Você está na página 1de 21

This article was published in an Elsevier journal.

The attached copy


is furnished to the author for non-commercial research and
education use, including for instruction at the author’s institution,
sharing with colleagues and providing to institution administration.
Other uses, including reproduction and distribution, or selling or
licensing copies, or posting to personal, institutional or third party
websites are prohibited.
In most cases authors are permitted to post their version of the
article (e.g. in Word or Tex form) to their personal website or
institutional repository. Authors requiring further information
regarding Elsevier’s archiving and manuscript policies are
encouraged to visit:

http://www.elsevier.com/copyright
Author's personal copy

Available online at www.sciencedirect.com

Comparative Biochemistry and Physiology, Part A 149 (2008) 109 – 128


www.elsevier.com/locate/cbpa

Review
Ghrelin: A multifunctional hormone in non-mammalian vertebrates
Hiroyuki Kaiya a,⁎, Mikiya Miyazato a , Kenji Kangawa a , Richard E. Peter b , Suraj Unniappan c,⁎
a
Department of Biochemistry, National Cardiovascular Center Research Institute, Osaka, Japan
b
Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
c
Department of Biology, York University, Toronto, Ontario, Canada
Received 31 October 2007; received in revised form 3 December 2007; accepted 4 December 2007
Available online 14 December 2007

Abstract

In mammals, ghrelin is a non-amidated peptide hormone, existing in both acylated and non-acylated forms, produced mainly from the X/A or
ghrelin cells present in the mucosal layer of the stomach. Ghrelin is a natural ligand of the growth hormone (GH) secretagogue-receptor (GHS-R),
and functions primarily as a GH-releasing hormone and an orexigen, as well as having several other biological actions. Among non-mammalian
vertebrates, amino acid sequence of ghrelin has been reported in two species of cartilaginous fish, seven species of teleosts, two species of
amphibians, one species of reptile and six species of birds. The structure and functions of ghrelin are highly conserved among vertebrates. This
review presents a concise overview of ghrelin biology in non-mammalian vertebrates.
© 2007 Elsevier Inc. All rights reserved.

Keywords: Amphibians; Birds; Brain; Fish; Food intake; Ghrelin; Gut; Hormones; Peptides; Reptiles

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
2. From growth hormone secretagogues to ghrelin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
3. Ghrelin — from gene to peptide . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111
3.1. Ghrelin gene. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111
3.2. Ghrelin precursor, mature peptide and obestatin. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111
3.2.1. Ghrelin precursor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111
3.2.2. Mature peptide . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111
3.2.3. Acylation at the third amino acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
3.2.4. Active core of ghrelin molecule . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
3.2.5. Des-acyl ghrelin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
3.2.6. Obestatin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
4. Tissue distribution and localization of ghrelin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
4.1. Stomach . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
4.2. Brain. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116
4.3. Ghrelin levels in plasma or tissues . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116
5. Ghrelin receptor (GHS-R) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116

⁎ Corresponding authors. H. Kaiya is to be contacted at Department of Biochemistry, National Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita,
Osaka 565-8565, Japan. Tel.: +81 6 6833 5012x2458; fax: +81 6 6835 5402. S. Unniappan, Department of Biology, York University, 4700 Keele Street, Toronto,
Ontario, Canada M3J 1P3.
E-mail addresses: kaiya@ri.ncvc.go.jp (H. Kaiya), suraju@yorku.ca (S. Unniappan).

1095-6433/$ - see front matter © 2007 Elsevier Inc. All rights reserved.
doi:10.1016/j.cbpa.2007.12.004
Author's personal copy

110 H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128

6. Biological actions of ghrelin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119


6.1. Regulation of hormone release and synthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
6.1.1. Fish . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
6.1.2. Amphibians . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120
6.1.3. Birds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
6.1.4. Reptiles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
6.2. Appetite regulatory effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
6.2.1. Fish . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
6.2.2. Amphibians . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
6.2.3. Birds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
6.2.4. Reptiles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
6.3. Reproduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
6.3.1. Fish . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
6.3.2. Birds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
6.4. Gastrointestinal motility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
6.5. Regulation of water intake . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
6.6. Immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
6.7. Growth effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
6.8. Behavior . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
7. Perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 124
8. Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 124
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 124
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 124

1. Introduction in vitro in chicken (Bowers et al., 1984; Geris et al., 1998;


2001). Geris et al. (2001) also reported that GHS stimulates the
Gut hormones regulate a myriad of physiological functions release of an endogenous GH-releasing hormone, thyrotropin-
including appetite and energy balance in vertebrates (Badman releasing hormone (TRH) from chicken brain. These results
and Flier, 2005). Ghrelin, the only known appetite-stimulatory indicate a direct effect of GHS on the hypothalamus as well as
(orexigenic) gut hormone, is an acylated peptide that was its role in the pituitary to regulate GH release in chicken. The
recently added to the growing list of gut regulatory peptides results observed in fish and birds are in agreement with the
(Kojima et al., 1999). Ghrelin is involved in various physiolo- hypophysiotropic functions of GHS in mammals (Tannenbaum
gical functions other than GH release and appetite in mammals and Bowers, 2001). Furthermore, GHS participates in the
(Korbonits et al., 2004; van der Lely et al., 2004; Kojima and regulation of food intake. Saito et al. (2002a) reported that
Kangawa, 2005; Hosoda et al., 2006). There are several lines of intracerebroventricular (ICV) injections of GHRP-6 reduce
evidence for the existence, wide distribution and multiple food intake in neonatal chicks. Effects of GHS, although studied
biological actions of ghrelin in non-mammalian vertebrates, as only in a limited number of non-mammals, suggest the potential
well as in mammals. This review focuses on a comparative presence of an endogenous GHS and its receptor in non-
discussion of ghrelin in fish, amphibians, reptiles and birds with mammalian vertebrates.
mammals in terms of its structure, distribution and functions. In 1996, GHS were found to bind to an orphan receptor
discovered from human and swine. Binding of GHS elicited an
2. From growth hormone secretagogues to ghrelin increase in intracellular calcium in cells stably expressing the
orphan receptor. This finding led to the naming of the receptor
Growth hormone secretagogues (GHS) are synthetic com- as the growth hormone secretagogue-receptor 1a (GHS-R1a)
pounds having growth hormone (GH)-releasing actions (Tan- (Howard et al., 1996). An inactive alternative splice variant of
nenbaum and Bowers, 2001). Some of these GHS also have the GHS-R subtype, termed GHS-R1b, has also been found,
appetite-stimulatory effects in mammals (Laferrere et al., 2005). although the function is unknown (Howard et al., 1996). Many
GHS have been used in several studies on the regulation of GH groups explored the endogenous ligand for the GHS-R1a,
release or feeding in non-mammalian vertebrates. Intraperito- Kojima and colleagues succeeded to de-orphanize the GHS-R1a
neal (IP) injections of a GHS, KP-102, increased circulating GH by discovering its endogenous ligand from rat and human
levels in Nile tilapia, Oreochromis niloticus (Shepherd et al., stomach (Kojima et al., 1999). The identified peptide is 28-
2000). However, other GHS, GHRP-6 and hexarelin, were amino acid long and has a unique fatty acid modification at the
found ineffective in stimulating GH release in vitro and in vivo N-terminal third amino acid, serine (Ser-3) (Fig. 1). The peptide
in a grass carp, Ctenopharyngodon idellus (Xiao et al., 2002), has been named “ghrelin”, a term derived from the Proto-Indo-
suggesting a species-specific action of GHS. Synthetic GHS, L- European word “ghre” meaning “grow” and the name can also
692,429 and L-163,255 stimulate the release of GH in vivo and indicate the abbreviation for GH, followed by “relin” a suffix
Author's personal copy

H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128 111

comprised of five exons and four introns as seen in rats, mice


and humans. The ghrelin gene organization of a reptile, the red-
eared slider turtle has been identified and it is composed of five
exons and four introns (H. Kaiya, unpublished data). The
ghrelin gene organization in amphibians is currently unknown.

3.2. Ghrelin precursor, mature peptide and obestatin

3.2.1. Ghrelin precursor


Fig. 2 shows a comparison of prepro-ghrelin sequences from
various non-mammalian vertebrates. Ghrelin precursor is
composed of a signal peptide, ghrelin mature peptide and the
C-terminal peptide (C-peptide). Length of ghrelin precursor
protein varies from species to species and it is 103-amino acid
long in goldfish, while only contains 124 amino acids in turtle.
Fig. 1. Primary structure of ghrelin in human and eel. Human ghrelin is a non- Mature peptide is processed from the precursor protein. A
amidated 28-amino acid peptide. Eel ghrelin is a 21-amino acid peptide and has dibasic processing sequence (arginine (Arg)-Arg) is present at
an amide structure at the C-terminal end.
the C-terminal end of mature ghrelin with some exceptions:
turkey has a proline (Pro)-Arg sequence, and the Perciformes,
meaning releasing substance. The fascinating story of ghrelin including tilapia, sea bass, seabream and grouper have a single
discovery has been described in some reviews (Kojima et al., Arg (Fig. 2). In addition to these, all teleost ghrelin has an amide
2001a; 2001b; Kojima and Kangawa, 2005). The International structure at the C-terminus of the mature peptide (Fig. 1). In
Union of Pharmacology Committee on Receptor Nomenclature contrast, tetrapod ghrelin including amphibians, reptiles and
and Drug Classification has accepted “GHS-R1a” as the name mammals do not have the structure (Fig. 1). A glycine residue
for the ghrelin receptor and “GRLN” as the abbreviation for existing at the C-terminus of the mature peptide after processing
ghrelin (Davenport et al., 2005). contributes to the formation of the amide structure. Presence of
non-amidated, glycine (Gly)-extended ghrelin has also been
3. Ghrelin — from gene to peptide reported in rainbow trout (Kaiya et al., 2003c). Although the
mature peptide is processed at the dibasic Arg residues, recently
Since ghrelin was discovered in rats and humans in 1999, the identified shark ghrelin has no amide structure at the C-terminus
existence of ghrelin has been demonstrated in different non- of the mature peptide. This is due to the lack of a Gly residue at
mammalian vertebrate species from cartilaginous fish to birds the C-terminus of the mature peptide. Goldfish (Unniappan et
(Table 1). al., 2002) ghrelin mRNA has two putative processing sites and
amidation signals after the 12th and 19th amino acids,
3.1. Ghrelin gene suggesting a possible presence of two putative amidated
peptides in this species. However, the endogenous forms of
It has been reported that human ghrelin gene consists of four ghrelin have not been purified.
exons and three introns (Wajnrajch et al., 2000). On the other
hand, in rats and mice, an additional 19-bp non-coding short 3.2.2. Mature peptide
exon was present (Tanaka et al., 2001) and rodent ghrelin gene Mature ghrelins identified in mammals so far are known to
results in five exons and four introns. Recently, the presence of be a 27- or 28-amino acid peptide (Kojima and Kangawa,
an additional 20-bp non-coding exon in human ghrelin gene 2005). Ghrelin mature peptide is identified in goldfish
was discovered after detailed sequence analyses. Based on this (Unniappan et al., 2002), eel (Kaiya et al., 2003a), Mozambique
new data, the human ghrelin gene is composed of five exons and tilapia (Kaiya et al., 2003b), Nile tilapia (Parhar et al., 2003),
four introns (Wei et al., 2005). rainbow trout (Kaiya et al., 2003c), channel catfish (Kaiya et al.,
In non-mammalian vertebrates, sequence of the ghrelin gene 2005), seabream (Yeung et al., 2006), European sea bass
and exon:intron organization have been reported in goldfish (Terova et al., 2008) and zebrafish (Napolitano, F., 2005
(Unniappan et al., 2002), Nile tilapia (Parhar et al., 2003), (CAJ20254), referred in Olsson et al., 2008), and has different
rainbow trout (Kaiya et al., 2003c), seabream (Yeung et al., lengths from 12- to 23-amino acids (Fig. 2). Recently, ghrelin
2006), chicken (Nie et al., 2004; Richards et al., 2006) and was identified from two species of sharks and shark ghrelin, the
turkey (Richards et al., 2006) (Table 1). The ghrelin gene in first known ghrelin from a cartilaginous fish consists of 26-
goldfish, tilapia and seabream has four exons and three introns. amino acids (Kawakoshi et al., 2007). Ghrelin in two species of
The sequence identity of the short, non-coding 1st exon found frog, Rana catesbeiana (Kaiya et al., 2001) and Rana esculenta
in humans, rats and mice ghrelin gene is currently not evident in (Napolitano, F., 2005 (AM055941)) are 28-amino acid long.
goldfish, tilapia and seabream. On the other hand, the ghrelin Turtle ghrelin, the only ghrelin reported so far from reptiles, has
gene of rainbow trout (Kaiya et al., 2003c), chicken (Nie et al., 25 amino acids (Kaiya et al., 2004). Ghrelin has been identified
2004; Richards et al., 2006) and turkey (Richards et al., 2006) is in six species of birds: chicken (Kaiya et al., 2002; Nie et al.,
Author's personal copy

112
Table 1
Information of ghrelin in non-mammalian vertebrates
Scientific name Ghrelin cDNA accession# Reference Remarks Ghrelin gene accession# Reference GHS-R1a cDNA accession# Reference
Carcharhinus AB254129 Kawakoshi et al. (2007)
melanopterus

