010, 107(16), 7467–7472. 10.1073/pnas.1002271107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Yang J; Zhao TJ; Goldstein JL; Brown MS Inhibition of ghrelin O-acyltransferase (GOAT) by octanoylated pentapeptides. Proc. Natl Acad. Sci. USA, 2008, 105(31), 10750–10755. 10.1073/pnas.0805353105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Solomou S; Korbonits M The role of ghrelin in weight-regulation disorders: implications in clinical practice. Hormones (Athens), 2014, 13(4), 458–475. 10.14310/horm.2002.1551 [DOI] [PubMed] [Google Scholar]
- [31].Schalla MA; Stengel A. Pharmacological modulation of ghrelin to induce weight loss: successes and challenges. Curr. Diab. Rep, 2019, 19(10), 102. 10.1007/s11892-019-1211-9 [DOI] [PubMed] [Google Scholar]
- [32].Weibert E; Stengel A The X/A-like cell revisited - spot-light on the peripheral effects of NUCB2/nesfatin-1 and ghrelin. J. Physiol. Pharmacol, 2017, 68(4), 497–520. [PubMed] [Google Scholar]
- [33].Tang-Christensen M; Vrang N; Ortmann S; Bidlingmaier M; Horvath TL; Tschöp M Central administration of ghrelin and agouti-related protein (83-132) increases food intake and decreases spontaneous locomotor activity in rats. Endocrinology, 2004, 145(10), 4645–4652. 10.1210/en.2004-0529 [DOI] [PubMed] [Google Scholar]
- [34].Wren AM; Small CJ; Ward HL; Murphy KG; Dakin CL; Taheri S; Kennedy AR; Roberts GH; Morgan DG; Ghatei MA; Bloom SR, The novel hypothalamic peptide ghrelin stimulates food intake and growth hormone secretion. Endocrinology, 2000, 141(11), 4325–4328. 10.1210/endo.141.11.7873 [DOI] [PubMed] [Google Scholar]
- [35].Druce MR; Wren AM; Park AJ; Milton JE; Patterson M; Frost G; Ghatei MA; Small C; Bloom SR Ghrelin increases food intake in obese as well as lean subjects. Int. J. Obes, 2005, 29(9), 1130–1136. 10.1038/sj.ijo.0803001 [DOI] [PubMed] [Google Scholar]
- [36].Tschöp M; Smiley DL; Heiman ML Ghrelin induces adiposity in rodents. Nature, 2000, 407(6806), 908–913. 10.1038/35038090 [DOI] [PubMed] [Google Scholar]
- [37].Theander-Carrillo C; Wiedmer P; Cettour-Rose P; Nogueiras R; Perez-Tilve D; Pfluger P; Castaneda TR; Muzzin P; Schürmann A; Szanto I; Tschöp MH; Rohner-Jeanrenaud F Ghrelin action in the brain controls adipocyte metabolism. J. din. Invest, 2006, 116(7), 1983–1993. 10.1172/JCI25811 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Davies JS; Kotokorpi P; Eccles SR; Barnes SK; Tokarczuk PF; Allen SK; Whitworth HS; Guschina IA; Evans BA; Mode A; Zigman JM; Wells T. Ghrelin induces abdominal obesity via GHS-R-dependent lipid retention. Mol. Endocrinol, 2009, 23(6), 914–924. 10.1210/me.2008-0432 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Rodríguez A; Gómez-Ambrosi J; Catalán V; Gil MJ; Becerril S; Sáinz N; Silva C; Salvador J; Colina I; Frühbeck G Acylated and desacyl ghrelin stimulate lipid accumulation in human visceral adipocytes. Int. J. Obes, 2009, 33(5), 541–552. 10.1038/ijo.2009.40 [DOI] [PubMed] [Google Scholar]
- [40].Willesen MG; Kristensen P; Rømer J Co-localization of growth hormone secretagogue receptor and NPY mRNA in the arcuate nucleus of the rat. Neuroendocrinology, 1999, 70(5), 306–316. 10.1159/000054491 [DOI] [PubMed] [Google Scholar]