H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128
Sphyrna lewini AB254128 Kawakoshi et al. (2007) AB254130 Kawakoshi et al. (2007)
Ictalurus punctatus AB196449 Kaiya et al. (2005) Prepro-ghrelin-1
AB196450 Kaiya et al. (2005) Prepro-ghrelin-2
Anguilla japonica AB062427 Kaiya et al. (2003a)
Carassius auratus AF454389 Unniappan et al. (2002) AF454390 Unniappan et al. (2002)
Daino rerio AM055940 Napolitano et al. (2005)⁎ XM_001340372
NM_001083872 XM_001335981 Olsson et al. (2008)
Oreochromis AB077764 Kaiya et al. (2003b) AB361053 Kaiya et al. (2007)⁎
mossambiqus
Oreochromis niloticus AB104859 Parhar et al. (2003) AB104860 Parhar et al. (2003)
Oreochromis urolepis EF371464 Gao et al. (2007)⁎
Epinephelus coioides DQ343147 Lu et al. (2005)⁎
Dicentrarchus labrax DQ665912 Terova et al. (2008)
Acanthopagrus schlegeli AY643808 Yeung et al. (2006) AY643809 Yeung et al. (2006) AY151040 Chan and Cheng (2004)
Oncorhynchus mykiss AB096919 Kaiya et al. (2003c) Prepro-ghrelin-1 AB100839 Kaiya et al. (2003c) AB362479 Kaiya et al. (2007)⁎
AB101443 Kaiya et al. (2003c) Prepro-ghrelin-2
Spheroides nephelus AF082209 Palyha et al. (2000)
Rana catesbeiana AB058510 Kaiya et al. (2001)
Rana esculenta AM055941 Napolitano et al. (2005)⁎
Trachemis scripta AB161457 Kaiya et al. (2002) Prepro-ghrelin-1
elegans
AB161458 Prepro-ghrelin-2

Gallus gallus AB075215 (broiler) Kaiya et al. (2002) AY303688 Nie et al. (2004) AB095995; AJ309542 Tanaka et al. (2003);
Geelissen et al. (2003)
AY299454 (Leghorn) Richards et al. (2006) AB158617 Tanaka et al. (2004)⁎
Anas platyrhynchos AY338466 Yuan et al. (2007)
Dromaius AY338467 Yuan et al. (2007)
novaehollandiae
Anser sp. AY338465 Yuan et al. (2007)
Meleagris gallapavo AY333783 Richards et al. (2006) AY497549 Richards et al. (2006)
Coturnix japonica AB244056 Yoshimura et al. (2006)
Homo sapiens AB029434 Kojima et al. (1999) AF296558 Wajnrajch et al. (2000)
Note: ⁎ These papers have not been published. Database only.
Author's personal copy

H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128 113

2004; Richards et al., 2006), turkey (Richards et al., 2006), emu 3.2.5. Des-acyl ghrelin
(Yuan et al., 2007), goose (Yuan et al., 2007), duck (Yuan et al., Hosoda et al. (2000a) reported presence of two forms of
2007) and Japanese quail (Yoshimura et al., 2005; Kaiya et al., ghrelin in gastrointestinal tissues of rats, ghrelin having
2007b). Ghrelin in birds is a 26-amino acid peptide. In rats, a acylation at Ser-3 and des-acyl ghrelin lacking the acyl
27-amino acid des-Gln14-ghrelin, generated by the alternative modification at Ser-3. Des-acyl ghrelin is also present in
splicing of the ghrelin gene, has been identified (Hosoda circulation and growing evidence suggests des-acyl ghrelin has
et al., 2000b). This peptide acts as a full agonist of GHS- some biological actions (Asakawa et al., 2005; Chen et al.,
R1a. A similar splice variant, des-V13R14Q15-ghrelin was 2005; Toshinai et al., 2006; Sato et al., 2006). In non-
reported in rainbow trout (Kaiya et al., 2003c). No nucleotide mammalian vertebrates, Kaiya et al. (2006a) revealed presence
sequences that cause similar alternative splicing in the region of des-acyl ghrelin-like substance in the stomach and plasma of
encoding the mature peptide was found in other non-mammalian bullfrog using radioimmunoassay (RIA) combined with high-
vertebrates. performance liquid chromatography (HPLC). Furthermore, des-
acyl ghrelin can inhibit ghrelin-induced food intake in goldfish,
3.2.3. Acylation at the third amino acid although des-acyl ghrelin alone has no effect (Matsuda et al.,
A unique feature of ghrelin is the modification of Ser-3 with 2006a). These results suggest synthesis and bioactivity of des-
fatty acid; rat and human ghrelin is modified by octanoic acid acyl ghrelin in non-mammalian vertebrates.
(Kojima et al., 1999; Kojima and Kangawa, 2005). In non-
mammalian vertebrates, almost of all species from cartilaginous 3.2.6. Obestatin
fish to birds have Ser-3, and the amino acid has been modified Zhang et al. (2005) discovered a peptide that antagonizes the
mainly by octanoic acid as in mammals. Only frog ghrelin has a stimulatory effect of ghrelin on food intake and gut motility
substitution of the amino acid from Ser to threonine (Thr), and though an orphan GPCR, GPR39. The peptide was found in the
the Thr residue has been modified by octanoic acid (Kaiya et al., C-terminal region of rat prepro-ghrelin. This peptide was named
2001). In fact both Ser-3 and Thr-3 can be modified by various obestatin, a contraction of obese from Latin “obedere”, meaning
kinds of middle-chain fatty acids, i.e., decanoic acid (C10:0) and to devour, and “statin” denoting suppression. Obestatin is a 23-
decenoic acid (C10:1) are attached in human ghrelin (Hosoda amino acid amidated peptide, and putative obestatin 13–23 has
et al., 2003). Similar variations of Ser-3 and Thr-3 modification also been present. However, several groups who followed up
have been reported in all non-mammalian vertebrates where the these studies were unable to reproduce the original findings on
peptide sequence has been identified (Kaiya et al., 2001, 2003c, obestatin binding and activation of GPR39 in vitro (Lauwers et
2004, 2005, 2007a; Kawakoshi et al., 2007). In Mozambique al., 2006; Holst et al., 2007; Chartrel et al., 2007). Zhang et al.
tilapia, ghrelin modified with decanoic acid is the major form of (2007) confirmed impurities in the initial batch of obestatin they
the mature ghrelin, and octanoylated ghrelin was not detected in used, and indicated that they could not reproduce their original
the stomach extracts (Kaiya et al., 2003b). finding on obestatin binding and activation of GPR39. Zhang et
The exact mechanisms governing acyl modification of al. (2007) claims reproducibility of inhibitory effects of
ghrelin remains to be fully elucidated. It is likely that ingested obestatin on food intake. However, several other researchers
or absorbed substrates contribute to acylation of ghrelin (Nishi were unable to find any inhibitory effect for obestatin on food
et al., 2005). It has been reported that oral (OR) or IP intake and body weight gain (Bresciani et al., 2006; Seoane et
administration of octanoic acid promotes production of acylated al., 2006; Sibilia et al., 2006; Zizzari et al., 2007). Nothing is
ghrelin in chicken proventriculus (Yamato et al., 2005). reported about the isolation of obestatin in non-mammalian
vertebrates, although possible existence has been proposed in
3.2.4. Active core of ghrelin molecule seabream (Yeung et al., 2006). However, the obestatin region in
In mammals, the N-terminal four amino acids of ghrelin non-mammalian prepro-ghrelin sequences is highly divergent
including the acyl modification is essential for receptor binding compared to mammalian obestatins (Fig. 2). It was proposed to
and this structure is named as the “active core” of the peptide obestatin as ghrelin-associated peptide (Gourcerol and Taché,
(Bednarek et al., 2000; Matsumoto et al., 2001). Comparison of 2007; Gourcerol et al., 2007). Given that the “obestatin”
ghrelin sequences among non-mammalian vertebrates displays sequence does not exist in non-mammalian vertebrates and its
high sequence homology within the seven amino acids at the N- synthesis from proghrelin in mammals is not clear, re-naming
terminal region (Fig. 2). Goldfish and zebrafish ghrelin have a this sequence as ghrelin-associated peptide could lead to further
substitution in the second amino acid from Ser to Thr confusions in this already controversial research field.
(Unniappan et al., 2002; Kojima and Kangawa, 2005). Bullfrog
ghrelin has two amino acids substituted in the second (Ser to 4. Tissue distribution and localization of ghrelin
Leucine (Leu)) and third (Ser to Thr) positions (Kaiya et al.,
2001). On the other hand, there is little identity of the region of In rats, ghrelin is primarily produced in the mucosal layer of
shark ghrelins (GVSFHPR), although Ser-3 has been modified stomach from the classically known X/A-like cells that is now
by octanoic or decanoic acid (Kawakoshi et al., 2007). known as ghrelin cells (Kojima et al., 1999; Date et al., 2000).
Interestingly, the shark ghrelin can increase intracellular Ghrelin immunoreactive cells are also found in the duodenum,
calcium concentration of the cells expressing rat GHS-R1a, and their number are decreased gradually in distal intestines from
indicating the peptide can bind to rat GHS-R1a. ileum to colon (Date et al., 2000). Ghrelin is also produced in the
Author's personal copy

114 H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128
Author's personal copy

H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128 115

Table 2
Tissue distribution of ghrelin in vertebrates
Hammerhead shark Eel Goldfish Catfish Rainbow trout Tilapia Bullfrog Turtle Chicken Rat
Brain ++ ++ +++ N.D. ++ +++ – – ++ +
Hypothalamus ++ N.D. + + ++ N.D. N.D. N.D. N.D. +
Pituitary N.D. N.D. – ++ N.D. N.D. N.D. N.D. N.D. +
Heart +++ + – + +++ – – – – +
Lung N.P. N.P. N.P. N.P. N.P. N.P. + – + +
Stomach ++++ ++++ N.P. ++++ ++++ ++++ ++++ ++++ ++++ ++++
Pancreas N.D. N.D. N.D. +++ N.D. N.D. + + – +
Spleen + N.D. ++++ + + N.D. – – + N.D.
Liver ++ N.D. + + – N.D. – – – +
Intestine +++ ++++ +++ ++ +++ – ++ ++ + +++
Rectal gland + N.P. N.P. N.P. N.P. N.P. N.P. N.P. N.P. N.P.
Adrenal gland N.P. N.P. N.P. N.P. N.P. N.P. – – N.D. +
Head kidney N.D. + N.D. + + N.D. N.P. N.P. N.P. N.P.
Trunk kidney + ++ – ++ + – – – – +
Gall bladder N.D. N.D. N.D. +++ N.D. N.D. + N.D. N.D. N.D.
Ovary N.D. N.D. – N.D. N.D. N.D. N.D. N.D. + N.D.
Testis N.D. N.D. – N.D. N.D. N.D. + N.D. N.D. +
Gill ++ – +++ ++ + + N.D. N.P. N.P. N.P.
This table showed relative level of ghrelin based on expression level of ghrelin mRNA or ghrelin content.
++++: very high; +++: high; ++: moderate; +: low; –: not detected; N.D.: not determined; N.P.: not present.

hypothalamic arcuate nucleus (Kojima et al., 1999; Lu et al., 2001, 2002). Ghrelin mRNA expression using reverse-tran-
2002; Mondal et al., 2005), pancreatic A-cells (Kageyama et al., scription-polymerase chain reaction (RT-PCR) was detected in
2005), kidney (Mori et al., 2000) and pituitary (Korbonits et al., the stomach of chicken (Kaiya et al., 2002; Wada et al., 2003),
2001) of rats, mice and humans. However, the expression is quail (Yoshimura et al., 2005), bullfrog (Kaiya et al., 2001),
relatively lower than that of the gastrointestinal tract (Hosoda et turtle (Kaiya et al., 2004), Japanese eel (Kaiya et al., 2003a),
al., 2000a,b). tilapia (Parhar et al., 2003; Kaiya et al., 2003b), rainbow trout
In non-mammalian vertebrates, ghrelin peptide was purified (Kaiya et al., 2003c), channel catfish (Kaiya et al., 2005),
from stomach extracts, and ghrelin immunoreactivity and its seabream (Yeung et al., 2006), sea bass (Terova et al., 2008) and
mRNA expression have been detected mainly in the stomach or hammerhead shark (Kawakoshi et al., 2007). Goldfish lacks a
intestine, indicating that gastrointestinal tract is the major site of stomach, but northern blot and RT-PCR analyses demonstrated
ghrelin production in vertebrates. However, expression of a strong expression of ghrelin mRNA in the gut (Unniappan
ghrelin mRNA was found in various organs of non-mammalian et al., 2002). Therefore, the gastrointestinal tract is recognized
vertebrates other than stomach and there is a species-specific as the main site for ghrelin production in non-mammalian
difference in ghrelin expression. For example, ghrelin expres- vertebrates.
sion in the brain was found in chicken, eel, Mozambique tilapia, Ghrelin-immunoreactive (ir) cells are detected in the
rainbow trout, catfish and hammerhead shark, but not in turtle. mucosal layer of the stomach in chicken (Wada et al., 2003;
Expression in the intestine was found in chicken, turtle, eel, Yamato et al., 2005), quail (Yoshimura et al., 2005), turtle
rainbow trout, catfish and hammerhead shark, but not in (Kaiya et al., 2004), frogs (Galas et al., 2002; Kaiya et al.,
Mozambique tilapia. Ghrelin mRNA is expressed in the kidney 2007b), rainbow trout (Sakata et al., 2004) and Japanese eel
of eel, rainbow trout and catfish, but not in chicken, turtle, (Kaiya et al., 2006b). Recently, Olsson et al. (2008) reported
Mozambique tilapia and hammerhead shark. Table 2 sum- localization of ghrelin-ir cells in the gut of zebrafish. The
marizes the tissue distribution of ghrelin in vertebrates. The antibody used in these studies recognizes only acylated (active-
following sections will highlight our knowledge of ghrelin form) ghrelin, indicating that immunopositive cells show the
distribution in the stomach and brain, and ghrelin levels in blood presence of bioactive ghrelin (Hosoda et al., 2000a). Also
and tissues in non-mammals. recently, Kaiya et al. (2006a) showed localization of ghrelin-ir
cells in the bullfrog stomach using the antisera raised against
4.1. Stomach bullfrog ghrelin C-terminal fragment 13–28. This antibody can
recognize both acylated and des-acyl ghrelin, and is character-
Ghrelin mRNA expression is found in the stomach of istically similar to the antisera for C-RIA (Hosoda et al., 2000a).
bullfrog and chicken by Northern blot analysis (Kaiya et al., In chicken, Neglia et al. (2005) found ghrelin-ir cells in the