- [41].Culmsee C; Monnig J; Kemp BE; Mattson MP AMP-activated protein kinase is highly expressed in neurons in the developing rat brain and promotes neuronal survival following glucose deprivation. J. Mol. Neurosci, 2001, 17(1), 45–58. 10.1385/JMN:17:1:45 [DOI] [PubMed] [Google Scholar]
- [42].Li Z; Yu R, Yin W; Qin Y; Ma L; Mulholland M; Zhang W. Mtor signaling in x/a-like cells contributes to lipid homeostasis in mice. Hepatology, 2019, 69(2), 860–875. 10.1002/hep.30229 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Li Z; Xu G; Qin Y; Zhang C; Tang H; Yin Y; Xiang X; Li Y; Zhao J; Mulholland M; Zhang W Ghrelin promotes hepatic lipogenesis by activation of mTOR-PPARγ signaling pathway. Proc. Natl. Acad. Sci. USA, 2014. 111(36), 13163–13168. 10.1073/pnas.1411571111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Zhang W; Chai B; Li JY; Wang H; Mulholland MW Effect of des-acyl ghrelin on adiposity and glucose metabolism. Endocrinology, 2008, 149(9), 4710–4716. 10.1210/en.2008-0263 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Zhang W; Zhao L; Lin TR; Chai B; Fan Y; Gantz I; Mulholland MW Inhibition of adipogenesis by ghrelin. Mol. Biol. Cell, 2004, 15(5), 2484–2491. 10.1091/mbc.e03-09-0657 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Zorrilla EP; Iwasaki S; Moss JA; Chang J; Otsuji J; Inoue K; Meijler MM; Janda KD Vaccination against weight gain. Proc. Natl. Acad. Sci. USA, 2006, 103(35), 13226–13231. 10.1073/pnas.0605376103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Vizcarra JA; Kirby JD; Kim SK; Galyean ML Active immunization against ghrelin decreases weight gain and alters plasma concentrations of growth hormone in growing pigs. Domest. Anim. Endocrinol, 2007, 33(2), 176–189. 10.1016/j.domaniend.2006.05.005 [DOI] [PubMed] [Google Scholar]
- [48].Azegami T; Yuki Y; Sawada S; Mejima M; Ishige K; Akiyoshi K; Itoh H; Kiyono H Nanogel-based nasal ghrelin vaccine prevents obesity. Mucosal Immunol., 2017, 10(5), 1351–1360. 10.1038/mi.2016.137 [DOI] [PubMed] [Google Scholar]
- [49].Biotechnology Cytos. Phase I/IIa clinical trial with obese individuals shows no effect of cyt009-ghrqb on weight loss, Press Release, 2006. (Accessed; July 1, 2011). [Google Scholar]
- [50].Altabas V; Zjačić-Rotkvić V Anti-ghrelin antibodies in appetite suppression: recent advances in obesity pharmacotherapy. Immunotargets Ther., 2015, 4, 123–130. 10.2147/itt.s60398 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Helmling S; Maasch C; Eulberg D; Buchner K; Schröder W; Lange C; Vonhoff S; Wlotzka B; Tschöp MH; Rosewicz S; Klussmann S Inhibition of ghrelin action in vitro and in vivo by an RNA-Spiegelmer. Proc. Natl. Acad. Sci. USA, 2004, 101(36). 13174–13179. 10.1073/pnas.0404175101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Klussmann S; Nolte A; Bald R; Erdmann VA; Fürste JP Mirror-image RNA that binds D-adenosine. Nat. Biotechnol, 1996, 14(9), 1112–1115. 10.1038/nbt0996-1112 [DOI] [PubMed] [Google Scholar]
- [53].Depoortere I. Targeting the ghrelin receptor to regulate food intake. Regul. Pept, 2009, 156(1-3), 13–23. 10.1016/j.regpep.2009.04.002 [DOI] [PubMed] [Google Scholar]
- [54].Becskei C; Bilik KU; Klussmann S; Jarosch F; Lutz TA; Riediger T The anti-ghrelin Spiegelmer NOX-B11-3 blocks ghrelin- but not fasting-induced neuronal activation in the hypothalamic arcuate nucleus. J. Neuroendocrinal, 2008, 20(1), 85–92. 10.1111/j.1365-2826.2007.01619.x [DOI] [PubMed] [Google Scholar]
- [55].Shearman LP; Wang SP; Helmling S; Stribling DS; Mazur P; Ge L; Wang L; Klussmann S; Macintyre DE; Howard AD; Strack AM Ghrelin neutralization by a ribonucleic acid-SPM ameliorates obesity in diet-induced obese mice. Endocrinology, 2006, 147(3), 1517–1526. 10.1210/en.2005-0993 [DOI] [PubMed] [Google Scholar]
- [56].Gagnon J; Zhu L; Anini Y; Wang Q Neutralizing circulating ghrelin by expressing a growth hormone secretagogue receptor-based protein protects against high-fat diet-induced obesity in mice. Gene Ther., 2015, 22(9), 750–757. 10.1038/gt.2015.38 [DOI] [PubMed] [Google Scholar]
- [57].Sun Y; Wang P; Zheng H; Smith RG Ghrelin stimulation of growth hormone release and appetite is mediated through the growth hormone sccretagogue receptor. Proc. Natl Acad. Sci. USA, 2004, 101(13), 4679–4684. 10.1073/pnas.0305930101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Zigman JM; Nakano Y; Coppari R; Balthasar N; Marcus JN; Lee CE; Jones JE; Deysher AE; Waxman AR; White RD; Williams TD; Lachey JL; Seeley RJ; Lowell BB; Elmquist JK Mice lacking ghrelin receptors resist the development of diet-induced obesity. J. Clin. Invest, 2005, 115(12), 3564–3572. 10.1172/JCI26002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Lin L; Saha PK; Ma X; Henshaw IO; Shao L; Chang BH; Buras ED; Tong Q; Chan L; McGuinness OP; Sun Y Ablation of ghrelin receptor reduces adiposity and improves insulin sensitivity during aging by regulating fat metabolism in white and brown adipose tissues. Aging Cell, 2011, 10(6), 996–1010. 10.1111/j.1474-9726.2011.00740.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Veeraragavan K; Sethumadhavan K; Bowers CY Growth hormone-releasing peptide (GHRP) binding to porcine anterior pituitary and hypothalamic membranes. Life Sci., 1992, 50(16), 1149–1155. 10.1016/0024-3205(92)90457-Z [DOI] [PubMed] [Google Scholar]
- [61].Ramirez VT; van Oeffelen WEPA; Torres-Fuentes C; Chruścicka B; Druelle C; Golubeva AV; van de Wouw M; Dinan TG; Cryan JF; Schellekens H Differential functional selectivity and downstream signaling bias of ghrelin reccptor antagonists and inverse agonists. FASEB J., 2019, 33(1), 518–531. 10.1096/fj.201800655R [DOI] [PubMed] [Google Scholar]