Fig. 2. Comparison of prepro-ghrelin peptides from various non-mammalian vertebrates with human prepro-ghrelin sequence. Signal peptide, mature peptide and C-
terminal peptide (C-peptide) regions of the prepro-ghrelin are shown separately. Number of amino acids in amidated mature ghrelin is provided in parentheses of the
middle panel. Black-boxed letter indicates a glycine residue that contributes to create an amide structure of mature ghrelin in teleosts. Parentheses in the lower panel for
C-peptide shows total number of amino acids for prepro-ghrelin with exceptions of edible frog and quail for incomplete sequence. Obestatin region in the C-peptide of
human sequence is shown with broken lines. Prepro-ghrelin sequences were obtained from publications reporting its cloning or from the GenBank (accession numbers
provided in Table 1). Sequences were aligned using the GENETYX-Mac software program.
Author's personal copy

116 H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128

gastrointestinal tissues, but Ahmed and Harvey (2002) failed to Date et al., 2000). Total ghrelin levels in plasma ranged from
detect ghrelin-ir cells in the proventriculus using commercially 150 to 640 fmol/mL in rats, mice and humans (Hosoda et al.,
available antibodies. The difference is due to the variation of 2000a; Date et al., 2000; Yoshimoto et al., 2002; Ariyasu et al.,
recognition site of the antibodies to ghrelin molecule. 2002; Akamizu et al., 2004). Total ghrelin levels in the stomach
of rats range from 1800 to 4600 fmol/mg tissue (Hosoda et al.,
4.2. Brain 2000a,b; Date et al., 2000).
In non-mammalian vertebrates, Mustonen et al. (2002)
Using RT-PCR, ghrelin mRNA expression is detected in the reported plasma levels of ghrelin in a fish, the burbot (Lota lota)
brain of chicken (Kaiya et al., 2002; Saito et al., 2005; Richards before spawn as approximately 470 pg/mL (140 fmol) using a
et al., 2006; Kaiya et al., 2007b), goldfish (Unniappan et al., commercial RIA kit for human ghrelin. They reported the levels
2002), Mozambique tilapia (Kaiya et al., 2003b), rainbow trout reduced during spawn to 350 pg/mL and increased to 780 pg/
(Kaiya et al., 2003c), Japanese eel (Kaiya et al., 2003a) and mL after spawn. Nieminen et al. (2003) reported similar levels
hammerhead shark (Kawakoshi et al., 2007). In chicken, various of plasma ir-ghrelin in burbot (400 pg/mL) and the levels
parts of brain, including corpus striatum, cerebellum, optic lobe reduced by two-week fasting at 2 °C. These results suggest that
and brainstem, express ghrelin mRNA, and the corpus striatum ghrelin may be involved in energy homeostasis in burbot. In
showed the highest expression. In goldfish, telencephalon, goldfish, serum ghrelin levels were found to be 50 fmol/mL at
followed by hypothalamus, express ghrelin mRNA. In hammer- normal feeding state, using a validated RIA that detected the
head shark, dorsal nucleus, followed by forebrain, cerebellum, active-form of ghrelin (Unniappan et al., 2004). Serum ghrelin
hypothalamus and optic tectum were expression sites for the levels significantly increased 3 days after fasting in goldfish,
ghrelin gene. On the other hand, no ghrelin mRNA expression and reached peak levels at 5 days. A post-prandial decrease in
was detected in the brain of the bullfrog (Kaiya et al., 2001), the serum ghrelin concentration was also observed. These
turtle (Kaiya et al., 2004), Nile tilapia (Parhar et al., 2003), results indicate that ghrelin participates in feeding regulation in
catfish (Kaiya et al., 2005), seabream (Yeung et al., 2006) and goldfish. Kaiya et al. (2006b) validated the N-RIA for eel
sea bass (Terova et al., 2008). Further studies are necessary to ghrelin; plasma ghrelin levels were approximately 40 fmol/mL
validate the presence of ghrelin mRNA in specific brain regions in freshwater (FW)-adapted Japanese eels starved for 1 week,
of these animals. and the levels transiently increased to 200 fmol/mL at 6 h after
In the mammalian brain, ghrelin-ir cells are localized in transfer from FW to seawater (SW), suggesting a possible
arcuate nucleus of the hypothalamus, as demonstrated in rats osmoregulatory action of ghrelin in eels. Acylated ghrelin
(Kojima et al., 1999). In addition, Cowley et al. (2003) reported content in the eel stomach was approximately 40 fmol/mg wet
a wide distribution of ghrelin-ir neurons and fibers in rats. tissue, and the levels did not change after transfer of eels from
Among non-mammalian vertebrates, the detailed distribution of FW to SW (Kaiya et al., 2006b). In rainbow trout, active ghrelin
ir-ghrelin in the brain has been reported only in frogs and levels in plasma have been reported as 4–8 fmol/mL (Jönsson et
chicken. Ghrelin-ir neurons were found in the diencephalon and al., 2007), and no pre-prandial and post-prandial changes in
mesencephalon of the frog, R. esculenta (Galas et al., 2002). In plasma ghrelin levels were observed.
addition, a wide distribution of ghrelin-ir nerve fibers was also In amphibians, Kaiya et al. (2006a) established a RIA for the
detected in the telencephalon, diencephalon and mesencephalon measurement of total ghrelin concentration of bullfrog. Ghrelin
(Galas et al., 2002). In chicken, ghrelin-ir cells and nerve fibers levels in plasma and stomach extracts were approximately 50–
are observed in the hypothalamus (Ahmed and Harvey, 2002). 150 fmol/mL and 83–135 fmol/mg wet tissue, respectively. In
However, as the rat ghrelin antibody did not detect ir-ghrelin in bullfrog plasma, the majority of ghrelin immunoreactivity was
chicken proventriculus, it has been suggested that the antibody des-acyl ghrelin and C-terminal fragments of ghrelin. On the
cross-reacts with a protein other than ghrelin in the chicken other hand, the major form of ghrelin stored in the bullfrog
hypothalamus (Wada et al., 2003). No staining for ghrelin was stomach was acylated ghrelin. Total ghrelin levels in plasma and
found in the pituitary and brain of a broiler strain using the stomach significantly increases in frogs fasted for 10 days and
antibody for the active-form of ghrelin (S. Geelissen, R. Wada for 20 days, suggesting a possible involvement of ghrelin in
and T. Sakai, personal communications). feeding behavior in bullfrog.
In birds, Kaiya et al. (2007b) have reported that active
4.3. Ghrelin levels in plasma or tissues ghrelin concentrations in chicken plasma are approximately
20 fmol/mL using the N-RIA for chicken ghrelin, and the levels
Hosoda et al. (2000a) validated two RIAs for ghrelin increase to about 30 fmol/mL 12 h after fasting. The increased
measurement in mammals; the N-RIA that can measure ghrelin levels returned to the control levels 6-h after re-feeding.
acylated, active-form of ghrelin and the C-RIA, that can These results suggest that ghrelin plays a role in “hunger signal”
measure the total concentration of ghrelin including acylated in chicken.
and unacylated des-acyl ghrelin. Using the N-RIA, acylated
ghrelin concentrations in rodent plasma are detected from 5. Ghrelin receptor (GHS-R)
approximately 5 to 80 fmol/mL (Hosoda et al., 2000a; Ariyasu
et al., 2005), and acylated ghrelin content in the rat stomach As mentioned earlier, GHS-R is a G-protein-coupled
ranges from 370 to 1800 fmol/mg tissue (Hosoda et al., 2000a; receptor (GPCR) firstly identified in pig and human (Howard
Author's personal copy

H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128 117

et al., 1996). Two types of GHS-R, GHS-R1a that mediates GHS-R1a-expressing mammalian cells (Howard et al., 1996;
biological actions of GHS and ghrelin, and an inactive, Kojima et al., 1999) or in rat and human pituitary cells
alternative splice variant GHS-R1b have been known. In (Herrington and Hille, 1994; Lania et al., 1998), biphasic Ca2+

Fig. 3. Multiple sequence alignment of the ghrelin receptor (GHS-R) 1a protein in human, chicken, pufferfish, seabream, zebrafish, tilapia and rainbow trout.
Accession number is human GHS-R1a (AY429112), chicken GHS-R1a (AB095995; AJ309542), pufferfish 78B7 (AF082209), seabream GHS-R1a (AY151040),
zebrafish GHS-R1a (XM001335981), zebrafish GHS-R2a (XM001340372), tilapia GHS-R1a (AB361053), rainbow trout GHS-R1a (AB362479).
Author's personal copy

118 H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128

Fig. 4. Multiple sequence alignment of the ghrelin receptor (GHS-R) 1b protein in human, chicken, seabream, tilapia and rainbow trout. Accession number is human
GHS-R1b (U60181), chicken GHS-R1aV (AB095996), seabream GHS-R1b (AY151041), tilapia GHS-R1b (AB361054) and rainbow trout GHS-R1b (AB362481).

increases, which due to a transient Ca2+ release from the truncated form of GHS-R1a. In addition, Sirotkin et al. (2006)
intracellular store and Ca2+ influx through voltage-dependent reported the possible presence of a third GHS-R variant
L-type calcium channel, are observed as the signal transduction. identified in chicken ovary, namely cGHS-R1tv. Unfortunately,
Among non-mammalian vertebrates, GHS-R has been the nucleotide and amino acid sequences of the cGHS-R1tv is
reported in two species of teleosts, black seabream and a unavailable. It is likely that this receptor may be an ortholog
pufferfish, and in a bird, chicken (Figs. 3 and 4). In the black of GHS-R1b as seen in other animals with judging from its
seabream, both types of receptor, GHS-R1a (385 amino acids generation process. Recently, GHS-R has been identified in
(aa)) and GHS-R1b (295 aa), have been identified (Chan and zebrafish (Olsson et al., 2008), Mozambique tilapia and rain-
Cheng, 2004; Chan et al., 2004b). In a pufferfish, Spheroides bow trout (unpublished observations by H. Kaiya). In zebrafish,
nephelus, GHS-R1a ortholog, namely 78B7 (374 aa) has been Olsson et al. (2008) reported 360-amino acid protein. On the
reported (Palyha et al., 2000). In chicken, GHS-R1a (347 aa) other hand, we discovered another GHS-R paralogous sequence
(Geelissen et al., 2003; Tanaka et al., 2003), GHS-R1aV (331 using commercial zebrafish genomic database, which has 365-
aa) (Tanaka et al., 2003) and GHS-R1c (Geelissen et al., 2003) amino acid and shows 74% homology with the a 360-amino
have been reported. Amino acid sequences of GHS-R1a of acid zebrafish GHS-R at amino acid level. Therefore, we
seabream, pufferfish and chicken GHS-R1a have 56, 55 and propose that nomenclature of the firstly identified 360-amino
72% identity to human GHS-R1a, respectively (Fig. 3). GHS- acid protein and the second 365-amino acid protein are named
R1aV and GHS-R1c lack the same portion of 48 bp in the as zebrafish GHS-R1a and GHS-R2a, respectively (Fig. 3).
transmembrane domain-6. Here we propose that nomenclature Whether these receptors are functional remain unknown. In
of these two receptors be standardized as GHS-R1aV since Mozambique tilapia, both GHS-R1a (384 aa) and GHS-R1b
these receptor proteins are identical, and since it is a partial (298 aa) have been identified (Kaiya et al., 2007b, database
Author's personal copy