- [62].Mosa R; Huang L; Li H; Grist M; LeRoith D; Chen C Long-term treatment with the ghrelin receptor antagonist [d-Lys3]-GHRP-6 does not improve glucose homeostasis in nonobese diabetic MKR mice. Am. J. Phvsiol. Regul. Integr. Comp. Physiol, 2018, 314(1), R71–R83. 10.1152/ajpregu.00157.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Asakawa A; Inui A; Kaga T; Katsuura G; Fujimiya M; Fujino MA; Kasuga M Antagonism of ghrelin receptor reduces food intake and body weight gain in mice. Gut, 2003, 52(7), 947–952. 10.1136/gut.52.7.947 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Beck B; Richy S; Stricker-Krongrad A Feeding response to ghrelin agonist and antagonist in lean and obese Zucker rats. Life Sci., 2004, 76(4), 473–478. 10.1016/j.lfs.2004.09.001 [DOI] [PubMed] [Google Scholar]
- [65].Rasineni K; Kubik JL; Casey CA; Kharbanda KK, Inhibition of ghrelin activity by receptor antagonist [d-lys-3] ghrp-6 attenuates alcohol-induced hepatic steatosis by regulating hepatic lipid metabolism. Biomolecules, 2019, 9(10), E517. 10.3390/biom9100517 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Demange L; Boeglin D; Moulin A; Mousseaux D; Ryan J; Bergé G; Gagne D; Heitz A; Perrissoud D; Locatelli V; Torsello A; Galleyrand JC; Fehrentz JA; Martinez J Synthesis and pharmacological in vitro and in vivo evaluations of novel triazole derivatives as ligands of the ghrelin receptor. 1. J. Med. Chem, 2007, 50(8), 1939–1957. 10.l021/jm070024h [DOI] [PubMed] [Google Scholar]
- [67].Moulin A; Demange L; Bergé G; Gagne D; Ryan J; Mousseaux D; Heitz A; Perrissoud D; Locatelli V; Torsello A; Galleyrand JC; Fehrentz JA; Martinez J. Toward potent ghrelin receptor ligands based on trisubstitutcd 1,2,4-triazole structure. 2. Synthesis and pharmacological in vitro and in vivo evaluations. J. Med. Chem, 2007, 50(23), 5790–5806. 10.1021/jm0704550 [DOI] [PubMed] [Google Scholar]
- [68].Gomez JL; Ryabinin AE The effects of ghrelin antagonists [D-Lys(3)]-GHRP-6 or JMV2959 on ethanol, water and food intake in C57BL/6J mice. Alcohol. Clin. Exp. Res, 2014, 38(9), 2436–2444. 10.1111/acer.12499 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Salomé N; Haage D; Perrissoud D; Moulin A; Demange L; Egecioglu E; Fehrentz JA; Martinez J; Dickson SL Anorexigenic and electrophysiological actions of novel ghrelin receptor (GHS-R1A) antagonists in rats. Eur. J. Pharmacol, 2009, 612(1-3), 167–173. 10.1016/j.ejphar.2009.03.066 [DOI] [PubMed] [Google Scholar]
- [70].Halem HA; Taylor JE; Dong JZ; Shen Y; Datta R; Abizaid A; Diano S; Horvath TL; Culler MD A novel growth hormone secretagogue-1 a receptor antagonist that blocks ghrelin-induced growth hormone secretion but induces increased body weight gain. Neuroendocrinology, 2005, 81(5), 339–349. 10.1159/000088796 [DOI] [PubMed] [Google Scholar]
- [71].Rudolph J; Esler WP; O’connor S; Coish PD; Wickens PL; Brands M; Bierer DE; Bloomquist BT; Bondar G; Chen L; Chuang CY; Claus TH; Fathi Z; Fu W; Khire UR; Kristie JA; Liu XG; Lowe DB; McClure AC; Michels M; Ortiz AA; Ramsden PD; Schoenleber RW; Shelekhin TE; Vakalopoulos A; Tang W; Wang L; Yi L; Gardell SJ; Livingston JN; Sweet LJ; Bullock WH Quinazolinone derivatives as orally available ghrelin receptor antagonists for the treatment of diabetes and obesity. J. Med. Chem, 2007, 50(21), 5202–5216. 10.102l/jm070071 [DOI] [PubMed] [Google Scholar]