H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128 119

only). Rainbow trout GHS-R1a and 1b were composed of 387 mammalian vertebrates, ghrelin is also involved in various
aa and 300 aa, respectively (Kaiya et al., 2007b, database only). physiological functions (Table 3). Unniappan and Peter (2005),
Amino acid sequences of GHS-R1a of Mozambique tilapia and and Kaiya et al. (2007b) have previously summarized ghrelin
rainbow trout have 55 and 57% identity to human GHS-R1a, function in fish and birds, respectively.
respectively.
It has been reported that the GHS-R gene has two exons and 6.1. Regulation of hormone release and synthesis
one intron in chicken (Tanaka et al., 2003), seabream (Chan and
Cheng, 2004) and pufferfish (Palyha et al., 2000). Recently A variety of evidence indicates that ghrelin directly acts on
identified tilapia and rainbow trout GHS-R genes have also the pituitary in fish, amphibians, and birds. In addition, it is
composed of two exons and one intron (Kaiya et al., 2007b, likely that circulating ghrelin directly or indirectly acts as an
database only). This gene organization and the insert position of endocrine peptide to stimulate the release of some hormones,
the intron are identical to those of the mammalian GHS-R gene although the regulatory mechanisms need further investigation.
(McKee et al., 1997; Petersenn, 2002), indicating that the GHS-
R gene has been evolutionary conserved from fish to mammals. 6.1.1. Fish
Yeung et al. (2004) isolated 5′-flanking region of the seabream Ghrelin directly affects on the pituitary of fish. In organ-
GHS-R gene, and reported presence of a number of unique cultured pituitary of Mozambique tilapia, the release of GH and
putative transcription factor-binding sites different from human prolactin (PRL) is simulated by ghrelin originated from rat, eel,
counterpart. Interestingly, a GHS-R agonist, GHS enhances the and tilapia (Riley et al., 2002; Kaiya et al., 2003a,b). Recently,
promoter activity, and the stimulatory effect is blocked by a Fox et al. (2007) reported that tilapia ghrelin-stimulated GH
GHS-R antagonist, indicating a homologous up-regulation of release, but not PRL release from dispersed pituitary cells of the
transcription of the GHS-R gene in seabream. This is opposite tilapia. This suggests a possible importance of cell-to-cell
to the regulation in human GHS-R, where the promoter activity communication for regulation of PRL release by ghrelin. In the
is inhibited by the treatment with a GHS-R agonist. study, it has been shown that an effect of tilapia ghrelin C-10
GHS-R1a mRNA is mainly expressed in the pituitary and was more potent than that of tilapia ghrelin C-8. In addition, a
various parts of the brain of chicken (Geelissen et al., 2003; GHS-R antagonist, [D-Lys3]-GHRP-6 blocked the ghrelin's
Tanaka et al., 2003; Saito et al., 2005; Richards et al., 2006) and effects, indicating that ghrelin's effects are mediated by GHS-
seabream (Chan and Cheng, 2004), suggesting direct actions of R1a on the pituitary cells. In seabream, GHS stimulates GH
ghrelin in these tissues. The GHS-R splice variants (GHS-R1b, release from the pituitary (Chan et al., 2004a). The direct action
1aVor 1c) are detected in almost all tissues that expressed GHS- of ghrelin on the pituitary are supported by the fact that GHS-
R1a (Geelissen et al., 2003; Tanaka et al., 2003; Chan and R1a is present in seabream pituitary (Chan and Cheng, 2004). In
Cheng, 2004). However, the expression level of GHS-R splice goldfish, synthetic goldfish ghrelin stimulates the release of GH
variant mRNAs is much lower than that of the GHS-R1a. from perifused pituitary cells and pituitary cells in static culture
Furthermore, the physiological function of GHS-R1b remains to (Unniappan and Peter, 2004). Ran et al. (2004) also observed a
be fully elucidated, although it has been reported that the GH-releasing activity from pituitary fragments by rat ghrelin in
seabream GHS-R1a activity is attenuated to some extent when orange-spotted grouper. In rainbow trout, trout ghrelin stimu-
co-expressed with seabream GHS-R1b in HEK293 cells (Chan lated the release of only GH, but not PRL and somatolactin (SL)
and Cheng, 2004). Expression of GHS-R1a and 1b mRNA were in organ-cultured pituitary (Kaiya et al., 2003c). Furthermore,
detected in the phagocytic leukocytes of rainbow trout, and ghrelin-stimulated GH release from goldfish pituitary cells is
GHS-R1a is involved in immune functions of the leukocytes inhibited by somatostatin-14 (Unniappan and Peter, 2004),
through production of local GH by ghrelin (Yada et al., 2006). suggesting a similar intracellular linkage of ghrelin with
The signal transduction mechanism of GHS-R in non- classical regulation of GH secretion.
mammalian vertebrates has been investigated using mammalian Ghrelin also affects on pituitary hormone secretion through
HEK293 cells expressing seabream GHS-R1a (Chan et al., circulation. Circulating levels of GH increase after IP injections
2004b). They concluded that seabream GHS-R1a is a of a GHS, KP-102 in Nile tilapia (Shepherd et al., 2000), after
constitutively active receptor, which triggers a PLC-PKC- ICV and IP injections of ghrelin in goldfish (Unniappan and
dependent [Ca2+] mobilization in association with L-type Peter, 2004), after IP injection of ghrelin in channel catfish
voltage-gated calcium channel by PKC. Thus, it is likely that (Kaiya et al., 2005) and rainbow trout (Kaiya et al., 2003c;
the characteristics of GHS-R on signal transduction in the cells Shepherd et al., 2007), and after IP injections of tilapia ghrelin
are similar among seabream, rat and human (Herrington and in Mozambique tilapia (Fox et al., 2007). Chronic administra-
Hille, 1994; Lania et al., 1998; Holst et al., 2005). tion of ghrelin using osmotic mini-pumps, however, caused no
change in circulating GH levels after 21 days post-implantation
6. Biological actions of ghrelin (Riley et al., 2005). Similar findings have been reported in rats
(Bowers et al., 1984; Date et al., 2000). It may be due to
In mammals, ghrelin participates in several physiological desensitization of the GH secretory response of somatotrophs
functions other than stimulation of GH release. Several reviews during ghrelin administration. Other than GH, no changes in
have discussed the details (van der Lely et al., 2004; Kojima and circulating levels of PRL are observed after IP injection of a
Kangawa, 2005; Peeters, 2005; Hosoda et al., 2006). In non- GHS in Nile tilapia, of trout ghrelin in rainbow trout or of tilapia
Author's personal copy

120 H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128

Table 3 Summary of ghrelin function examined in non-mammalian vertebrates


Cartilaginous fish Teleostian Amphibian Reptilian Avian Mammalian
Hormone secretion
GH N.D. ↑ ↑ N.D. ↑ ↑
PRL N.D. ↑→ ↑ N.D. N.D. ↑(weak)
SL – → – – – –
ACTH N.D. N.D. N.D. N.D. (↑) ↑(weak)
FSH N.D. – → N.D. → →
LH N.D. ↑ → N.D. N.D. ↑? →
TSH N.D. N.D. N.D. N.D. → ↓? →
TRH N.D. N.D. N.D. N.D. ↑→ →
CRF N.D. N.D. N.D. N.D. ↑→ ↑?↓
Corticosteroids N.D. N.D. N.D. N.D. ↑ ↑(weak)
Insulin N.D. N.D. N.D. N.D. N.D. ↑?↓
Behaviour
Food intake N.D. ↑→ (↑) N.D. ↓ ↑
Water intake N.D. ↓ N.D. N.D. ↓ ↓
Locomotor activity N.D. ↑ N.D. N.D. ↑↓ ↑↓
Metabolism
Adiposity N.D. ↑ N.D. N.D. N.D. ↑
Blood glucose N.D. N.D. N.D. N.D. N.D. ↑?↓
Gastrointestinal function
Acid secretion N.D. N.D. N.D. N.D. N.D. ↑
Motility N.D. N.D. N.D. N.D. ↑ ↑
Turnover of mucosa N.D. N.D. N.D. N.D. N.D. ↑
Cardiovascular function
Cardiac output N.D. N.D. N.D. N.D. N.D. ↑
Blood pressure N.D. N.D. N.D. N.D. N.D. ↓
Note: N.D.: not determined; ↑: increase; ↓: decrease; →: no change.

ghrelin in Mozambique tilapia (Shepherd et al., 2000; Kaiya et goldfish brain (Canosa et al., 2005), indicating possible
al., 2003c; Fox et al., 2007). As mentioned before, ghrelin can interactions between ghrelin and somatostatin in the regulation
stimulate PRL release of Mozambique tilapia from organ- of GH release in goldfish. Hepatic IGF-I mRNA increased by IP
cultured pituitary (Riley et al., 2002; Kaiya et al., 2003a,b). injections of tilapia ghrelin in Mozambique tilapia (Fox et al.,
Further studies are necessary to identify the reasons for different 2007), suggesting that the GH-IGF-I axis is mediated by
PRL response in tilapia and rainbow trout. Besides, plasma SL ghrelin. Recent studies suggest the possible interactions of
levels did not change after IP injections of trout ghrelin in ghrelin with other peptides such as gastrin-releasing peptide/
rainbow trout (Kaiya et al., 2003c). Unniappan and Peter (2004) bombesin (GRP/BBS) to regulate the post-prandial GH surge in
have reported a small increase in serum LH levels in goldfish goldfish (Canosa et al., 2005). Further studies are necessary to
after ICV or IP injection of goldfish ghrelin. In rainbow trout, elucidate the actions and interactions of ghrelin with other
Shepherd et al. (2007) observed increases in plasma IGF-I releasing and release-inhibitory factors in regulating GH
levels 3 h after IP injection of KP-102 or rat ghrelin. Similarly, secretion and release of other pituitary hormones in fish.
Fox et al. (2007) reported IGF-I increase in tilapia plasma 10 h
after IP injections of tilapia ghrelin. Ghrelin affects gene 6.1.2. Amphibians
expression of pituitary hormones. Ghrelin increased GH mRNA Bullfrog and rat ghrelin stimulate the release of GH and PRL
expression in the pituitary of orange-spotted grouper (Ran et al., from dispersed pituitary cells of the bullfrog without any effects
2004) and goldfish (Unniappan and Peter, 2004) in vitro, on LH and FSH release (Kaiya et al., 2001). This result indicates
suggesting a role for ghrelin in regulating GH synthesis in fish. that ghrelin directly acts on the somatotrophs and lactotrophs of
IP injections of ghrelin also increased GH mRNA expression in bullfrog. Interestingly, the potency is 100–1000 times greater
the pituitary of channel catfish and Mozambique tilapia (Kaiya for bullfrog ghrelin than for rat ghrelin, which may be due to the
et al., 2005; Fox et al., 2007). Riley et al. (2005) reported a differences in the structure and affinity of these peptides to the
different stimulatory action on GH synthesis in Mozambique bullfrog ghrelin receptor. Bullfrog ghrelin increases plasma GH
tilapia with tilapia ghrelin C-8 and C-10; ghrelin C-8 increased and PRL levels 15 min after injection when injected IP into
GH mRNA but ghrelin C-10 did not. These in vitro and in vivo juvenile bullfrogs and the elevated levels returns to the basal
results indicate that ghrelin stimulates not only the release but levels by 60 min after the injection (H. Kaiya, A. Koda, K.
also the synthesis of GH in some fish. In goldfish, LH mRNA Yamamoto, S. Kikuyama, unpublished data). This suggests a
expression, as well as LH secretion, is stimulated by ghrelin direct action of circulating ghrelin on somatotrophs and
(Unniappan and Peter, 2004). lactotrophs and the possible existence of a signal transduction
IP injections of ghrelin were found to cause a significant with the vagal system in the bullfrog, as demonstrated in rats
reduction in prepro-somatostatin-II mRNA expression in the (Date et al., 2002).
Author's personal copy

H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128 121

6.1.3. Birds weight gain of the Mozambique tilapia (Riley et al., 2005). It is
Ghrelin stimulates GH release in vivo and in vitro in chicken interesting to note that the effect has been observed with
(Ahmed and Harvey, 2002; Kaiya et al., 2002; Baudet and decanoylated tilapia ghrelin C-10, which is the major form in
Harvey, 2003). Ahmed and Harvey (2002) observed an increase the tilapia, but not with octanoylated tilapia ghrelin C-8.
in plasma GH levels 10 min after IV injection of rat ghrelin in Shepherd et al. (2007) observed increased food intake 2 or 5 h
chicken. The effect was comparable with that of GHRH, but after IP injections of several GH secretagogues such as KP-102
much weaker than that of TRH. Kaiya et al. (2002) observed the and rat ghrelin in juvenile rainbow trout. On the other hand,
same effects using a homologous chicken ghrelin. In this study, Jönsson et al. (2007) has observed no effect of trout ghrelin on
plasma GH levels transiently increased 15 min after IV food intake in two-year-old rainbow trout during 12-h post-IP
injection. In addition to the GH increase, plasma corticosterone injections of trout ghrelin. These results may suggest a species-
levels also increased and the dose–response relationship was specific and a developmental stage-specific action of ghrelin on
much clearer in corticosterone than GH (Kaiya et al., 2002). feeding regulation in fish.
ICV injections of chicken ghrelin also increase plasma It has been reported that ghrelin mRNA expression and
corticosterone levels (Saito et al., 2005). Thus, ghrelin would ghrelin secretion changes during feeding conditions. A post-
act on pituitary or adrenal gland directly or indirectly through prandial decrease in ghrelin mRNA expression in the
the hypothalamo-pituitary-adrenal axis (HPA-axis). Geelissen hypothalamus and gut, and a post-prandial decrease in serum
et al. (2006) reported no effects for chicken ghrelin on thyroxine ghrelin levels are observed in goldfish (Unniappan et al., 2004;
(T3) levels in chicken after IV injections, suggesting no effect Canosa et al., 2005). Fasting for 7 days caused a significant
for ghrelin on at least TSH release from the pituitary. increase in ghrelin mRNA expression in the hypothalamus and
gut of goldfish (Unniappan et al., 2004). A significant increase
6.1.4. Reptiles in serum ghrelin levels is observed 3 days after fasting, the
There are no reports currently available on the in vitro or in increase reaches the maximum at 5 days, and a little decrease
vivo effects of ghrelin in the regulation of any hormone release in serum ghrelin is observed at 7 days after fasting. Matsuda et
in reptiles. From a phylogenic point of view, it is valuable to al. (2006b) also observed a significant increase in ghrelin
investigate the role of ghrelin in the regulation of hormone mRNA expression in the gut, but such an increase was not
release in reptiles. observed in the brain. This may be due to the very low
expression of the ghrelin gene in the hypothalamus. These
6.2. Appetite regulatory effects meal-related increases in ghrelin mRNA expression in the
brain and gut, and ghrelin levels in blood support the view that
Ghrelin is an orexigen in mammals (Nakazato et al., 2001; ghrelin is involved in the regulation of food intake in goldfish.
Tschöp et al., 2000; Wren et al., 2000, 2001). Ghrelin stimulates In burbot (Lota lota), however, a significant reduction in
food intake when injected centrally or peripherally, and it is plasma ir-ghrelin levels was observed after a two-week fast at
known that various hypothalamic neurons such as NPY/AGRP, 2 °C, but not at 10 °C (Nieminen et al., 2003). This is an
orexin, and POMC interact with the ghrelin signal system opposite response compared with other animals. Furthermore,
(Horvath et al., 2001; Date et al., 2006; Gil-Campos et al., 2006). in Nile tilapia, it has been demonstrated using quantitative RT-
A variety of evidence supports the participation of ghrelin in PCR that ghrelin mRNA levels remain unchanged after 7-day
modifying feeding behaviors in non-mammalian vertebrates. food deprivation (Parhar et al., 2003). Food intake in fish is
regulated by the co-ordination of multiple, redundant neu-
6.2.1. Fish roendocrine pathways (Volkoff et al., 2005). The lack of an up-
The regulation of food intake has been studied using regulation in ghrelin mRNA expression in Nile tilapia after
goldfish, tilapia and rainbow trout. Central administration of 7 days of starvation might be due to a compensatory decrease
human ghrelin, octanoylated 19-amino acid and 12-amino acid or increase in one or more of the appetite regulatory peptides.
goldfish ghrelins stimulate food intake in goldfish (Unniappan IP injections of the anorexigen GRP/BBS inhibit ghrelin
et al., 2002; Unniappan et al., 2004). Matsuda et al. (2006a,b) mRNA expression in the gut of goldfish (Canosa et al., 2005),
also found that ICV injections of octanoylated, 12-amino acid suggesting interactions between ghrelin and other appetite
goldfish ghrelin stimulate food intake in goldfish. Recently, regulatory peptides in fish.
Miura et al. revealed that ICV ghrelin-induced food intake in Des-acyl ghrelin, which is a form lacking acyl modification
goldfish is mediated by NPY and orexin, as seen in rodents of ghrelin Ser-3, has been known to have some biological
(Miura et al., 2006, 2007). actions such as anti-apoptotic, anti-proliferative and adipogenic
IP injections of ghrelin are also effective to stimulate food effects in mammals (Baldanzi et al., 2002; Cassoni et al., 2001,
intake (Unniappan et al., 2004; Matsuda et al., 2006a,b). The 2004; Thompson et al., 2004). Stimulatory and inhibitory
peripheral ghrelin-induced food intake in goldfish is mediated effects of des-acyl ghrelin on food intake through different
by a mechanism through capsaicin-sensitive vagal afferent, as regulatory pathways have been reported (Asakawa et al., 2005;
demonstrated in rats (Date et al., 2002; Matsuda et al., 2006a). Toshinai et al., 2006). Central and peripheral injections of des-
In agreement with these studies using acute injections of acyl goldfish ghrelin were found ineffective in regulating food
ghrelin, chronic administration of ghrelin using osmotic mini- intake of goldfish (Matsuda et al., 2006a). However, ICV or IP
pumps for 21 days also resulted in an increase in food intake and pretreatment of the des-acyl ghrelin attenuates ghrelin-induced
Author's personal copy

122 H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128

food intake in a dose-dependent manner (Matsuda et al., 2006a). induced gonadotropin secretion, but direct stimulatory actions
The mechanisms underlying the inhibitory action of des-acyl on basal LH and FSH secretion by long-term treatment (Garcia
ghrelin is currently unknown. et al., 2007; Tena-Sempere, 2005a,b).

6.2.2. Amphibians 6.3.1. Fish


There are no reports about a role of ghrelin on feeding There are some reports that suggest possible involvement of
regulation in amphibians. However, Kaiya et al. (2006a) reported ghrelin in reproduction in non-mammalian vertebrates. Plasma
that plasma ghrelin levels, stomach ghrelin content and stomach ir-ghrelin levels were found to increase after spawning in burbot
ghrelin mRNA levels did not change after 5-day starvation in (Mustonen et al., 2002) although the functional significance of
bullfrog. However, ghrelin mRNA was significantly increased this is currently unknown. They have suggested stimulation of
after 10-day starvation, suggesting a possible involvement of gluconeogenesis by ghrelin-released GH and increase in
ghrelin in energy metabolism and feeding behavior in bullfrog. appetite in post-spawning fish. A sexually dimorphic expression
of ghrelin in the stomach of the Nile tilapia has been reported
6.2.3. Birds (Parhar et al., 2003), pointing towards a linkage between ghrelin
In discordance with the orexigenic actions of ghrelin and the reproductive axis. In goldfish, ghrelin stimulates LH
reported in mammals and fish, ICV administration of ghrelin release and LH-β mRNA expression in pituitary cells
decreases food intake in neonatal chicks (Furuse et al., 2001; (Unniappan and Peter, 2004), indicating direct actions of
Saito et al., 2002a, 2005). Saito et al. (2005) revealed that the ghrelin on goldfish gonadotrophs. The minimal effective dose
anorectic effect of ghrelin is mediated via corticotropin- of ghrelin is comparable to that of gonadotropin-releasing
releasing factor (CRF): ICV injections of ghrelin increased hormone (GnRH) and NPY (Marchant et al., 1989; Peng et al.,
plasma corticosterone levels in chicken and the increase in 1993), suggesting physiological importance of ghrelin on the
corticosterone and anorexia was attenuated by the co-adminis- regulation of LH release and synthesis in goldfish. Further
tration of ghrelin and a CRF receptor antagonist, astressin. studies, including those examining the role of steroid hormones
Similar to what has been reported in chicken, ICV injections of in the regulation of ghrelin synthesis and release, will be helpful
ghrelin inhibit food intake in Japanese quails (Shousha et al., in unraveling the role of ghrelin in the reproductive physiology
2005). However, IP injections of high doses of ghrelin inhibited of fish.
food intake, and small doses of ghrelin were found to stimulate
food intake in Japanese quail. The reason of the different effect 6.3.2. Birds
by the doses injected has considered a different permeability of Yoshimura et al. (2005) identified ir-ghrelin and its mRNA in
ghrelin into central nervous system (Shousha et al., 2005). the oviduct of laying Japanese quails. Ghrelin immunoreactivity
IV injections of chicken ghrelin (1 nmol/100 g body weight) and mRNA expression were also found in the infundibulum and
decrease food intake during 1 h post-injection, as well as magnum, but not in the isthmus, uterus and vagina. The intensity
respiratory quotient for 14 h post-injection in chickens of ghrelin immunoreactivity in the infundibulum and magnum
(Geelissen et al., 2006). However, Kaiya et al. (2007b) reported reduced post-ovulation, suggesting secretory activity of ghrelin
no change in food intake during 2 h after IV injection of chicken during the passage of eggs through oviduct. The physiological
ghrelin (600 pmol/100 g body weight). The difference of relevance remains unknown, but a possible involvement of
response might be due to the chicken strain used for each study: ghrelin in embryonic development through GH has been
Geelissen et al. (2006) used a broiler strain, and Kaiya et al. proposed (Decuypere and Buyse, 2005). Furthermore, Sirotkin
(2007b) used a layer strain. and colleagues have reported possible involvement of ghrelin in
the control of gonadal functions in chicken, such as proliferation,
6.2.4. Reptiles apoptosis, and hormone secretion (Sirotkin et al., 2006). Ghrelin
There are no reports currently available on the effects of and GHS-R1a mRNA expression was found in chicken ovarian
ghrelin in regulating food intake in reptiles. Given that ghrelin tissue and cultured chicken ovarian cells. A ghrelin analog 1–18
regulates appetite in fish and birds, it will be interesting to study increased makers of proliferation (PCNA, cyclin) and decreased
the role of ghrelin in regulating energy balance reptiles. makers of apoptosis (caspase-3, bax, bcl-2 and TUNEL-positive
cells) in ovarian granulosa cells. In chicken ovarian follicular
6.3. Reproduction fragments, ghrelin 1–18 increased PCNA, cyclin, bax and p53,
where ghrelin stimulated the release of progesterone, estradiol,
Kojima et al. (1999) reported no effects of ghrelin on the arginin–vasotocin (AVT) and IGF-1, but not testosterone. These
release of LH and follicle-stimulating hormone (FSH) in secretory activities of ghrelin on ovarian cell are mediated through
dispersed rat pituitary cells, supposed no reproductive effects. TK-, MAPK-, CDK- or OKA-dependent intracellular mechan-
However, recent studies have indicated some involvement of isms (Sirotkin and Grossmann, 2007).
ghrelin in gonadal and reproductive function in mammals. For
example, ghrelin and/or GHS-R1a express in testicular Sertoli 6.4. Gastrointestinal motility
and Leydig cells of rats, regulate seminiferous tubule functions
and inhibit testosterone secretion. In addition, ghrelin shows Ghrelin promotes gastric emptying in mammals when
predominant inhibitory effects at central level and on GnRH- injected centrally and peripherally. The motor effects of ghrelin
Author's personal copy

H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128 123

are regulated by efferent pathways from the central nervous ghrelin. It is possible that similar regulatory systems are present
system, vagal afferents and the enteric nervous system (Peeters, among vertebrates.
2005). There are two reports about the effect of ghrelin on
gastrointestinal motility in chickens. In layer chicks, ICV 6.7. Growth effects
injections of a peptidyl GHS, GHRP-6 did not affect
gastrointestinal motility (Khan et al., 2006). Since GHS inhibits Ghrelin mRNA expression increases before the “rapid
food intake in neonatal chicks when injected centrally (Saito growth period” in Nile tilapia, suggesting its direct role in the
et al., 2002a,b; Khan et al., 2006), these results were found growth of tilapia (Parhar et al., 2003). It has been reported that
supportive for the anorexigenic actions. However, Kitazawa ghrelin affects IGF-I release in Mozambique tilapia and rainbow
et al. (2007) clearly demonstrated different actions of various trout (Fox et al., 2007; Shepherd et al., 2007). In rainbow trout,
GHS-R agonists on contractile effect on the chicken gastro- plasma levels of the 32 kDa, 42 kDa and 50 kDa IGF binding
intestinal tract. Homologous chicken ghrelin stimulated the protein (IGFBP) vary after KP102 and rat ghrelin (Shepherd et
contraction of the crop and proventriculus, but GHRP-6 and rat al., 2007), suggesting a ghrelin action on the somatotropic axis.
and human ghrelins showed only weak stimulatory effects. Continuous infusion of tilapia ghrelin C-10 but not C-8 for
Kitazawa at al. (2007) also showed that chicken ghrelin caused 21 days using osmotic mini-pumps resulted in increases in liver
contractile responses in smooth muscle strips isolated from weight, total lipid content in the liver and muscle of
several parts of the gastrointestinal tract such as esophagus, Mozambique tilapia although the longitudinal growth did not
crop, proventriculus, duodenum, jejunum and colon, and that increase and plasma IGF-1 levels were decreased (Riley et al.,
localization of functional GHS-R in the gastrointestinal wall is 2005). The reduction of fat mobilization and stimulation of
region- and species-specific. In addition, chicken ghrelin and lipogenesis are similar with that observed in rodents (Tschöp et
motilin, a ghrelin-related gut peptide, both caused contraction of al., 2000). In chicken, bolus IV injections of ghrelin had no
chicken gastrointestinal tract with a region-dependent manner: apparent effects on lipid metabolism, as plasma glucose,
ghrelin mainly stimulated the contractions of upper (crop) and triglyceride, free fatty acids protein levels, as well as
lower (colon) intestines, while motilin principally stimulated thermogenesis were unaffected (Geelissen et al., 2006).
those of middle and small intestines. It will be interesting to
compare the effects of ghrelin on the motility of different parts 6.8. Behavior
of the gastrointestinal tracts of other non-mammalian
vertebrates. In goldfish, ICV or IP injection of octanoylated 12-amino
acid goldfish ghrelin increases locomotor activity of goldfish,
6.5. Regulation of water intake although des-acyl ghrelin has no effect, suggesting specific
actions of the ghrelin–GHS-R system on locomotor behavior of
Kozaka et al. (2003) demonstrated that the intracranial fish (Matsuda et al., 2006a,b).
injection of ghrelin inhibited water intake in the seawater- In chicken, ICV injections of ghrelin induced hyperactiv-
adapted eel. This result provides further evidence for the central ity and a sleep-like behavior (Tachibana et al., 2001; Saito et
actions of ghrelin in fish. Furthermore, it has been shown that
ghrelin potently inhibits water intake in neonatal chicks when
injected ICV (Tachibana et al., 2006). Recently, similar
inhibitory effects on water intake have been reported in rats
(Hashimoto et al., 2007), via actions on the area postrema (AP)
and the nucleus of the tractus solitarius (NTS). These results
from fish to mammals indicate that regulation of water intake by
ghrelin may be a conserved physiological action in vertebrates.

6.6. Immunity

In mammals, ghrelin participates in immune functions.


Hattori et al. (2001) has reported expression of the GHS-R gene
in human T cells, B cells and neutrophils. Dixit and Taub (2005)
revealed that ghrelin inhibits expression of proinflammatory
cytokine in human T cells and monocytes. These results suggest
that ghrelin acts on the immune cells through GHS-R.
In non-mammalian vertebrates, Yada et al. (2006) demon-
strated actions by ghrelin on the fish immune system using Fig. 5. Multiple physiological functions of ghrelin in non-mammalian
rainbow trout. Ghrelin stimulates superoxide production and vertebrates. Biological actions that ghrelin was found to regulate at least in
one non-mammalian vertebrate have been included. Information used to develop
activates phagocytosis in leukocytes derived from the head this figure was obtained from published articles on ghrelin in non-mammalian
kidney of rainbow trout. Interestingly, these effects were found vertebrates, and in some instances based on unpublished, preliminary
mediated by GH, secreted from leucocytes in response to observations.
Author's personal copy