- [72].Esler WP; Rudolph J; Claus TH; Tang W; Barucci N; Brown SE; Bullock W; Daly M; Decarr L; Li Y; Milardo L; Molstad D; Zhu J; Gardell SJ; Livingston JN; Sweet LJ Small-molecule ghrelin receptor antagonists improve glucose tolerance, suppress appetite, and promote weight loss. Endocrinology, 2007, 145(11). 5175–5185. 10.1210/en.2007-0239 [DOI] [PubMed] [Google Scholar]
- [73].Longo KA; Govek EK; Nolan A; McDonagh T; Charoenthongtrakul S; Giuliana DJ; Morgan K; Hixon J; Zhou C; Kelder B; Kopchick JJ; Saunders JO; Navia MA; Curtis R; DiStefano PS; Geddes BJ Pharmacologic inhibition of ghrelin receptor signaling is insulin sparing and promotes insulin sensitivity. J. Pharmacol. Exp. Ther, 2011, 339(1), 115–124. 10.1124/jpet.111.183764 [DOI] [PubMed] [Google Scholar]
- [74].Sabbatini FM; Di Fabio R; Corsi M; Cavanni P; Bromidge SM; St-Denis Y; D’Adamo L; Contini S; Rinaldi M; Guery S; Savoia C; Mundi C; Perini B; Carpenter AJ; Dal Forno G; Faggioni F; Tessari M; Pavone F; Di Francesco C; Buson A; Mattioli M; Perdona E; Melotto S Discovery process and characterization of novel carbohydrazide derivatives as potent and selective GHSR1a antagonists. Chem. Med. Chem, 2010, 5(9), 1450–1455. 10.1002/cmdc.201000185 [DOI] [PubMed] [Google Scholar]
- [75].Costantini VJ; Vicentini E; Sabbatini FM; Valerio E; Lepore S; Tessari M; Sartori M; Michielin F; Melotto S; Bifone A; Pich EM; Corsi M GSK 1614343, a novel ghrelin receptor antagonist, produces an unexpected increase of food intake and body weight in rodents and dogs. Neuroendocrinology, 2011, 94(2), 158–168. 10.1159/000328968 [DOI] [PubMed] [Google Scholar]
- [76].Hoist B; Cygankiewicz A; Jensen TH; Ankersen M; Schwartz TW High constitutive signaling of the ghrelin receptor-identification of a potent inverse agonist. Mol. Endocrinol, 2003, 17(11), 2201–2210. 10.1210/me.2003-0069 [DOI] [PubMed] [Google Scholar]
- [77].Holliday ND; Holst B; Rodionova EA; Schwartz TW; Cox HM Importance of constitutive activity and arrestin-independent mechanisms for intracellular trafficking of the ghrelin receptor. Mol. Endocrinol, 2007, 21(12), 3100–3112. 10.1210/me.2007-0254 [DOI] [PubMed] [Google Scholar]
- [78].Els S; Beck-Sickinger AG; Chollet C. Ghrelin receptor: high constitutive activity and methods for developing inverse agonists. Methods Enzymol., 2010, 485, 103–121. 10.1016/B978-0-12-381296-4.00006-3 [DOI] [PubMed] [Google Scholar]
- [79].Hoist B; Schwartz TW Constitutive ghrelin receptor activity as a signaling set-point in appetite regulation. Trends Pharmacol. Sci, 2004, 25(3), 113–117. 10.1016/j.tips.2004.01.010 [DOI] [PubMed] [Google Scholar]
- [80].Ge X; Yang H; Bednarek MA; Galon-Tilleman H; Chen P; Chen M; Lichtman JS; Wang Y; Dalmas O; Yin Y; Tian H; Jermutus L; Grimsby J; Rondinone CM; Konkar A; Kaplan DD Leap2 is an endogenous antagonist of the ghrelin receptor. Cell Metab., 2018, 27(2), 461–469.e6. 10.1016/j.cmet.2017.10.016 [DOI] [PubMed] [Google Scholar]
- [81].M’Kadmi C; Cabral A; Barrile F; Giribaldi J; Cantel S; Damian M; Mary S; Denoyelle S; Dutertre S; Péraldi-Roux S; Neasta J; Oiry C; Banères JL; Marie J; Perello M; Fehrentz JA N-terminal liver-expressed antimicrobial peptide 2 (leap2) region exhibits inverse agonist activity toward the ghrelin rcceptor. J. Med. Chem 2019, 62(2), 965–973. 10.1021/acs.jmedchem.8b01644 [DOI] [PubMed] [Google Scholar]