124 H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128

al., 2002b). These behaviors are contradictory to each other References


but are observed during a different time course: hyperactivity
was induced during the first 30 min, and then sleep-like Ahmed, S., Harvey, S., 2002. Ghrelin: a novel hypothalamic GH-releasing factor
in domestic fowl (Gallus domesticus). J. Endocrinol. 172, 117–125.
behavior occurred during the next 30 min. The hyperactivity
Akamizu, T., Takaya, K., Irako, T., Hosoda, H., Teramukai, S., Matsuyama, A.,
is due to the activation of hypothalamic CRF system by Tada, H., Miura, K., Shimizu, A., Fukushima, M., Yokode, M., Tanaka, K.,
ghrelin in relation to inhibition of food intake (Saito et al., Kangawa, K., 2004. Pharmacokinetics, safety, and endocrine and appetite
2005). effects of ghrelin administration in young healthy subjects. Eur. J.
Endocrinol. 150, 447–455.
Ariyasu, H., Takaya, K., Hosoda, H., Iwakura, H., Ebihara, K., Mori, K.,
7. Perspectives
Ogawa, Y., Hosoda, K., Akamizu, T., Kojima, M., Kangawa, K., Nakao, K.,
2002. Delayed short-term secretory regulation of ghrelin in obese animals:
There is enough evidence available now to highlight the evidenced by a specific RIA for the active form of ghrelin. Endocrinology
importance of ghrelin as a multifaceted endocrine regulator in 143, 3341–3350.
non-mammalian vertebrates (Fig. 5). The high similarity in the Ariyasu, H., Takaya, K., Iwakura, H., Hosoda, H., Akamizu, T., Arai, Y.,
Kangawa, K., Nakao, K., 2005. Transgenic mice overexpressing des-acyl
structure of ghrelin gene, peptide and GHS-R1a, distribution
ghrelin show small phenotype. Endocrinology 146, 355–364.
pattern of ghrelin and GHS-R1a and the commonality of many of Asakawa, A., Inui, A., Fujimiya, M., Sakamaki, R., Shinfuku, N., Ueta, Y.,
ghrelin's functions among vertebrates indicate that ghrelin is an Meguid, M.M., Kasuga, M., 2005. Stomach regulates energy balance via
evolutionarily conserved, essential hormone in vertebrates. While acylated ghrelin and desacyl ghrelin. Gut 54, 18–24.
ghrelin shares some biological functions across the vertebrates, it Badman, M.K., Flier, J.S., 2005. The gut and energy balance: visceral allies in
the obesity wars. Science 307, 1909–1914.
is also clear that some functions are more species-specific. Based
Baldanzi, G., Filigheddu, N., Cutrupi, S., Catapano, F., Bonissoni, Fubini, A.,
on the results reported so far, it is reasonable to conclude that the Malan, D., Baj, G., Granata, R., Broglio, F., Papotti, M., Surico, N.,
regulation of GH release and feeding would be two major Bussolino, F., Isgaard, J., Deghenghi, R., Sinigaglia, F., Prat, M., Muccioli,
functions of ghrelin in vertebrates. Future studies employing G., Ghigo, E., Graziani, A., 2002. Ghrelin and des-acyl ghrelin inhibit cell
cellular, molecular and physiological tools and in vitro and in death in cardiomyocytes and endothelial cells through ERK1/2 and PI 3-
kinase/AKT. J. Cell Biol. 159, 1029–1037.
vivo approaches could enable us to better understand the biology
Baudet, M.L., Harvey, S., 2003. Ghrelin-induced GH secretion in domestic fowl
of ghrelin, not only in non-mammalian vertebrates but also in all in vivo and in vitro. J. Endocrinol. 179, 97–105.
vertebrates. Some of the properties of ghrelin, especially its role Bednarek, M.A., Feighner, S.D., Pong, S.S., McKee, K.K., Hreniuk, D.L., Silva,
in the regulation of food intake and GH release are currently M.V., Warren, V.A., Haward, A.D., Van Der Ploeg, L.H., Heck, J.V., 2000.
being explored to develop potential therapies for the treatment of Structure-function studies on the new growth hormone-releasing peptide,
ghrelin: minimal sequence of ghrelin necessary for activation of growth
several endocrine pathophysiologies in humans. Utilizing the
hormone secretagogue receptor. J. Med. Chem. 43, 4370–4376.
potential of the effects of ghrelin on feeding and pituitary Bowers, C.Y., Momany, F.A., Reynolds, G.A., Hong, A., 1984. On the in
hormone release in non-mammalian vertebrates could also be vitro and in vivo activity of a new synthetic hexapeptide that acts on the
beneficial, for example, in poultry and aquaculture to promote pituitary to specifically release growth hormone. Endocrinology 114,
growth and weight gain of cultured species. 1537–1545.
Bresciani, E., Rapetti, D., Donà, F., Bulgarelli, I., Tamiazzo, L., Locatelli, V.,
Torsello, A., 2006. Obestatin inhibits feeding but does not modulate GH and
8. Summary corticosterone secretion in the rat. J. Endocrinol. Investig. 29, RC16–RC18.
Canosa, L.F., Unniappan, S., Peter, R.E., 2005. Periprandial changes in growth
In this review, we discussed many aspects of ghrelin from hormone release in goldfish: role of somatostatin, ghrelin, and gastrin-
fish to birds, in comparison to mammals. Ghrelin is a conserved releasing peptide. Am. J. Physiol. Regul. Integr. Comp. Physiol. 289,
R125–R133.
hormone during vertebrate evolution and have multiple
Cassoni, P., Papotti, M., Ghe, C., Catapano, F., Sapino, A., Graziani, A.,
fundamental biological effects. Research on non-mammalian Deghenghi, R., Reissmann, T., Ghigo, E., Muccioli, G., 2001. Identification,
ghrelin is at its infancy. Comparative endocrinological characterization, and biological activity of specific receptors for natural
approaches using non-mammalian vertebrates would act as (ghrelin) and synthetic growth hormone secretagogues and analogs in
strong tools to better understand the commonality of ghrelin in human breast carcinomas and cell lines. J. Clin. Endocrinol. Metab. 86,
1738–1745.
vertebrates.
Cassoni, P., Ghe, C., Marrocco, T., Tarabra, E., Allia, E., Catapano, F.,
Deghenghi, R., Ghigo, E., Papotti, M., Muccioli, G., 2004. Expression of
Acknowledgments ghrelin and biological activity of specific receptors for ghrelin and des-acyl
ghrelin in human prostate neoplasms and related cell lines. Eur. J.
SU is funded by a Discovery Grant and a Research Tools and Endocrinol. 150, 173–184.
Chan, C.B., Cheng, C.H., 2004. Identification and functional characterization of
Instruments Grant from the Natural Sciences and Engineering
two alternatively spliced growth hormone secretagogue receptor transcripts
Research Council (NSERC) of Canada. REP received funding from the pituitary of black seabream, Acanthopagrus schlegeli. Mol. Cell.
from the NSERC of Canada. HK, MM and KK are supported by Endocrinol. 214, 81–95.
a Grant-in-Aid for Scientific Research from the Ministry of Chan, C.B., Fung, C.K., Fung, W., Tse, M.C., Cheng, C.H., 2004a. Stimulation
Education, Culture, Science, Sports and Technology (MEXT) of of growth hormone secretion from seabream pituitary cells in primary
culture by growth hormone secretagogues is independent of growth
Japan, the Ministry of Health, Labor and Welfare of Japan, and
hormone transcription. Comp. Biochem. Physiol. C. 139, 77–85.
the Promotion of Fundamental Studies in Health Science from Chan, C.B., Leung, P.K., Wise, H., Cheng, C.H., 2004b. Signal transduction
the Organization for Pharmaceutical Safety and Research mechanism of the seabream growth hormone secretagogue receptor. FEBS
(ORSR) of Japan. Lett. 577, 147–153.
Author's personal copy

H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128 125

Chartrel, N., Alvear-Perez, R., Leprince, J., Iturrioz, X., Reaux-Le Goazigo, A., Gourcerol, G., Tach, é, Y., 2007. Obestatin—a ghrelin-associated peptide that
Audinot, V., Chomarat, P., Coge, F., Nosjean, O., Rodriguez, M., Galizzi, does not hold its promise to suppress food intake and motility.
J.P., Boutin, J.A., Vaudry, H., Llorens-Cortes, C., 2007. Comment on Neurogastroenterol. Motil. 19, 161–165.
“Obestatin, a peptide encoded by the ghrelin gene, opposes ghrelin's Gourcerol, G., St-Pierre, D.H., Tach, é, Y., 2007. Lack of obestatin effects on
effects on food intake”. Science 315, 766. food intake: should obestatin be renamed ghrelin-associated peptide (GAP)?
Chen, C.Y., Inui, A., Asakawa, A., Fujino, K., Kato, I., Chen, C.C., Ueno, N., Regul. Pept. 141, 1–7.
Fujimiya, M., 2005. Des-acyl ghrelin acts by CRF type 2 receptors to disrupt Hashimoto, H., Fujihara, H., Kawasaki, M., Saito, T., Shibata, M., Otsubo, H.,
fasted stomach motility in conscious rats. Gastroenterology 129, 8–25. Takei, Y., Ueta, Y., 2007. Centrally and peripherally administered ghrelin
Cowley, M.A., Smith, R.G., Diano, S., Tschöp, M., Pronchuk, N., Grove, K.L., potently inhibits water intake in rats. Endocrinology 148, 1638–1647.
Strasburger, C.J., Bidlingmaier, M., Esterman, M., Heiman, M.L., Garcia- Hattori, N., Saito, T., Yagyu, T., Jiang, B.H., Kitagawa, K., Inagaki, C., 2001.
Segura, L.M., Nillni, E.A., Mendez, P., Low, M.J., Sotonyi, P., Friedman, J.M., GH, GH receptor, GH secretagogue receptor, and ghrelin expression in
Liu, H., Pinto, S., Colmers, W.F., Cone, R.D., Horvath, T.L., 2003. The human T cells, B cells, and neutrophils. J. Clin. Endocrinol. Metab. 86,
distribution and mechanism of action of ghrelin in the CNS demonstrates a 4284–4291.
novel hypothalamic circuit regulating energy homeostasis. Neuron 37, Herrington, J., Hille, B., 1994. Growth hormone-releasing hexapeptide elevates
649–661. intracellular calcium in rat somatotropes by two mechanisms. Endocrinol-
Date, Y., Kojima, M., Hosoda, H., Sawaguchi, A., Mondal, M.S., Suganuma, T., ogy 135, 1100–1108.
Matsukura, S., Kangawa, K., Nakazato, M., 2000. Ghrelin, a novel growth Holst, B., Brandt, E., Bach, A., Heding, A., Schwartz, T.W., 2005. Nonpeptide
hormone-releasing acylated peptide, is synthesized in a distinct endocrine and peptide growth hormone secretagogues act both as ghrelin receptor
cell type in the gastrointestinal tracts of rats and humans. Endocrinology agonist and as positive or negative allosteric modulators of ghrelin signaling.
141, 4255–4261. Mol. Endocrinol. 19, 2400–2411.
Date, Y., Murakami, N., Toshinai, K., Matsukura, S., Niijima, A., Matsuo, H., Holst, B., Egerod, K.L., Schild, E., Vickers, S.P., Cheetham, S., Gerlach, L.O.,
Kangawa, K., Nakazato, M., 2002. The role of the gastric afferent vagal Storjohann, L., Stidsen, C.E., Jones, R., Beck-Sickinger, A.G., Schwartz,
nerve in ghrelin-induced feeding and growth hormone secretion in rats. T.W., 2007. GPR39 signaling is stimulated by zinc ions but not by
Gastroenterology 123, 1120–1128. obestatin. Endocrinology 148, 13–20.
Date, Y., Shimbara, T., Koda, S., Toshinai, K., Ida, T., Murakami, N., Miyazato, Horvath, T.L., Diano, S., Sotonyi, P., Heiman, M., Tschop, M., 2001.
M., Kokame, K., Ishizuka, Y., Ishida, Y., Kageyama, H., Shioda, S., Minireview: ghrelin and the regulation of energy balance—a hypothalamic
Kangawa, K., Nakazato, M., 2006. Peripheral ghrelin transmits orexigenic perspective. Endocrinology 142, 4163–4169.
signals through the noradrenergic pathway from the hindbrain to the Hosoda, H., Kojima, M., Matsuo, H., Kangawa, K., 2000a. Ghrelin and des-acyl
hypothalamus. Cell Metab. 4, 323–331. ghrelin: two major forms of rat ghrelin peptide in gastrointestinal tissue.
Davenport, A.P., Bonner, T.I., Foord, S.M., Harmar, A.J., Neubig, R.R., Pin, Biochem. Biophys. Res. Commun. 279, 909–913.
J.P., Spedding, M., Kojima, M., Kangawa, K., 2005. International Union Hosoda, H., Kojima, M., Matsuo, H., Kangawa, K., 2000b. Purification and
of Pharmacology. LVI. Ghrelin receptor nomenclature, distribution, and characterization of rat des-Gln14-ghrelin, a second endogenous ligand for
function. Pharmacol. Rev. 57, 541–546. the growth hormone secretagogue receptor. J. Biol. Chem. 275,
Decuypere, E., Buyse, J., 2005. Endocrine control of postnatal growth in 21995–22000.
poultry. J. Poult. Sci. 42. Hosoda, H., Kojima, M., Mizushima, T., Shimizu, S., Kangawa, K., 2003.
Dixit, V.D., Taub, D.D., 2005. Ghrelin and immunity: a young player in an old Structural divergence of human ghrelin. Identification of multiple ghrelin-
field. Exp. Gerontol. 40, 900–910. derived molecules produced by post-translational processing. J. Biol. Chem.
Fox, B.K., Riley, L.G., Dorough, C., Kaiya, H., Hirano, T., Grau, E.G., 2007. 278, 64–70.
Effects of homologous ghrelins on the growth hormone/insulin-like Hosoda, H., Kojima, M., Kangawa, K., 2006. Biological, physiological and
growth factor-I in the tilapia, Oreochromis mossambicus. Zool. Sci. 24, pharmacological aspects of ghrelin. J. Pharmacol. Sci. 100, 398–410.
391–400. Howard, A.D., Feighner, S.D., Cully, D.F., Arena, J.P., Liberator, P.A.,
Furuse, M., Tachibana, T., Ohgushi, A., Ando, R., Yoshimatsu, T., Denbow, Rosenblum, C.I., Hamelin, M., Hreniuk, D.L., Palyha, O.C., Anderson, J.,
D.M., 2001. Intracerebroventricular injection of ghrelin and growth Paress, P.S., Diaz, C., Chou, M., Liu, K.K., McKee, K.K., Pong, S.S.,
hormone releasing factor inhibits food intake in neonatal chicks. Neurosci. Chaung, L.Y., Elbrecht, A., Dashkevicz, M., Heavens, R., Rigby, M.,
Lett. 301, 123–126. Sirinathsinghji, D.J., Dean, D.C., Melillo, D.G., Patchett, A.A., Nargund, R.,
Galas, L., Chartrel, N., Kojima, M., Kangawa, K., Vaudry, H., 2002. Griffin, P.R., DeMartino, Ja., Gupta, S.K., Schaeffer, J.M., Smith, R.G., Van
Immunohistochemical localization and biochemical characterization of der Ploeg, L.H., 1996. A receptor in pituitary and hypothalamus that
ghrelin in the brain and stomach of the frog, Rana esculenta. J. Comp. functions in growth hormone release. Science 273, 974–977.
Neurol. 450, 34–44. Jönsson, E., Forsman, A., Einarsdottir, I.E., Kaiya, H., Ruohonen, K.,
Garcia, M.C., Lopez, M., Alvarez, C.V., Casanueva, F., Tena-Sempere, M., Dieguez, Björnsson, B.T., 2007. Plasma ghrelin levels in rainbow trout in response
C., 2007. Role of ghrelin in reproduction. Reproduction 133, 531–540. to fasting, feeding and food composition, and effects of ghrelin on voluntary
Geelissen, S.M., Beck, I.M., Darras, V.M., Kuhn, E.R., Van der Geyten, S., food intake. Comp. Biochem. Physiol. A 147, 1116–1124.
2003. Distribution and regulation of chicken growth hormone secretagogue Kageyama, H., Funahashi, H., Hirayama, M., Takenoya, F., Kita, T., Kato, S.,
receptor isoforms. Gen. Comp. Endocrinol. 134, 167–174. Sakurai, J., Lee, E.Y., Inoue, S., Date, Y., Nakazato, M., Kangawa, K.,
Geelissen, S.M., Swennen, Q., Geyten, S.V., Kuhn, E.R., Kaiya, H., Kangawa, Shioda, S., 2005. Morphological analysis of ghrelin and its receptor
K., Decuypere, E., Buyse, J., Darras, V.M., 2006. Peripheral ghrelin reduces distribution in the rat pancreas. Regul. Pept. 126, 67–71.
food intake and respiratory quotient in chicken. Domest. Anim. Endocrinol. Kaiya, H., Kojima, M., Hosoda, H., Koda, A., Yamamoto, K., Kitajima, Y.,
30, 108–116. Matsumoto, M., Minamitake, Y., Kikuyama, S., Kangawa, K., 2001.
Geris, K.L., Hickey, G.J., Berghman, L.R., Visser, T.J., Kühn, E.R., Darras, Bullfrog ghrelin is modified by n-octanoic acid at its third threonine residue.
V.M., 1998. Pituitary and extrapituitary action sites of the novel nonpeptidyl J. Biol. Chem. 276, 40441–40448.
growth hormone (GH) secretagogue L-692,429 in the chicken. Gen. Comp. Kaiya, H., Van der Gayten, S., Kojima, M., Hosoda, H., Kitajima, Y.,
Endocrinol. 111, 186–196. Matsumoto, M., Geelissen, S., Darra, V.M., Kangawa, K., 2002. Chicken
Geris, K.L., Hickey, G.J., Vanderghote, A., Kuhn, E.R., Darras, V.M., 2001. ghrelin: purification, cDNA cloning, and biological activity. Endocrinology
Synthetic growth hormone secretagogues control growth hormone 143, 3445–3463.
secretion in the chicken at pituitary and hypothalamic levels. Endocrine Kaiya, H., Kojima, M., Hosoda, H., Riley, L.G., Hirano, T., Grau, E.G.,
14, 67–72. Kangawa, K., 2003a. Amidated fish ghrelin: purification, cDNA cloning
Gil-Campos, M., Aguilera, C.M., Canete, R., Gil, A., 2006. Ghrelin: a hormone in the Japanese eel and its biological activity. J. Endocrinol. 176,
regulating food intake and energy homeostasis. Br. J. Nutr. 96, 201–226. 415–423.
Author's personal copy

126 H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128

Kaiya, H., Kojima, M., Hosoda, H., Riley, L.G., Hirano, T., Grau, E.G., Lauwers, E., Landuyt, B., Arckens, L., Schoofs, L., Luyten, W., 2006. Obestatin
Kangawa, K., 2003b. Identification of tilapia ghrelin and its effects on does not activate orphan G protein-coupled receptor GPR39. Biochem.
growth hormone and prolactin release in the tilapia, Oreochromis Biophys. Res. Commun. 351, 21–25.
mossambicus. Comp. Biochem. Physiol. B. Biochem. Mol. Biol. 135, Lu, S., Guan, J.L., Wang, Q.P., Uehara, K., Yamada, S., Goto, N., Date, Y.,
421–429. Nakazato, M., Kojima, M., Kangawa, K., Shioda, S., 2002. Immunocyto-
Kaiya, H., Kojima, M., Hosoda, H., Moriyama, S., Takahashi, A., Kawauchi, H., chemical observation of ghrelin-containing neurons in the rat arcuate
Kangawa, K., 2003c. Peptide purification, cDNA and genomic DNA nucleus. Neurosci Lett. 321, 157–160.
cloning, and functional characterization of ghrelin in rainbow trout. Marchant, T.A., Chang, J.P., Nahorniak, C.S., Peter, R.E., 1989. Evidence that
Endocrinology 144, 5215–5226. gonadotropin-releasing hormone also functions as a growth hormone-
Kaiya, H., Sakata, I., Kojima, M., Hosoda, H., Sakai, T., Kangawa, K., 2004. releasing factor in the goldfish. Endocrinology 124, 2509–2518.
Structural determination and histochemical localization of ghrelin in the red- Matsuda, K., Miura, T., Kaiya, H., Maruyama, K., Shimakura, S., Uchiyama,
eared slider turtle, Trachemys scripta elegans. Gen. Comp. Endocrinol. 138, M., Kangawa, K., Shioda, S., 2006a. Regulation of food intake by acyl and
50–57. des-acyl ghrelins in the goldfish. Peptides 27, 2321–2325.
Kaiya, H., Small, B.C., Lelania Bilodeau, A., Shepherd, B.S., Kojima, M., Matsuda, K., Miura, T., Kaiya, H., Maruyama, K., Uchiyama, M., Kangawa, K.,
Hosoda, H., Kangawa, K., 2005. Purification, cDNA cloning, and Shioda, S., 2006b. Stimulatory effect of n-octanoylated ghrelin on locomotor
characterization of ghrelin in channel catfish, Ictalurus punctatus. Gen. activity in the goldfish, Carassius auratus. Peptides 27, 1335–1340.
Comp. Endocrinol. 143, 201–210. Matsumoto, M., Hosoda, H., Kitajima, Y., Morozumi, N., Minamitake, Y.,
Kaiya, H., Sakata, I., Yamamoto, K., Koda, A., Sakai, T., Kangawa, K., Tanaka, S., Matsuo, H., Kojima, M., Hayashi, Y., Kangawa, K., 2001.
Kikuyama, S., 2006a. Identification of immunoreactive plasma and stomach Structure–activity relationship of ghrelin: pharmacological study of ghrelin
ghrelin, and expression of stomach ghrelin mRNA in the bullfrog, Rana peptides. Biochem. Biophys. Res. Commun. 287, 142–146.
catesbeiana. Gen Comp. Endocrinol. 148, 236–244. McKee, K.K., Palyha, O.C., Feighner, S.D., Hreniuk, D.L., Tan, C.P., Phillips,
Kaiya, H., Tsukada, T., Yuge, S., Mondo, H., Kangawa, K., Takei, Y., 2006b. M.S., Smith, R.G., van der Ploeg, L.H., Howard, A.D., 1997. Molecular
Identification of eel ghrelin in plasma and stomach by radioimmunoassay analysis of rat pituitary and hypothalamic growth hormone secretagogue
and histochemistry. Gen. Comp. Endocrinol. 148, 375–382. receptors. Mol. Endocrinol. 11, 415–423.
Kaiya, H., Darras, V.M., Kangawa, K., 2007a. Ghrelin in birds: its structure, Miura, T., Maruyama, K., Shimakura, S., Kaiya, H., Uchiyama, M., Kangawa,
distribution and function. J. Poult. Sci. 44, 1–18. K., Shioda, S., Matsuda, K., 2006. Neuropeptide Y mediates ghrelin-
Kaiya, H., Saito, E.S., Tachibana, T., Furuse, M., Kangawa, K., 2007b. Changes induced feeding in the goldfish,Carassius. auratus Neurosci. Lett. 407,
in ghrelin levels of plasma and proventriculus and ghrelin mRNA of 279–283.
proventriculus in fasted and refed layer chicks. Domest. Anim. Endocrinol. Miura, T., Maruyama, K., Shimakura, S., Kaiya, H., Uchiyama, M., Kangawa,
32, 247–259. K., Shioda, S., Matsuda, K., 2007. Regulation of food intake in the goldfish
Kawakoshi, A., Kaiya, H., Riley, L.G., Hirano, T., Grau, E.G., Miyazato, M., by interaction between ghrelin and orexin. Peptides 28, 1207–1213.
Hosoda, H., Kangawa, K., 2007. Identification of a ghrelin-like peptide in Mondal, M.S., Date, Y., Yamaguchi, H., Toshinai, K., Tsuruta, T., Kangawa, K.,
two species of shark, Sphyrna lewini and Carcharhinus melanopterus. Gen. Nakazato, M., 2005. Identification of ghrelin and its receptor in neurons of
Comp. Endocrinol. 151, 259–268. the rat arcuate nucleus. Regul. Pept. 126, 55–59.
Khan, M.S.I., Dodo, K., Yahata, K., Nishimoto, S., Ueda, H., Taneike, T., Mori, K., Yoshimoto, A., Takaya, K., Hosoda, K., Ariyasu, H., Yahata, K.,
Kitazawa, T., Hosaka, Y., Bungo, T., 2006. Intracerebroventricular Mukoyama, M., Sugawara, A., Hosoda, H., Kojima, M., Kangawa, K.,
administration of growth hormone releasing peptide-6 (GHRP-6) inhibits Nakao, K., 2000. Kidney produces a novel acylated peptide, ghrelin. FEBS
food intake, but not food retention of crop and stomach in neonatal chicks. J. Lett. 486, 213–216.
Poult. Sci. 43, 35–40. Mustonen, A.M., Nieminen, P., Hyvarinen, H., 2002. Leptin, ghrelin, and
Kitazawa, T., Kaiya, H., Taneike, T., 2007. Contractile effects of ghrelin-related energy metabolism of the spawning burbot (Lota lota, L.). J. Exp. Zool. 293,
peptides on the chicken gastrointestinal tract in vitro. Peptides 28, 617–624. 119–126.
Kojima, M., Kangawa, K., 2005. Ghrelin: structure and function. Physiol. Rev. Nakazato, M., Murakami, N., Date, Y., Kojima, M., Matsuo, H., Kangawa, K.,
85, 495–522. Matsukura, S., 2001. A role for ghrelin in the central regulation of feeding.
Kojima, M., Hosoda, H., Date, Y., Nakazato, M., Matuso, H., Kangawa, K., Nature 409, 194–198.
1999. Ghrelin is a growth-hormone-releasing acylated peptide. Nature 402, Neglia, S., Arcamone, N., Esposito, V., Gargiulo, G., de Girolamo, P., 2005.
656–660. Presence and distribution of ghrelin-immunopositive cells in the chicken
Kojima, M., Hosoda, H., Kangawa, K., 2001a. Purification and distribution of gastrointestinal tract. Acta Histochem. 107, 3–9.
ghrelin: the natural endogenous ligand for the growth hormone secretagogue Nie, Q., Zeng, H., Lei, M., Ishag, N.A., Fang, M., Sun, B., Yang, G., Zhang, X.,
receptor. Horm. Res. 56 (Suppl 1), 93–97. 2004. Genomic organisation of the chicken ghrelin gene and its single
Kojima, M., Hosoda, H., Matsuo, H., Kangawa, K., 2001b. Ghrelin: discovery nucleotide polymorphisms detected by denaturing high-performance liquid
of the natural endogenous ligand for the growth hormone secretagogue chromatography. Br. Poult. Sci. 45, 611–618.
receptor. Trends Endocrinol. Metab. 12, 118–122. Nieminen, P., Mustonen, A.M., Hyvarinen, H., 2003. Fasting reduces plasma
Korbonits, M., Bustin, S.A., Kojima, M., Jordan, S., Adams, E.F., Lowe, D.G., leptin-and ghrelin-immunoreactive peptide concentrations of the burbot
Kangawa, K., Grossman, A.B., 2001. The expression of the growth hormone (Lota lota) at 2 degrees C but not at 10 degrees C. Zool. Sci. 20, 1109–1115.
secretagogue receptor ligand ghrelin in normal and abnormal human pituitary Nishi, Y., Hiejima, H., Hosoda, H., Kaiya, H., Mori, K., Fukue, Y., Yanase, T.,
and other neuroendocrine tumors. J. Clin. Endocrinol. Metab. 86, 881–887. Nawata, H., Kangawa, K., Kojima, M., 2005. Ingested medium-chain fatty
Korbonits, M., Goldstone, A.P., Gueorguiev, M., Grossman, A.B., 2004. acids are directly utilized for the acyl modification of ghrelin. Endocrinology
Ghrelin—a hormone with multiple functions. Front. Neuroendocrinol. 25, 146, 2255–2264.
27–68. Olsson, C., Holbrook, J.D., Bompadre, G., Jönsson, E., Hoyle, C.H., Sanger,
Kozaka, T., Fujii, Y., Ando, M., 2003. Central effects of various ligands on G.J., Holmgren, S., Andrews, P.L., 2008. Identification of genes for the
drinking behavior in eels acclimated to seawater. J. Exp. Biol. 206, 687–692. ghrelin and motilin receptors and a novel related gene in fish, and stimulation of
Lania, A., Ballare, E., Corbetta, S., Filopanti, M., Persani, L., Spada, A., 1998. intestinal motility in zebrafish (Danio rerio) by ghrelin and motilin. Gen.
Growth hormone-releasing hexapeptide (GHRP-6) increases intracellular Comp. Endocrinol. 155, 217–226.
calcium concentrations in cultured cells from human pituitary adenomas of Palyha, O.C., Feighner, S.D., Tan, C.P., McKee, K.K., Hreniuk, D.L., Gao, Y.D.,
different types. Eur. J. Endocrinol. 139, 343–348. Schleim, K.D., Yang, L., Morriello, G.J., Nargund, R., Patchett, A.A.,
Laferrere, B., Abraham, C., Russell, C.D., Bowers, C.Y., 2005. Growth hormone Howard, A.D., Smith, R.G., 2000. Ligand activation domain of human
releasing peptide-2 (GHRP-2), like ghrelin, increases food intake in healthy orphan growth hormone (GH) secretagogue receptor (GHS-R) conserved
men. J. Clin. Endocrinol. Metab. 90, 611–614. from pufferfish to humans. Mol. Endocrinol. 14, 160–169.
Author's personal copy