- [82].Mani BK; Puzziferri N; He Z; Rodriguez JA; Osborne-Lawrence S; Metzger NP; Chhina N; Gaylinn C; Thorner MO; Thomas EL; Bell JD; Williams KW; Goldstone AP; Zigman JM LEAP2 changes with body mass and food intake in humans and mice. J. Clin. Invest, 2019, 129(9), 3909–3923. 10.1172/JCI125332 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Takahashi B; Funami H; Iwaki T; Maruoka H; Shibata M; Koyama M; Nagahira A; Kamiide Y; Kanki S; Igawa Y; Muto T Orally active ghrelin receptor inverse agonists and their actions on a rat obesity model. Bioorg. Med. Chem, 2015, 23(15), 4792–4803. 10.10l6/j.bmc.2015.05.047 [DOI] [PubMed] [Google Scholar]
- [84].Takahashi B; Funami H; Iwaki T; Maruoka H; Nagahira A; Koyama M; Kamiide Y; Matsuo T; Muto T; Annoura H 2-Aminoalkyl nicotinamide derivatives as pure inverse agonists of the ghrelin receptor. Bioorg. Med. Chem. Lett, 2015, 25(13), 2707–2712. 10.1016/j.bmcl.2015.04.040 [DOI] [PubMed] [Google Scholar]
- [85].McCoull W; Barton P; Brown AJ; Bowker SS; Cameron J; Clarke DS; Davies RD; Dossetter AG; Ertan A; Fenwick M; Green C; Holmes JL; Martin N; Masters D; Moore JE; Newcombe NJ; Newton C; Pointon H; Robb GR; Sheldon C; Stokes S; Morgan D Identification, optimization, and pharmacology of acylurea GHS-R1a inverse agonists. J. Med. Chem, 2014, 57(14), 6128–6140. 10.1021/jm50061on [DOI] [PubMed] [Google Scholar]
- [86].Abegg K; Bemasconi L; Hutter M; Whiting L; Pietra C; Giuliano C; Lutz TA; Riediger T Ghrelin receptor inverse agonists as a novel therapeutic approach against obesity-related metabolic disease. Diabetes Obes. Metab, 2017, 19(12), 1740–1750. 10.1111/dom.13020 [DOI] [PubMed] [Google Scholar]
- [87].Bhattacharya SK; Andrews K; Beveridge R; Cameron KO; Chen C; Dunn M; Fernando D; Gao H; Hepworth D; Jackson VM; Khot V; Kong J; Kosa RH; Lapham K; Loria PM; Londregan AT; McClure KF; Orr ST; Patel J; Rose C; Saenz J; Stock LA; Storer G; VanVolkenburg M; Vrieze D; Wang G; Xiao J; Zhang Y Discovery of pf-5190457, a potent, selective Activation of mTORC1 signaling in gastric X/A-like cells induces spontaneous pancreatic fibrosis and derangement of glucose metabolism by reducing ghrelin production and orally bioavailable ghrelin receptor inverse agonist clinical candidate. ACS Med. Chem. Lett, 2014, 5(5), 474–479. 10.1021/m1400473x [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Denney WS; Sonnenberg GE; Carvajal-Gonzalez S; Tuthill T; Jackson VM Pharmacokinetics and pharmacodynamics of PF-05190457: The first oral ghrelin receptor inverse agonist to be profiled in healthy subjects. Br. J. Clin. Pharmacol, 2017, 83(2), 326–338. 10.1111/bcp.13127 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Lee MR; Tapocik JD; Ghareeb M; Schwandt ML; Dias AA; Le AN; Cobbina E; Farinelli LA; Bouhlal S; Farokhnia M; Heilig M; Akhlaghi F; Leggio L The novel ghrelin receptor inverse agonist PF-5190457 administered with alcohol: preclinical safety experiments and a phase lb human laboratory study. Mol. Psychiatry, 2020, 25(2), 461–475. 10.1038/s41380-0l8-0064-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Barnett BP; Hwang Y; Taylor MS; Kirchner H; Pfluger PT; Bernard V; Lin YY; Bowers EM; Mukherjee C; Song WJ; Longo PA; Leahy DJ; Hussain MA; Tschöp MH; Boeke JD; Cole PA Glucose and weight control in mice with a designed ghrelin O-acyltransferase inhibitor. Science, 2010, 330(6011), 1689–1692. 10.1126/science.1196154 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Teubner BJ; Bartness TJ Anti-ghrelin Spiegelmer inhibits exogenous ghrelin-induced increases in food intake, hoarding, and neural activation, but not food deprivation-induced increases. Am. J. Physiol. Regul. Integr. Comp. Physiol, 2013, 305(4), R323–R333. 10.1152/ajpregu.00097.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Zhang S; Mao Y; Fan X Inhibition of ghrelin o-acyltransferase attenuated lipotoxicity by inducing auto-phagy via AMPK-mTOR pathway. Drug Des. Devel. Ther, 2018, 12, 873–885. 10.2147/DDDT.S158985 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Yoneyama-Hirozane M; Deguchi K; Hirakawa T; Ishii T; Odani T; Matsui J; Nakano Y; Imahashi K; Takakura N; Chisaki I; Takekawa S; Sakamoto J Identification and characterization of a new series of ghrelin o-acyl transferase inhibitors. SLAS Discov., 2018, 23(2), 154–163. 10.1177/2472555217727097 [DOI] [PubMed] [Google Scholar]
- [94].Xu G; Wang Z; Li Y; Li Z; Tang H; Zhao J; Xiang X; Ding L; Ma L; Yuan F; Fei J; Wang W; Wang N; Guan Y; Tang C; Mulholland M; Zhang W Ghrelin contributes to derangements of glucose metabolism induced by rapamycin in mice. Diahetologia, 2012, 55(6), 1813–1823. 10.1007/s00125-012-2509-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Xu G; Li Y; An W; Li S; Guan Y; Wang N; Tang C; Wang X; Zhu Y; Li X; Mulholland MW; Zhang W Gastric mammalian target of rapamycin signaling regulates ghrelin production and food intake. Endocrinology, 2009, 150(8), 3637–3644. 10.1210/en.2009-0372 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Yu R; Li Z; Liu S; Huwatibieke B; Li Y; Yin Y; Zhang W Activation of mTORC1 signaling in gastric X/A-like cells induces spontaneous pancreatic fibrosis and derangement of glucose metabolism by reducing ghrelin production. EBiomedicine, 2018, 36, 304–315. 10.1016/j.ebiom.2018.09.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Janssen S; Laermans J; Verhulst PJ; Thijs T; Tack J; Depoortere I Bitter taste receptors and α-gustducin regulate the secretion of ghrelin with functional effects on food intake and gastric emptying. Proc. Natl. Acad. Sci. USA, 2011, 108(5), 2094–2099. 10.1073/pnas.1011508108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Wang Q; Liszt KI; Deloose E; Canovai E; Thijs T; Farré R; Ceulemans LJ; Lannoo M; Tack J; Depoortere I Obesity alters adrenergic and chemosensory signaling pathways that regulate ghrelin secretion in the human gut. FASEB J., 2019, 33(4). 4907–4920. 10.1096/fj.201801661RR [DOI] [PubMed] [Google Scholar]