H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128 127

Parhar, I.S., Sato, H., Sakuma, Y., 2003. Ghrelin gene in cichlid fish is Tachibana, T., Ohgushi, A., Furuse, M., 2001. Intracerebroventricular injection
modulated by sex and development. Biochem. Biophys. Res. Commun. 305, of ghrelin induces sleep-like behavior in neonatal chicks. J. Poult. Sci. 38,
169–175. 358–363.
Peng, C., Chang, J.P., Yu, K.L., Wong, A.O., Van Goor, F., Peter, R.E., Rivier, Tachibana, T., Kaiya, H., Denbow, D.M., Kangawa, K., Furuse, M., 2006.
J.E., 1993. Neuropeptide-Y stimulates growth hormone and gonadotropin-II Central ghrelin acts as an anti-dipsogenic peptide in chicks. Neurosci. Lett.
secretion in the goldfish pituitary: involvement of both presynaptic and 405, 241–245.
pituitary cell actions. Endocrinology 132, 1820–1829. Tanaka, M., Hayashida, Y., Iguchi, T., Nakao, N., Nakai, N., Nakashima, K.,
Peeters, T.L., 2005. Ghrelin: a new player in the control of gastrointestinal 2001. Organization of the mouse ghrelin gene and promoter: occurrence of a
functions. Gut 54, 1638–1649. short non-coding first exon. Endocrinology 142, 3697–3700.
Petersenn, S., 2002. Structure and regulation of the growth hormone Tanaka, M., Miyazaki, T., Yamamoto, I., Nakai, N., Ohta, Y., Tsushima, N.,
secretagogue receptor. Minerva Endocrinol. 27, 243–256. Wakita, M., Shimada, K., 2003. Molecular characterization of chicken growth
Ran, X., Li, W., Lin, H., 2004. Rat ghrelin stimulates GH release and mRNA hormone secretagogue receptor gene. Gen. Comp. Endocrinol. 134, 198–202.
expression in the pituitary of orange-spotted grouper, Epinephelus coioides. Tannenbaum, G.S., Bowers, C.Y., 2001. Interactions of growth hormone
Fish Physiol. Biochem. 30, 95–102. secretagogues and growth hormone-releasing hormone/somatostatin. Endo-
Richards, M.P., Poch, S.M., McMurtry, J.P., 2006. Characterization of turkey crine 14, 21–27.
and chicken ghrelin genes, and regulation of ghrelin and ghrelin receptor Tena-Sempere, M., 2005a. Ghrelin: novel regulator of gonadal function. J.
mRNA levels in broiler chickens. Gen. Comp. Endocrinol. 145, 298–310. Endocrinol. Investig. 28 (5 Suppl), 26–29.
Riley, L.G., Hirano, T., Grau, E.G., 2002. Rat ghrelin stimulates growth Tena-Sempere, M., 2005b. Exploring the role of ghrelin as novel regulator of
hormone and prolactin release in the tilapia, Oreochromis mossambicus. gonadal function. Growth Horm. IGF Res. 15, 83–88.
Zool. Sci. 19, 797–800. Terova, G., Rimoldi, S., Bernardini, G., Gornati, R., Saroglia, M., 2008. Sea
Riley, L.G., Fox, B.K., Kaiya, H., Hirano, T., Grau, E.G., 2005. Long-term bass ghrelin: Molecular cloning and mRNA quantification during fasting
treatment of ghrelin stimulates feeding, fat deposition, and alters the GH/ and refeeding. Gen. Comp. Endocrinol. 155, 341–351.
IGF-I axis in the tilapia, Oreochromis mossambicus. Gen. Comp. Thompson, N.M., Gill, D.A., Davies, R., Loveridge, N., Houston, P.A.,
Endocrinol. 142, 234–240. Robinson, I.C., Wells, T., 2004. Ghrelin and des-octanoyl ghrelin promote
Sato, M., Nakahara, K., Goto, S., Kaiya, H., Miyazato, M., Date, Y., Nakazato, adipogenesis directly in vivo by a mechanism independent of the type 1a
M., Kangawa, K., Murakami, N., 2006. Effects of ghrelin and des-acyl growth hormone secretagogue receptor. Endocrinology 145, 234–242.
ghrelin on neurogenesis of the rat fet al spinal cord. Biochem. Biophys. Res. Toshinai, K., Yamaguchi, H., Sun, Y., Smith, R.G., Yamanaka, A., Sakurai, T.,
Commun. 350, 598–603. Date, Y., Mondal, M.S., Shimbara, T., Kawagoe, T., Murakami, N.,
Saito, E.S., Kaiya, H., Takagi, T., Yamasaki, I., Denbow, D.M., Kangawa, K., Miyazato, M., Kangawa, K., Nakazato, M., 2006. Des-acyl ghrelin induces
Furuse, M., 2002a. Chicken ghrelin and growth hormone-releasing peptide- food intake by a mechanism independent of the growth hormone
2 inhibit food intake of neonatal chicks. Eur. J. Pharmacol. 453, 75–79. secretagogue receptor. Endocrinology 147, 2306–2314.
Saito, E.S., Takagi, T., Nakanishi, T., Sashihara, K., Furuse, M., 2002b. Ghrelin Tschöp, M., Smiley, D.L., Heiman, M.L., 2000. Ghrelin induces adiposity in
activates behavior of neonatal chicks in a short period of post- rodents. Nature 407, 908–913.
intracerebroventricular injection. J. Appl. Anim. Res. 22, 33–41. Unniappan, S., Peter, R.E., 2004. In vitro and in vivo effects of ghrelin on
Saito, E.S., Kaiya, H., Tachibanat, T., Tomonaga, S., Denbow, D.M., Kangawa, luteinizing hormone and growth hormone release in goldfish. Am. J.
K., Furuse, M., 2005. Inhibitory effect of ghrelin on food intake is mediated Physiol. Regul. Integr. Comp. Physiol. 286, R1093–R1101.
by the corticotropin-releasing factor system in neonatal chicks. Regul. Pept. Unniappan, S., Peter, R.E., 2005. Structure, distribution and physiological
125, 201–208. functions of ghrelin in fish. Comp. Biochem. Physiol. A 140, 396–408.
Sakata, I., Mori, T., Kaiya, H., Yamazaki, M., Kangawa, K., Inoue, K., Sakai, T., Unniappan, S., Lin, X., Cervini, L., Rivier, J., Kaiya, H., Kangawa, K., Peter,
2004. Localization of ghrelin-producing cells in the stomach of the rainbow R.E., 2002. Goldfish ghrelin: molecular characterization of the comple-
trout (Oncorhynchus mykiss). Zool. Sci. 21, 757–762. mentary deoxyribonucleic acid, partial gene structure, and evidence for its
Seoane, L.M., Al-Massadi, O., Pazos, Y., Pagotto, U., Casanueva, F.F., 2006. stimulatory role in food intake. Endocrinology 143, 4143–4146.
Central obestatin administration does not modify either spontaneous or Unniappan, S., Canosa, L.F., Peter, R.E., 2004. Orexigenic actions of ghrelin in
ghrelin-induced food intake in rats. J. Endocrinol. Investig. 29, goldfish: feeding-induced changes in brain and gut mRNA expression and
RC13–RC15. serum levels, and responses to central and peripheral injections. Neuroen-
Shepherd, B.S., Eckert, S.M., Parhar, I.S., Vijayan, M.M., Wakabayashi, I., docrinology 79, 100–108.
Hirano, T., Grau, E.G., Chen, T.T., 2000. The hexapeptide KP-102 (D-ala-D- van der Lely, A.J., Tschöp, M., Heiman, M.L., Ghigo, E., 2004. Biological,
beta-Nal-ala-trp-D-phe-lys-NH(2)) stimulates growth hormone release in a physiological, pathophysiological and pharmacological aspects of ghrelin.
cichlid fish (Oreochromis mossambicus). J. Endocrinol. 167, R7–R10. Endocr. Rev. 25, 426–457.
Shepherd, B.S., Johnson, J.K., Silverstein, J.T., Parhar, I.S., Vijayan, M.M., Volkoff, H., Canosa, L.F., Unniappan, S., Cerda-Reverter, J.M., Bernier, N.J.,
McGuire, A., Weber, G.M., 2007. Endocrine and orexigenic actions of Kelly, S.P., Peter, R.E., 2005. Neuropeptides and the control of food intake
growth hormone secretagogues in rainbow trout (Oncorhynchus mykiss). in fish. Gen. Comp. Endocrinol. 142, 3–19.
Comp. Biochem. Physiol. A. 146, 390–399. Wada, R., Sakata, I., Kaiya, H., Nakamura, K., Hayashi, Y., Kangawa, K., Sakai,
Shousha, S., Nakahara, K., Kojima, M., Miyazato, M., Hosoda, H., Kangawa, T., 2003. Existence of ghrelin-immunopositive and -expressing cells in the
K., Murakami, N., 2005. Different effects of peripheral and central ghrelin proventriculus of the hatching and adult chicken. Regul. Pept. 111,
on regulation of food intake in the Japanese quail. Gen. Comp. Endocrinol. 123–128.
141, 178–183. Wajnrajch, M.P., Ten, I.S., Gertner, J.M., Leibel, R.L., 2000. Genomic
Sibilia, V., Bresciani, E., Lattuada, N., Rapetti, D., Locatelli, V., De Luca, V., organization of the human ghrelin gene. J. Endocrinol. Genet. 1, 231–233.
Dona, F., Netti, C., Torsello, A., Guidobono, F., 2006. Intracerebroven- Wei, W., Wang, G., Qi, X., Englander, E.W., Greeley Jr., G.H., 2005.
tricular acute and chronic administration of obestatin minimally affect food Characterization and regulation of the rat and human ghrelin promoters.
intake but not weight gain in the rat. J. Endocrinol. Investig. 29, Endocrinology 146, 1611–1625.
RC31–RC34. Wren, A.M., Small, C.J., Ward, H.L., Murphy, K.G., Dakin, C.L., Taheri, S.,
Sirotkin, A.V., Grossmann, R., 2007. The role of ghrelin and some intracellular Kennedy, A.R., Roberts, G.H., Morgan, D.G., Ghatei, M.A., Bloom, S.R.,
mechanisms in controlling the secretory activity of chicken ovarian cells. 2000. The novel hypothalamic peptide ghrelin stimulates food intake and
Comp. Biochem. Physiol. A 147, 239–246. growth hormone secretion. Endocrinology 141, 4325–4328.
Sirotkin, A.V., Grossmann, R., Maria-Peon, M.T., Roa, J., Tena-Sempere, M., Wren, A.M., Seal, L.J., Cohen, A.E., Brynes, A.E., Frost, G.S., Murphy, K.G.,
Klein, S., 2006. Novel expression and functional role of ghrelin in chicken Dhillo, W.S., Ghatei, M.A., Bloom, S.R., 2001. Ghrelin enhances appetite and
ovary. Mol. Cell. Endocrinol. 257–258, 15–25. increases food intake in humans. J. Clin. Endocrinol. Metab. 86, 5992–5995.
Author's personal copy

128 H. Kaiya et al. / Comparative Biochemistry and Physiology, Part A 149 (2008) 109–128

Xiao, D., Wong, A.O.L., Lin, H.-R., 2002. Lack of growth hormone releasing Yoshimoto, A., Mori, K., Sugawara, A., Mukoyama, M., Yahata, K., Suganami,
peptide-6 action on in vivo and in vitro growth hormone secretion in T., Takaya, K., Hosoda, H., Kojima, M., Kangawa, K., Nakao, K., 2002.
sexually immature and grass carp, Ctenopharyngodon idellus. Fish Physiol. Plasma ghrelin and desacyl ghrelin concentrations in renal failure. J. Am.
Biochem. 26, 315–327. Soc. Nephrol. 13, 2748–2752.
Yada, T., Kaiya, H., Mutoh, K., Azuma, T., Hyodo, S., Kangawa, K., 2006. Yoshimura, Y., Nagano, K., Subedi, K., Kaiya, H., 2005. Identification of
Ghrelin stimulates phagocytosis and superoxide production in fish immunoreactive ghrelin and its mRNA in the oviduct of laying Japanese
leucocytes. J. Endocrinol. 189, 57–65. quail, Coturnix japonica. J. Poult. Sci. 42, 291–300.
Yamato, M., Sakata, I., Wada, R., Kaiya, H., Sakai, T., 2005. Exogenous Yuan, J., Zhou, J., Hu, X., Li, N., 2007. Molecular cloning and comparison of
administration of octanoic acid accelerates octanoylated ghrelin production avian preproghrelin genes. Biochem. Genet. 45, 185–194.
in the proventriculus of neonatal chicks. Biochem. Biophys. Res. Commun. Zhang, J.V., Ren, P.G., Avsian-Kretchmer, O., Luo, C.W., Rauch, R., Klein, C.,
333, 583–589. Hsueh, A.J., 2005. Obestatin, a peptide encoded by the ghrelin gene,
Yeung, C.M., Chan, C.B., Cheng, C.H., 2004. Isolation and characterization of opposes ghrelin's effects on food intake. Science 310, 996–999.
the 5'-flanking region of the growth hormone secretagogue receptor gene Zhang, J.V., Klein, C., Ren, P.G., Kass, S., Donck, L.V., Moechars, D., Hsueh, A.J.,
from black seabream Acanthopagrus schlegeli. Mol. Cell. Endocrinol. 223, 2007. Response to comment on “Obestatin, a peptide encoded by the ghrelin
5–15. gene, opposes ghrelin's effects on food intake”. Science 315, 766 author reply.
Yeung, C.M., Chan, C.B., Woo, N.Y., Cheng, C.H., 2006. Seabream ghrelin: Zizzari, P., Longchamps, R., Epelbaum, J., Bluet-Pajot, M.T., 2007. Obestatin
cDNA cloning, genomic organization and promoter studies. J. Endocrinol. partially affects ghrelin stimulation of food intake and growth hormone
189, 365–379. secretion in rodents. Endocrinology 148, 1648–1653.

Você também pode gostar